1
|
Jomova K, Alomar SY, Nepovimova E, Kuca K, Valko M. Heavy metals: toxicity and human health effects. Arch Toxicol 2025; 99:153-209. [PMID: 39567405 PMCID: PMC11742009 DOI: 10.1007/s00204-024-03903-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024]
Abstract
Heavy metals are naturally occurring components of the Earth's crust and persistent environmental pollutants. Human exposure to heavy metals occurs via various pathways, including inhalation of air/dust particles, ingesting contaminated water or soil, or through the food chain. Their bioaccumulation may lead to diverse toxic effects affecting different body tissues and organ systems. The toxicity of heavy metals depends on the properties of the given metal, dose, route, duration of exposure (acute or chronic), and extent of bioaccumulation. The detrimental impacts of heavy metals on human health are largely linked to their capacity to interfere with antioxidant defense mechanisms, primarily through their interaction with intracellular glutathione (GSH) or sulfhydryl groups (R-SH) of antioxidant enzymes such as superoxide dismutase (SOD), catalase, glutathione peroxidase (GPx), glutathione reductase (GR), and other enzyme systems. Although arsenic (As) is believed to bind directly to critical thiols, alternative hydrogen peroxide production processes have also been postulated. Heavy metals are known to interfere with signaling pathways and affect a variety of cellular processes, including cell growth, proliferation, survival, metabolism, and apoptosis. For example, cadmium can affect the BLC-2 family of proteins involved in mitochondrial death via the overexpression of antiapoptotic Bcl-2 and the suppression of proapoptotic (BAX, BAK) mechanisms, thus increasing the resistance of various cells to undergo malignant transformation. Nuclear factor erythroid 2-related factor 2 (Nrf2) is an important regulator of antioxidant enzymes, the level of oxidative stress, and cellular resistance to oxidants and has been shown to act as a double-edged sword in response to arsenic-induced oxidative stress. Another mechanism of significant health threats and heavy metal (e.g., Pb) toxicity involves the substitution of essential metals (e.g., calcium (Ca), copper (Cu), and iron (Fe)) with structurally similar heavy metals (e.g., cadmium (Cd) and lead (Pb)) in the metal-binding sites of proteins. Displaced essential redox metals (copper, iron, manganese) from their natural metal-binding sites can catalyze the decomposition of hydrogen peroxide via the Fenton reaction and generate damaging ROS such as hydroxyl radicals, causing damage to lipids, proteins, and DNA. Conversely, some heavy metals, such as cadmium, can suppress the synthesis of nitric oxide radical (NO·), manifested by altered vasorelaxation and, consequently, blood pressure regulation. Pb-induced oxidative stress has been shown to be indirectly responsible for the depletion of nitric oxide due to its interaction with superoxide radical (O2·-), resulting in the formation of a potent biological oxidant, peroxynitrite (ONOO-). This review comprehensively discusses the mechanisms of heavy metal toxicity and their health effects. Aluminum (Al), cadmium (Cd), arsenic (As), mercury (Hg), lead (Pb), and chromium (Cr) and their roles in the development of gastrointestinal, pulmonary, kidney, reproductive, neurodegenerative (Alzheimer's and Parkinson's diseases), cardiovascular, and cancer (e.g. renal, lung, skin, stomach) diseases are discussed. A short account is devoted to the detoxification of heavy metals by chelation via the use of ethylenediaminetetraacetic acid (EDTA), dimercaprol (BAL), 2,3-dimercaptosuccinic acid (DMSA), 2,3-dimercapto-1-propane sulfonic acid (DMPS), and penicillamine chelators.
Collapse
Affiliation(s)
- Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences, Constantine The Philosopher University in Nitra, 949 74, Nitra, Slovakia
| | - Suliman Y Alomar
- Doping Research Chair, Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50005, Hradec Kralove, Czech Republic
- Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, 708 00, Ostrava-Poruba, Czech Republic
| | - Kamil Kuca
- Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, 708 00, Ostrava-Poruba, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37, Bratislava, Slovakia.
| |
Collapse
|
2
|
Zhang Y, Zhou M, Wang D, Liang R, Liu W, Wang B, Chen W. Arsenic exposure and oxidative damage to lipid, DNA, and protein among general Chinese adults: A repeated-measures cross-sectional and longitudinal study. J Environ Sci (China) 2025; 147:382-391. [PMID: 39003056 DOI: 10.1016/j.jes.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 07/15/2024]
Abstract
Arsenic-related oxidative stress and resultant diseases have attracted global concern, while longitudinal studies are scarce. To assess the relationship between arsenic exposure and systemic oxidative damage, we performed two repeated measures among 5236 observations (4067 participants) in the Wuhan-Zhuhai cohort at the baseline and follow-up after 3 years. Urinary total arsenic, biomarkers of DNA oxidative damage (8-hydroxy-2'-deoxyguanosine (8-OHdG)), lipid peroxidation (8-isoprostaglandin F2alpha (8-isoPGF2α)), and protein oxidative damage (protein carbonyls (PCO)) were detected for all observations. Here we used linear mixed models to estimate the cross-sectional and longitudinal associations between arsenic exposure and oxidative damage. Exposure-response curves were constructed by utilizing the generalized additive mixed models with thin plate regressions. After adjusting for potential confounders, arsenic level was significantly and positively related to the levels of global oxidative damage and their annual increased rates in dose-response manners. In cross-sectional analyses, each 1% increase in arsenic level was associated with a 0.406% (95% confidence interval (CI): 0.379% to 0.433%), 0.360% (0.301% to 0.420%), and 0.079% (0.055% to 0.103%) increase in 8-isoPGF2α, 8-OHdG, and PCO, respectively. More importantly, arsenic was further found to be associated with increased annual change rates of 8-isoPGF2α (β: 0.147; 95% CI: 0.130 to 0.164), 8-OHdG (0.155; 0.118 to 0.192), and PCO (0.050; 0.035 to 0.064) in the longitudinal analyses. Our study suggested that arsenic exposure was not only positively related with global oxidative damage to lipid, DNA, and protein in cross-sectional analyses, but also associated with annual increased rates of these biomarkers in dose-dependent manners.
Collapse
Affiliation(s)
- Yongfang Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Min Zhou
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dongming Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ruyi Liang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wei Liu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bin Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Weihong Chen
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
3
|
Ilic-Tomic T, Kramar A, Kostic M, Vojnovic S, Milovanovic J, Petkovic M, D’Agostino PM, Gulder TAM, Nikodinovic-Runic J. Functionalization of silk with actinomycins from Streptomyces anulatus BV365 for biomedical applications. Front Bioeng Biotechnol 2024; 12:1466757. [PMID: 39364265 PMCID: PMC11447452 DOI: 10.3389/fbioe.2024.1466757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/05/2024] [Indexed: 10/05/2024] Open
Abstract
Silk, traditionally acclaimed as the "queen of fiber," has been widely used thanks to its brilliant performance such as gentleness, smoothness and comfortableness. Owing to its mechanical characteristics and biocompatibility silk has a definitive role in biomedical applications, both as fibroin and fabric. In this work, the simultaneous dyeing and functionalization of silk fabric with pigments from Streptomyces anulatus BV365 were investigated. This strain produced high amounts of orange extracellular pigments on mannitol-soy flour agar, identified as actinomycin D, C2 and C3. The application of purified actinomycins in the dyeing of multifiber fabric was assessed. Actinomycins exhibited a high affinity towards protein fibers (silk and wool), but washing durability was maintained only with silk. Acidic condition (pH5) and high temperature (65°C) facilitated the silk dyeing. The morphologies and chemical components of the treated silk fabrics were analyzed using scanning electron microscopy and Fourier transform infrared spectroscopy. The results showed the pigments bind to the silk through interaction with the carbonyl group in silk fibroin rendering the functionalized, yet surface that does not cause skin irritation. The treated silk exhibited a remarkable antibacterial effect, while the biocompatibility test performed with 3D-reconstructed human epidermis model indicated safe biological properties, paving the way for future application of this material in medicine.
Collapse
Affiliation(s)
- Tatjana Ilic-Tomic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Ana Kramar
- Department of Textile Engineering, Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
- Novel Materials and Nanotechnology Group, Institute of Agrochemistry and Food Technology (IATA), Spanish Council for Scientific Research (CSIC), Paterna, Spain
| | - Mirjana Kostic
- Department of Textile Engineering, Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | - Sandra Vojnovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Jelena Milovanovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Milos Petkovic
- Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Paul M. D’Agostino
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Saarland, Germany
- Technical University of Dresden, Dresden, Saxony, Germany
| | - Tobias A. M. Gulder
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Saarland, Germany
- Technical University of Dresden, Dresden, Saxony, Germany
| | | |
Collapse
|
4
|
Liu J, Hermon T, Gao X, Dixon D, Xiao H. Arsenic and Diabetes Mellitus: A Putative Role for the Immune System. ALL LIFE 2023; 16:2167869. [PMID: 37152101 PMCID: PMC10162781 DOI: 10.1080/26895293.2023.2167869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 01/02/2023] [Indexed: 02/04/2023] Open
Abstract
Diabetes mellitus (DM) is an enormous public health issue worldwide. Recent data suggest that chronic arsenic exposure is linked to the risk of developing type 1 and type 2 DM, albeit the underlying mechanisms are unclear. This review discusses the role of the immune system as a link to possibly explain some of the mechanisms of developing T1DM or T2DM associated with arsenic exposure in humans, animal models, and in vitro studies. The rationale for the hypothesis includes: (1) Arsenic is a well-recognized modulator of the immune system; (2) arsenic exposures are associated with increased risk of DM; and (3) dysregulation of the immune system is one of the hallmarks in the pathogenesis of both T1DM and T2DM. A better understanding of DM in association with immune dysregulation and arsenic exposures may help to understand how environmental exposures modulate the immune system and how these effects may impact the manifestation of disease.
Collapse
Affiliation(s)
- Jingli Liu
- Molecular Pathogenesis Group, Mechanistic Toxicology Branch, Division of the National Toxicology Program (DNTP), National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, 111 TW Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Tonia Hermon
- Molecular Pathogenesis Group, Mechanistic Toxicology Branch, Division of the National Toxicology Program (DNTP), National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, 111 TW Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Xiaohua Gao
- Molecular Pathogenesis Group, Mechanistic Toxicology Branch, Division of the National Toxicology Program (DNTP), National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, 111 TW Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Darlene Dixon
- Molecular Pathogenesis Group, Mechanistic Toxicology Branch, Division of the National Toxicology Program (DNTP), National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, 111 TW Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Hang Xiao
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, Jiangsu, China
| |
Collapse
|
5
|
Rachamalla M, Chinthada J, Kushwaha S, Putnala SK, Sahu C, Jena G, Niyogi S. Contemporary Comprehensive Review on Arsenic-Induced Male Reproductive Toxicity and Mechanisms of Phytonutrient Intervention. TOXICS 2022; 10:toxics10120744. [PMID: 36548577 PMCID: PMC9784647 DOI: 10.3390/toxics10120744] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 05/26/2023]
Abstract
Arsenic (As) is a poisonous metalloid that is toxic to both humans and animals. Drinking water contamination has been linked to the development of cancer (skin, lung, urinary bladder, and liver), as well as other disorders such as diabetes and cardiovascular, gastrointestinal, neurological, and developmental damage. According to epidemiological studies, As contributes to male infertility, sexual dysfunction, poor sperm quality, and developmental consequences such as low birth weight, spontaneous abortion, and small for gestational age (SGA). Arsenic exposure negatively affected male reproductive systems by lowering testicular and accessory organ weights, and sperm counts, increasing sperm abnormalities and causing apoptotic cell death in Leydig and Sertoli cells, which resulted in decreased testosterone synthesis. Furthermore, during male reproductive toxicity, several molecular signalling pathways, such as apoptosis, inflammation, and autophagy are involved. Phytonutrient intervention in arsenic-induced male reproductive toxicity in various species has received a lot of attention over the years. The current review provides an in-depth summary of the available literature on arsenic-induced male toxicity, as well as therapeutic approaches and future directions.
Collapse
Affiliation(s)
- Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | - Joshi Chinthada
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S Nagar 160062, India
| | - Sapana Kushwaha
- Department of Pharmacology and Toxicology, Transit Campus, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Sravan Kumar Putnala
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | - Chittaranjan Sahu
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S Nagar 160062, India
| | - Gopabandhu Jena
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S Nagar 160062, India
| | - Som Niyogi
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| |
Collapse
|
6
|
Lou Q, Zhang M, Zhang K, Liu X, Zhang Z, Zhang X, Yang Y, Gao Y. Arsenic exposure elevated ROS promotes energy metabolic reprogramming with enhanced AKT-dependent HK2 expression. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 836:155691. [PMID: 35525345 DOI: 10.1016/j.scitotenv.2022.155691] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/22/2022] [Accepted: 04/30/2022] [Indexed: 06/14/2023]
Abstract
Exposure to inorganic or organic arsenic compounds continues to pose substantial public health concerns for hundreds of millions of people around the globe. Highly exposed individuals are susceptible to various illnesses, including impairments and cancers of the lung, liver, skin and bladder. Long-term exposure to low-dose arsenic has been identified to induce aerobic glycolysis, which contributes to cells aberrant proliferation. However, the mechanism underlying arsenic-induced aerobic glycolysis is still unclear. Here, mtDNA copy number is enhanced in arsenic-exposed populations and a positive correlation between serum HK2 and urinary total arsenic was observed in the individuals with high urine arsenic (≥ 0.032 mg/L). In a rat model of trivalent arsenic (iAs3+) exposure, the levels of HK2, NDUFA9 and NDUFB8 were increased in the rats treated with iAs3+ daily by gavage for 12 weeks than those in the control rats. Subsequently, in a low-dose arsenic exposure cell model we found that 0.2 μmol/L iAs3+ induced aerobic glycolysis to promote L-02 cells proliferation and inhibit apoptosis, in which HK2 played an important role. Further studies showed accumulated ROS determined the metabolic reprogramming via activating AKT and then increasing HK2 expression. On the one hand, activated AKT induced aerobic glycolysis by increasing HK2 to promote L-02 cells viability and DNA synthesis; on the other hand, phosphorylated AKT induced HK2 mitochondrial outer-membrane location with VDAC1 to inhibit apoptosis. Taken together, our results indicated that ROS induced by low-dose arsenic exposure determined energy metabolic reprogramming and acted a critical regulator for AKT-dependent HK2 expression and aerobic glycolysis.
Collapse
Affiliation(s)
- Qun Lou
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Meichen Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Kunyu Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Xiaona Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Zaihong Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Xin Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Yanmei Yang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China.
| | - Yanhui Gao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China.
| |
Collapse
|
7
|
Zhou X, Speer RM, Volk L, Hudson LG, Liu KJ. Arsenic co-carcinogenesis: Inhibition of DNA repair and interaction with zinc finger proteins. Semin Cancer Biol 2021; 76:86-98. [PMID: 33984503 PMCID: PMC8578584 DOI: 10.1016/j.semcancer.2021.05.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/17/2022]
Abstract
Arsenic is widely present in the environment and is associated with various population health risks including cancers. Arsenic exposure at environmentally relevant levels enhances the mutagenic effect of other carcinogens such as ultraviolet radiation. Investigation on the molecular mechanisms could inform the prevention and intervention strategies of arsenic carcinogenesis and co-carcinogenesis. Arsenic inhibition of DNA repair has been demonstrated to be an important mechanism, and certain DNA repair proteins have been identified to be extremely sensitive to arsenic exposure. This review will summarize the recent advances in understanding the mechanisms of arsenic carcinogenesis and co-carcinogenesis, including DNA damage induction and ROS generation, particularly how arsenic inhibits DNA repair through an integrated molecular mechanism which includes its interactions with sensitive zinc finger DNA repair proteins.
Collapse
Affiliation(s)
- Xixi Zhou
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Rachel M Speer
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Lindsay Volk
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Laurie G Hudson
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA.
| | - Ke Jian Liu
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA.
| |
Collapse
|
8
|
Zhang Z, Costa M. p62 functions as a signal hub in metal carcinogenesis. Semin Cancer Biol 2021; 76:267-278. [PMID: 33894381 PMCID: PMC9161642 DOI: 10.1016/j.semcancer.2021.04.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 04/06/2021] [Accepted: 04/15/2021] [Indexed: 12/13/2022]
Abstract
A number of metals are toxic and carcinogenic to humans. Reactive oxygen species (ROS) play an important role in metal carcinogenesis. Oxidative stress acts as the converging point among various stressors with ROS being the main intracellular signal transducer. In metal-transformed cells, persistent expression of p62 and erythroid 2-related factor 2 (Nrf2) result in apoptosis resistance, angiogenesis, inflammatory microenvironment, and metabolic reprogramming, contributing to overall mechanism of metal carcinogenesis. Autophagy, a conserved intracellular process, maintains cellular homeostasis by facilitating the turnover of protein aggregates, cellular debris, and damaged organelles. In addition to being a substrate of autophagy, p62 is also a crucial molecule in a myriad of cellular functions and in molecular events, which include oxidative stress, inflammation, apoptosis, cell proliferation, metabolic reprogramming, that modulate cell survival and tumor growth. The multiple functions of p62 are appreciated by its ability to interact with several key components involved in various oncogenic pathways. This review summarizes the current knowledge and progress in studies of p62 and metal carcinogenesis with emphasis on oncogenic pathways related to oxidative stress, inflammation, apoptosis, and metabolic reprogramming.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Environmental Medicine, NYU School of Medicine, 341 East 25th Street, New York, NY 10010, USA
| | - Max Costa
- Department of Environmental Medicine, NYU School of Medicine, 341 East 25th Street, New York, NY 10010, USA.
| |
Collapse
|
9
|
Wu Z, Sainz AG, Shadel GS. Mitochondrial DNA: cellular genotoxic stress sentinel. Trends Biochem Sci 2021; 46:812-821. [PMID: 34088564 PMCID: PMC9809014 DOI: 10.1016/j.tibs.2021.05.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/24/2021] [Accepted: 05/08/2021] [Indexed: 02/08/2023]
Abstract
High copy number, damage prone, and lean on repair mechanisms are unique features of mitochondrial DNA (mtDNA) that are hard to reconcile with its essentiality for oxidative phosphorylation, the primary function ascribed to this maternally inherited component of our genome. We propose that mtDNA is also a genotoxic stress sentinel, as well as a direct second messenger of this type of cellular stress. Here, we discuss existing evidence for this sentinel/effector role through the ability of mtDNA to escape the confines of the mitochondrial matrix and activate nuclear DNA damage/repair responses via interferon-stimulated gene products and other downstream effectors. However, this arrangement may come at a cost, leading to cancer chemoresistance and contributing to inflammation, disease pathology, and aging.
Collapse
Affiliation(s)
- Zheng Wu
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA,Graduate Program in Genetics, Yale School of Medicine, New Haven, CT 06437, USA,These authors contributed equally to this work
| | - Alva G. Sainz
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA,Graduate Program in Experimental Pathology, Yale School of Medicine, New Haven, CT 06437, USA,These authors contributed equally to this work
| | - Gerald S. Shadel
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA,Correspondence: (G.S. Shadel)
| |
Collapse
|
10
|
Thakur M, Rachamalla M, Niyogi S, Datusalia AK, Flora SJS. Molecular Mechanism of Arsenic-Induced Neurotoxicity including Neuronal Dysfunctions. Int J Mol Sci 2021; 22:10077. [PMID: 34576240 PMCID: PMC8471829 DOI: 10.3390/ijms221810077] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 12/15/2022] Open
Abstract
Arsenic is a key environmental toxicant having significant impacts on human health. Millions of people in developing countries such as Bangladesh, Mexico, Taiwan, and India are affected by arsenic contamination through groundwater. Environmental contamination of arsenic leads to leads to various types of cancers, coronary and neurological ailments in human. There are several sources of arsenic exposure such as drinking water, diet, wood preservatives, smoking, air and cosmetics, while, drinking water is the most explored route. Inorganic arsenic exhibits higher levels of toxicity compared its organic forms. Exposure to inorganic arsenic is known to cause major neurological effects such as cytotoxicity, chromosomal aberration, damage to cellular DNA and genotoxicity. On the other hand, long-term exposure to arsenic may cause neurobehavioral effects in the juvenile stage, which may have detrimental effects in the later stages of life. Thus, it is important to understand the toxicology and underlying molecular mechanism of arsenic which will help to mitigate its detrimental effects. The present review focuses on the epidemiology, and the toxic mechanisms responsible for arsenic induced neurobehavioral diseases, including strategies for its management from water, community and household premises. The review also provides a critical analysis of epigenetic and transgenerational modifications, mitochondrial oxidative stress, molecular mechanisms of arsenic-induced oxidative stress, and neuronal dysfunction.
Collapse
Affiliation(s)
- Manisha Thakur
- Department of Pharmacology and Toxicology, Transit Campus, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India; (M.T.); (A.K.D.)
| | - Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada; (M.R.); (S.N.)
| | - Som Niyogi
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada; (M.R.); (S.N.)
- Toxicology Centre, Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | - Ashok Kumar Datusalia
- Department of Pharmacology and Toxicology, Transit Campus, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India; (M.T.); (A.K.D.)
| | - Swaran Jeet Singh Flora
- Department of Pharmacology and Toxicology, Transit Campus, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India; (M.T.); (A.K.D.)
| |
Collapse
|
11
|
Naqvi S, Kumar P, Flora SJS. Comparative efficacy of Nano and Bulk Monoisoamyl DMSA against arsenic-induced neurotoxicity in rats. Biomed Pharmacother 2020; 132:110871. [PMID: 33069968 DOI: 10.1016/j.biopha.2020.110871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 10/23/2022] Open
Abstract
Chelation therapy is considered as a safe and effective strategy to combat metal poisoning. Arsenic is known to cause neurological dysfunctions such as impaired memory, encephalopathy, and peripheral neuropathy as it easily crosses the blood-brain barrier. Oxidative stress is one of the mechanisms suggested for arsenic-induced neurotoxicity. We prepared Solid Lipid nanoparticles loaded with Monoisoamyl 2, 3-dimercaptosuccinic acid (Nano-MiADMSA), and compared their efficacy with bulk MiADMSA for treating arsenic-induced neurological and other biochemical effects. Solid lipid nanoparticles entrapping MiADMSA were synthesized and particle characterization was carried out by transmission electron microscopy (TEM) and dynamic light scattering (DLS). An in vivo study was planned to investigate the therapeutic efficacy of MiADMSA-encapsulated solid lipid nanoparticles (Nano-MiADMSA; 50 mg/kg orally for 5 days) and compared it with bulk MiADMSA against sodium meta-arsenite exposed rats (25 ppm in drinking water, for 12 weeks) in male rats. The results suggested the size of Nano-MiADMSA was between 100-120 nm ranges. We noted enhanced chelating properties of Nano-MiADMSA compared with bulk MiADMSA as evident by the reversal of oxidative stress variables like blood δ-aminolevulinic acid dehydratase (δ-ALAD), Reactive Oxygen Species (ROS), Catalase activity, Superoxide Dismutase (SOD), Thiobarbituric Acid Reactive Substances (TBARS), Reduced Glutathione (GSH) and Oxidized Glutathione (GSSG), Glutathione Peroxidase (GPx), Glutathione-S-transferase (GST) and efficient removal of arsenic from the blood and tissues. Recoveries in neurobehavioral parameters further confirmed nano-MiADMSA to be more effective than bulk MiADMSA. We conclude that treatment with Nano-MiADMSA is a better therapeutic strategy than bulk MiADMSA in reducing the effects of arsenic-induced oxidative stress and associated neurobehavioral changes.
Collapse
Affiliation(s)
- Saba Naqvi
- National Institute of Pharmaceutical Education and Research (NIPER-Raebareli), Bijnor-Sisendi Road, CRPF Base Camp, P.O. Mati, Sarojini Nagar, Lucknow, UP, 226002, India
| | - Prince Kumar
- National Institute of Pharmaceutical Education and Research (NIPER-Raebareli), Bijnor-Sisendi Road, CRPF Base Camp, P.O. Mati, Sarojini Nagar, Lucknow, UP, 226002, India
| | - S J S Flora
- National Institute of Pharmaceutical Education and Research (NIPER-Raebareli), Bijnor-Sisendi Road, CRPF Base Camp, P.O. Mati, Sarojini Nagar, Lucknow, UP, 226002, India.
| |
Collapse
|
12
|
The Role of Reactive Oxygen Species in Arsenic Toxicity. Biomolecules 2020; 10:biom10020240. [PMID: 32033297 PMCID: PMC7072296 DOI: 10.3390/biom10020240] [Citation(s) in RCA: 211] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/13/2022] Open
Abstract
Arsenic poisoning is a global health problem. Chronic exposure to arsenic has been associated with the development of a wide range of diseases and health problems in humans. Arsenic exposure induces the generation of intracellular reactive oxygen species (ROS), which mediate multiple changes to cell behavior by altering signaling pathways and epigenetic modifications, or cause direct oxidative damage to molecules. Antioxidants with the potential to reduce ROS levels have been shown to ameliorate arsenic-induced lesions. However, emerging evidence suggests that constructive activation of antioxidative pathways and decreased ROS levels contribute to chronic arsenic toxicity in some cases. This review details the pathways involved in arsenic-induced redox imbalance, as well as current studies on prophylaxis and treatment strategies using antioxidants.
Collapse
|
13
|
Arsenic trioxide: insights into its evolution to an anticancer agent. J Biol Inorg Chem 2018; 23:313-329. [DOI: 10.1007/s00775-018-1537-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/22/2018] [Indexed: 01/01/2023]
|
14
|
Ahangarpour A, Zeidooni L, Samimi A, Alboghobeish S, Khorsandi LS, Moradi M. Chronic exposure to arsenic and high fat diet additively induced cardiotoxicity in male mice. Res Pharm Sci 2018; 13:47-56. [PMID: 29387111 PMCID: PMC5772081 DOI: 10.4103/1735-5362.220967] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Diet is one of the important risk factors that could potentially affect arsenic-induced cardiotoxicity. The present study was undertaken to investigate the effect of high fat diet on arsenic-induced cardiotoxicity in mice. Mice were divided into six different groups (n = 12), two control groups received either low fat diet (LFD) or high fat diet (HFD) along with deionized drinking water and four test groups given LFD + 25 ppm arsenic, LFD + 50 ppm arsenic, HFD + 25 ppm arsenic, and HFD + 50 ppm arsenic in drinking water for 5 months. The body weight, heart weight to body weight ratio, cardiac biochemical markers, lipid profile, and histological examination of heart were evaluated. The results demonstrated that arsenic exposure led to a significant decrease in heart glutathione level, catalase enzyme activity, and a significant increase in reactive oxygen species (ROS), malondialdehyde levels, and biochemical enzymes. The administration of HFD resulted in above-mentioned changes as well as an alteration in lipid profile; however, arsenic exposure alone or along with HFD caused a reduction in lipid profile factors, except HDL level. Our results revealed that HFD increased arsenic-induced heart injury in the mice. This effect may be because of reduction in antioxidant activities and/or increase in oxidative stress and ROS in mice heart tissues. These findings could be important for clinical intervention to protect against or prevent arsenic-induced cardiotoxicity in humans.
Collapse
Affiliation(s)
- Akram Ahangarpour
- Health Research Institute, Diabetes Research Center, Department of Physiology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, I.R. Iran
| | - Leila Zeidooni
- Department of Toxicology and Student Research Committee, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, I.R. Iran
| | - Azin Samimi
- Department of Toxicology and Student Research Committee, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, I.R. Iran
| | - Soheila Alboghobeish
- Department of Pharmacology and Student Research Committee, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, I.R. Iran
| | - Laya Sadat Khorsandi
- Cell and Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, I.R. Iran
| | - Mitra Moradi
- Health Research Institute, Diabetes Research Center, Department of Physiology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, I.R. Iran
| |
Collapse
|
15
|
Eckstein M, Eleazer R, Rea M, Fondufe-Mittendorf Y. Epigenomic reprogramming in inorganic arsenic-mediated gene expression patterns during carcinogenesis. REVIEWS ON ENVIRONMENTAL HEALTH 2017; 32:93-103. [PMID: 27701139 DOI: 10.1515/reveh-2016-0025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 08/08/2016] [Indexed: 05/22/2023]
Abstract
Arsenic is a ubiquitous metalloid that is not mutagenic but is carcinogenic. The mechanism(s) by which arsenic causes cancer remain unknown. To date, several mechanisms have been proposed, including the arsenic-induced generation of reactive oxygen species (ROS). However, it is also becoming evident that inorganic arsenic (iAs) may exert its carcinogenic effects by changing the epigenome, and thereby modifying chromatin structure and dynamics. These epigenetic changes alter the accessibility of gene regulatory factors to DNA, resulting in specific changes in gene expression both at the levels of transcription initiation and gene splicing. In this review, we discuss recent literature reports describing epigenetic changes induced by iAs exposure and the possible epigenetic mechanisms underlying these changes.
Collapse
|
16
|
Keshtzar E, Khodayar MJ, Javadipour M, Ghaffari MA, Bolduc DL, Rezaei M. Ellagic acid protects against arsenic toxicity in isolated rat mitochondria possibly through the maintaining of complex II. Hum Exp Toxicol 2016; 35:1060-72. [DOI: 10.1177/0960327115618247] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chronic arsenic exposure has been linked to many health problems including diabetes and cancer. In the present study, we assessed the protective effect of ellagic acid (EA) against toxicity induced by arsenic in isolated rat liver mitochondria. Reactive oxygen species (ROS) and mitochondrial membrane potential decline were assayed using dichlorofluorescein diacetate and rhodamine 123, respectively, and dehydrogenase activity obtained by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide conversion assay. Arsenic increased ROS levels and mitochondrial dysfunction, which led to a reduction in mitochondrial total dehydrogenase activity. Mitochondria pretreated with EA exposed to arsenic at various concentrations led to a reversal of ROS production and mitochondrial damage. Our results showed that mitochondria were significantly affected when exposed to arsenic, which resulted in excessive ROS production and mitochondrial membrane disruption. Pretreatment with EA, reduced ROS amounts, mitochondrial damage, and restored total dehydrogenase activity specifically associated with mitochondrial complex II. EA protective characteristics may be accomplished particularly throughout the mitochondrial maintenance either directly by its antioxidant property or indirectly through its maintaining of complex II. These findings also suggest a potential role for EA in treating or preventing mitochondria associated disorders.
Collapse
Affiliation(s)
- E Keshtzar
- Diabetes Research Center, Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - MJ Khodayar
- Department of Pharmacology and Toxicology, School of Pharmacy, Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - M Javadipour
- Diabetes Research Center, Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - MA Ghaffari
- Cellular and Molecular Research Center, Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - DL Bolduc
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - M Rezaei
- Department of Pharmacology and Toxicology, School of Pharmacy, Jundishapur University of Medical Sciences, Ahvaz, Iran
- Cellular and Molecular Research Center, Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
17
|
Choudhury S, Gupta P, Ghosh S, Mukherjee S, Chakraborty P, Chatterji U, Chattopadhyay S. Arsenic-induced dose-dependent modulation of the NF-κB/IL-6 axis in thymocytes triggers differential immune responses. Toxicology 2016; 357-358:85-96. [PMID: 27289040 DOI: 10.1016/j.tox.2016.06.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/01/2016] [Accepted: 06/08/2016] [Indexed: 12/22/2022]
Abstract
Arsenic contamination of drinking water is a matter of global concern. Arsenic intake impairs immune responses and leads to a variety of pathological conditions including cancer. In order to understand the intricate tuning of immune responses elicited by chronic exposure to arsenic, a mouse model was established by subjecting mice to different environmentally relevant concentrations of arsenic in drinking water for 30days. Detailed study of the thymus, a primary immune organ, revealed arsenic-mediated tissue damage in both histological specimens and scanning electron micrographs. Analysis of molecular markers of apoptosis by Western blot revealed a dose-dependent activation of the apoptotic cascade. Enzymatic assays supported oxidative stress as an instigator of cell death. Interestingly, assessment of inflammatory responses revealed disparity in the NF-κB/IL-6/STAT3 axis, where it was found that in animals consuming higher amounts of arsenic NF-κB activation did not lead to the classical IL-6 upregulation response. This deviation from the canonical pathway was accompanied with a significant rise in numbers of CD4+ CD25+ FoxP3 expressing cells in the thymus. The cytokine profile of the animals exposed to higher doses of arsenic also indicated an immune-suppressed milieu, thus validating that arsenic shapes the immune environment in context to its dose of exposure and that at higher doses it leads to immune-suppression. Our study establishes a novel role of arsenic in regulating immune homeostasis in context to its dose, where, at higher doses, arsenic related upregulation of NF-κB cascade takes on an alternative role that is correlated with increased immune-suppression.
Collapse
Affiliation(s)
- Sreetama Choudhury
- Department of Physiology, UCSTA, University of Calcutta, 92, A.P.C. Road, Kolkata 700009, India
| | - Payal Gupta
- Department of Physiology, UCSTA, University of Calcutta, 92, A.P.C. Road, Kolkata 700009, India
| | - Sayan Ghosh
- Department of Physiology, UCSTA, University of Calcutta, 92, A.P.C. Road, Kolkata 700009, India
| | - Sudeshna Mukherjee
- Department of Physiology, UCSTA, University of Calcutta, 92, A.P.C. Road, Kolkata 700009, India
| | - Priyanka Chakraborty
- Department of Physiology, UCSTA, University of Calcutta, 92, A.P.C. Road, Kolkata 700009, India
| | - Urmi Chatterji
- Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India
| | - Sreya Chattopadhyay
- Department of Physiology, UCSTA, University of Calcutta, 92, A.P.C. Road, Kolkata 700009, India.
| |
Collapse
|
18
|
Ronchetti SA, Bianchi MS, Duvilanski BH, Cabilla JP. In Vivo and In Vitro Arsenic Exposition Induces Oxidative Stress in Anterior Pituitary Gland. Int J Toxicol 2016; 35:463-75. [PMID: 27151894 DOI: 10.1177/1091581816645797] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Inorganic arsenic (iAs) is at the top of toxic metalloids. Inorganic arsenic-contaminated water consumption is one of the greatest environmental health threats worldwide. Human iAs exposure has been associated with cancers of several organs, neurological disorders, and reproductive problems. Nevertheless, there are no reports describing how iAs affects the anterior pituitary gland. The aim of this study was to investigate the mechanisms involved in iAs-mediated anterior pituitary toxicity both in vivo and in vitro. We showed that iAs administration (from 5 to 100 ppm) to male rats through drinking water increased messenger RNA expression of several oxidative stress-responsive genes in the anterior pituitary gland. Serum prolactin levels diminished, whereas luteinizing hormone (LH) levels were only affected at the higher dose tested. In anterior pituitary cells in culture, 25 µmol/L iAs significantly decreased prolactin release in a time-dependent fashion, whereas LH levels remained unaltered. Cell viability was significantly reduced mainly by apoptosis evidenced by morphological and phosphatidylserine externalization studies. This process is characterized by early depolarization of mitochondrial membrane potential and increased levels of reactive oxygen species. Expression of some key oxidative stress-responsive genes, such as heme oxygenase-1 and metallothionein-1, was also stimulated by iAs exposure. The antioxidant N-acetyl cysteine prevented iAs-induced effects on the expression of oxidative stress markers, prolactin release, and apoptosis. In summary, the present work demonstrates for the first time that iAs reduces prolactin release both in vivo and in vitro and induces apoptosis in anterior pituitary cells, possibly resulting from imbalanced cellular redox status.
Collapse
Affiliation(s)
- Sonia A Ronchetti
- Facultad de Medicina, Instituto de Investigaciones Biomédicas (UBA-CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María S Bianchi
- Instituto de Biología y Medicina Experimental-CONICET, Buenos Aires, Argentina
| | - Beatriz H Duvilanski
- Facultad de Medicina, Instituto de Investigaciones Biomédicas (UBA-CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jimena P Cabilla
- Facultad de Medicina, Instituto de Investigaciones Biomédicas (UBA-CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
19
|
Guidarelli A, Fiorani M, Azzolini C, Cerioni L, Scotti M, Cantoni O. U937 cell apoptosis induced by arsenite is prevented by low concentrations of mitochondrial ascorbic acid with hardly any effect mediated by the cytosolic fraction of the vitamin. Biofactors 2015; 41:101-10. [PMID: 25809564 DOI: 10.1002/biof.1204] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/12/2015] [Indexed: 02/03/2023]
Abstract
Arsenite directly triggers cytochrome c and Smac/DIABLO release in mitochondria isolated from U937 cells. These effects were not observed in mitochondria pre-exposed for 15 min to 10 µM L-ascorbic acid (AA). In other experiments, intact cells treated for 24-72 h with arsenite were found to die by apoptosis through a mechanism involving mitochondrial permeability transition. Pre-exposure (15 min) to low micromolar concentrations of AA and dehydroascorbic acid (DHA), resulting in identical cytosolic levels of the vitamin, had a diverse impact on cell survival, as cytoprotection was only observed after treatment with AA. Also the mitochondrial accumulation of the vitamin was restricted to AA exposure. An additional indication linking cytoprotection to the mitochondrial fraction of the vitamin was obtained in experiments measuring susceptibility to arsenite in parallel with loss of mitochondrial and cytosolic AA at different times after vitamin exposure. Finally, we took advantage of our recent findings that DHA potently inhibits AA transport to demonstrate that DHA abolishes all the protective effects of AA, under the same conditions in which the mitochondrial accumulation of the vitamin is prevented without affecting the overall cellular accumulation of the vitamin.
Collapse
Affiliation(s)
- Andrea Guidarelli
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", Urbino, Italy
| | | | | | | | | | | |
Collapse
|
20
|
Zhang Z, Pratheeshkumar P, Budhraja A, Son YO, Kim D, Shi X. Role of reactive oxygen species in arsenic-induced transformation of human lung bronchial epithelial (BEAS-2B) cells. Biochem Biophys Res Commun 2014; 456:643-8. [PMID: 25499816 DOI: 10.1016/j.bbrc.2014.12.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 12/04/2014] [Indexed: 11/27/2022]
Abstract
Arsenic is an environmental carcinogen, its mechanisms of carcinogenesis remain to be investigated. Reactive oxygen species (ROS) are considered to be important. A previous study (Carpenter et al., 2011) has measured ROS level in human lung bronchial epithelial (BEAS-2B) cells and arsenic-transformed BEAS-2B cells and found that ROS levels were higher in transformed cells than that in parent normal cells. Based on these observations, the authors concluded that cell transformation induced by arsenic is mediated by increased cellular levels of ROS. This conclusion is problematic because this study only measured the basal ROS levels in transformed and parent cells and did not investigate the role of ROS in the process of arsenic-induced cell transformation. The levels of ROS in arsenic-transformed cells represent the result and not the cause of cell transformation. Thus question concerning whether ROS are important in arsenic-induced cell transformation remains to be answered. In the present study, we used expressions of catalase (antioxidant against H2O2) and superoxide dismutase 2 (SOD2, antioxidant against O2(-)) to decrease ROS level and investigated their role in the process of arsenic-induced cell transformation. Our results show that inhibition of ROS by antioxidant enzymes decreased arsenic-induced cell transformation, demonstrating that ROS are important in this process. We have also shown that in arsenic-transformed cells, ROS generation was lower and levels of antioxidants are higher than those in parent cells, in a disagreement with the previous report. The present study has also shown that the arsenic-transformed cells acquired apoptosis resistance. The inhibition of catalase to increase ROS level restored apoptosis capability of arsenic-transformed BEAS-2B cells, further showing that ROS levels are low in these cells. The apoptosis resistance due to the low ROS levels may increase cells proliferation, providing a favorable environment for tumorigenesis of arsenic-transformed cells.
Collapse
Affiliation(s)
- Zhuo Zhang
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY 40536, USA.
| | - Poyil Pratheeshkumar
- Center for Research on Environmental Diseases, University of Kentucky, Lexington, KY 40536, USA
| | - Amit Budhraja
- Center for Research on Environmental Diseases, University of Kentucky, Lexington, KY 40536, USA
| | - Young-Ok Son
- Center for Research on Environmental Diseases, University of Kentucky, Lexington, KY 40536, USA
| | - Donghern Kim
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY 40536, USA
| | - Xianglin Shi
- Center for Research on Environmental Diseases, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
21
|
Sumedha NC, Miltonprabu S. Retracted: Cardiac mitochondrial oxidative stress and dysfunction induced by arsenic and
its amelioration by diallyl trisulphide. Toxicol Res (Camb) 2014. [DOI: 10.1039/c4tx00097h] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Naorem Chanu Sumedha
- Department of Zoology, Annamalai University, Annamalai Nagar-608002, Tamilnadu, India. Fax: +91 04144-238080; Tel: +91 04144-238282, +91 9842325222
| | - Selvaraj Miltonprabu
- Department of Zoology, Annamalai University, Annamalai Nagar-608002, Tamilnadu, India. Fax: +91 04144-238080; Tel: +91 04144-238282, +91 9842325222
| |
Collapse
|
22
|
Qi Y, Li H, Zhang M, Zhang T, Frank J, Chen G. Autophagy in arsenic carcinogenesis. ACTA ACUST UNITED AC 2014; 66:163-8. [DOI: 10.1016/j.etp.2014.01.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 01/10/2014] [Accepted: 01/20/2014] [Indexed: 12/31/2022]
|
23
|
Regoli F, Giuliani ME. Oxidative pathways of chemical toxicity and oxidative stress biomarkers in marine organisms. MARINE ENVIRONMENTAL RESEARCH 2014; 93:106-17. [PMID: 23942183 DOI: 10.1016/j.marenvres.2013.07.006] [Citation(s) in RCA: 593] [Impact Index Per Article: 53.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 07/16/2013] [Accepted: 07/17/2013] [Indexed: 05/04/2023]
Abstract
The antioxidant system of marine organisms consists of low molecular weight scavengers and antioxidant enzymes which interact in a sophisticated network. Environmental pollutants can unbalance this system through closely related mechanisms, indirect relationships and cascade effects acting from pre-transcriptional to catalytic levels. Chemically-mediated pathways have the potential to greatly enhance intracellular formation of reactive oxygen species (ROS); at the same time, excessive levels of oxyradicals down-regulate xenobiotics metabolism, with important environmental implications for organisms exposed to chemical mixtures. Interactions between different classes of chemicals, generation of ROS and onset of oxidative stress conditions are partly modulated by changes in levels and functions of redox-sensitive signaling proteins and transcription factors. The Nrf2-Keap1 pathway still remains largely unexplored in marine organisms, despite the elevated degree of identity and similarity with homolog transcripts and proteins from different species. Recent evidences on transcriptional up-regulation of this system are consistent with the capability to provide a prolonged expression of ARE-regulated cytoprotective genes, and to efficiently switch off this mechanism when oxidative pressure decreases. Although gene expression and catalytic activities of antioxidants are often measured as alternative biomarkers in monitoring biological effects of contaminants, conflicting results between molecular and biochemical responses are quite frequent. The links between effects occurring at various intracellular levels can be masked by non-genomic processes affecting mRNA stability and protein turnover, different timing for transcriptional and translational mechanisms, metabolic capability of tissues, post-transcriptional modifications of proteins, bi-phasic responses of antioxidant enzymes and interactions occurring in chemical mixtures. In this respect, caution should be taken in monitoring studies where mRNA levels of antioxidants could represent a snapshot of cell activity at a given time, not an effective endpoint of environmental pollutants.
Collapse
Affiliation(s)
- Francesco Regoli
- Dipartimento di Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, Via Ranieri Monte d'Ago, Ancona 60100, Italy.
| | - Maria Elisa Giuliani
- Dipartimento di Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, Via Ranieri Monte d'Ago, Ancona 60100, Italy
| |
Collapse
|
24
|
Ramsey KA, Foong RE, Sly PD, Larcombe AN, Zosky GR. Early life arsenic exposure and acute and long-term responses to influenza A infection in mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2013; 121:1187-93. [PMID: 23968752 PMCID: PMC3801203 DOI: 10.1289/ehp.1306748] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 07/23/2013] [Indexed: 05/04/2023]
Abstract
BACKGROUND Arsenic is a significant global environmental health problem. Exposure to arsenic in early life has been shown to increase the rate of respiratory infections during infancy, reduce childhood lung function, and increase the rates of bronchiectasis in early adulthood. OBJECTIVE We aimed to determine if early life exposure to arsenic exacerbates the response to early life influenza infection in mice. METHODS C57BL/6 mice were exposed to arsenic in utero and throughout postnatal life. At 1 week of age, a subgroup of mice were infected with influenza A. We then assessed the acute and long-term effects of arsenic exposure on viral clearance, inflammation, lung structure, and lung function. RESULTS Early life arsenic exposure reduced the clearance of and exacerbated the inflammatory response to influenza A, and resulted in acute and long-term changes in lung mechanics and airway structure. CONCLUSIONS Increased susceptibility to respiratory infections combined with exaggerated inflammatory responses throughout early life may contribute to the development of bronchiectasis in arsenic-exposed populations.
Collapse
Affiliation(s)
- Kathryn A Ramsey
- Division of Clinical Sciences, Telethon Institute for Child Health Research, Subiaco, Western Australia, Australia
| | | | | | | | | |
Collapse
|
25
|
Lee CH, Wu SB, Hong CH, Chen GS, Wei YH, Yu HS. Involvement of mtDNA Damage Elicited by Oxidative Stress in the Arsenical Skin Cancers. J Invest Dermatol 2013; 133:1890-900. [DOI: 10.1038/jid.2013.55] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
26
|
Bhattacharjee P, Chatterjee D, Singh KK, Giri AK. Systems biology approaches to evaluate arsenic toxicity and carcinogenicity: an overview. Int J Hyg Environ Health 2013; 216:574-86. [PMID: 23340121 DOI: 10.1016/j.ijheh.2012.12.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 12/11/2012] [Accepted: 12/13/2012] [Indexed: 01/08/2023]
Abstract
Long term exposure to arsenic, either through groundwater, food stuff or occupational sources, results in a plethora of dermatological and non-dermatological health effects including multi-organ cancer and early mortality. Several epidemiological studies, across the globe have reported arsenic-induced health effects and cancerous outcomes; but the prevalence of such diseases varies depending on environmental factors (geographical location, exposure level), and genetic makeup (and variants thereof); which is further modulated by several other factors like ethnicity, age-sex, smoking status, diet, etc. It is also interesting to note that, chronic arsenic exposure to a similar extent, even among the same family members, result in wide inter-individual variations. To understand the adverse effect of this toxic metabolite on biological system (cellular targets), and to unravel the underlying molecular basis (at the level of transcript, proteome, or metabolite), a holistic, systems biology approach was taken. Due to the paradoxical nature and unavailability of any suitable animal model system; the literature review is primarily based on cell line and population based studies. Thus, here we present a comprehensive review on the systems biology approaches to explore the underlying mechanism of arsenic-induced carcinogenicity, along with our own observations and an overview of mitigation strategies and their effectiveness till date.
Collapse
Affiliation(s)
- Pritha Bhattacharjee
- Molecular and Human Genetics Division, Indian Institute of Chemical Biology, Kolkata, India
| | | | | | | |
Collapse
|
27
|
Pei Q, Ma N, Zhang J, Xu W, Li Y, Ma Z, Li Y, Tian F, Zhang W, Mu J, Li Y, Wang D, Liu H, Yang M, Ma C, Yun F. Oxidative DNA damage of peripheral blood polymorphonuclear leukocytes, selectively induced by chronic arsenic exposure, is associated with extent of arsenic-related skin lesions. Toxicol Appl Pharmacol 2013; 266:143-9. [DOI: 10.1016/j.taap.2012.10.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 10/27/2012] [Accepted: 10/30/2012] [Indexed: 10/27/2022]
|
28
|
Li Y, Gao Y, Zhao L, Wei Y, Feng H, Wang C, Wei W, Ding Y, Sun D. Changes in serum thioredoxin among individuals chronically exposed to arsenic in drinking water. Toxicol Appl Pharmacol 2012; 259:124-32. [DOI: 10.1016/j.taap.2011.12.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Revised: 12/03/2011] [Accepted: 12/13/2011] [Indexed: 11/27/2022]
|
29
|
Ghosh S, Dungdung SR, Chowdhury ST, Mandal AK, Sarkar S, Ghosh D, Das N. Encapsulation of the flavonoid quercetin with an arsenic chelator into nanocapsules enables the simultaneous delivery of hydrophobic and hydrophilic drugs with a synergistic effect against chronic arsenic accumulation and oxidative stress. Free Radic Biol Med 2011; 51:1893-902. [PMID: 21914470 DOI: 10.1016/j.freeradbiomed.2011.08.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 08/19/2011] [Accepted: 08/19/2011] [Indexed: 11/18/2022]
Abstract
Chronic arsenic exposure causes oxidative stress and mitochondrial dysfunction in the liver and brain. The ideal treatment would be to chelate arsenic and prevent oxidative stress. meso-2,3-Dimercaptosuccinic acid (DMSA) is used to chelate arsenic but its hydrophilicity makes it membrane-impermeative. Conversely, quercetin (QC) is a good antioxidant with limited clinical application because of its hydrophobic nature and limited bioavailability, and it is not possible to solubilize these two compounds in a single nontoxic solvent. Nanocapsules have emerged as a potent drug delivery system and make it feasible to incorporate both hydrophilic and lipophilic compounds. Nanoencapsulated formulations with QC and DMSA either alone or coencapsulated in polylactide-co-glycolide [N(QC+DMSA)] were synthesized to explore their therapeutic application in a rat model of chronic arsenic toxicity. These treatments were compared to administration of quercetin or DMSA alone using conventional delivery methods. Both nanoencapsulated quercetin and nanoencapsulated DMSA were more effective at decreasing oxidative injury in liver or brain compared to conventional delivery methods, but coencapsulation of quercetin and DMSA into nanoparticles had a marked synergistic effect, decreasing liver and brain arsenic levels from 9.5 and 4.8μg/g to 2.2 and 1.5μg/g, respectively. Likewise, administration of coencapsulated quercetin and DMSA virtually normalized changes in mitochondrial function, formation of reactive oxygen species, and liver injury. We conclude that coencapsulation of quercetin and DMSA may provide a more effective therapeutic strategy in the management of arsenic toxicity and also presents a novel way of combining hydrophilic and hydrophobic drugs into a single delivery system.
Collapse
Affiliation(s)
- Swarupa Ghosh
- Biomembrane Division, Indian Institute of Chemical Biology, Kolkata 700032, India
| | | | | | | | | | | | | |
Collapse
|
30
|
Lee CH, Wu SB, Hong CH, Liao WT, Wu CY, Chen GS, Wei YH, Yu HS. Aberrant cell proliferation by enhanced mitochondrial biogenesis via mtTFA in arsenical skin cancers. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:2066-76. [PMID: 21514422 DOI: 10.1016/j.ajpath.2011.01.056] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Revised: 01/14/2011] [Accepted: 01/24/2011] [Indexed: 11/26/2022]
Abstract
Arsenic-induced Bowen's disease (As-BD), a cutaneous carcinoma in situ, is thought to arise from gene mutation and uncontrolled proliferation. However, how mitochondria regulate the arsenic-induced cell proliferation remains unclear. The aim of this study was to clarify whether arsenic interfered with mitochondrial biogenesis and function, leading to aberrant cell proliferation in As-BD. Skin biopsy samples from patients with As-BD and controls were stained for cytochrome c oxidase (Complex IV), measured for mitochondrial DNA (mtDNA) copy number and the expression levels of mitochondrial biogenesis-related genes, including peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), nuclear respiratory factor 1 (NRF-1), and mitochondrial transcription factor A (mtTFA). The results showed that expression of cytochrome c oxidase, mtTFA, NRF-1, and PGC-1α was increased in As-BD compared with in healthy subjects. Treatment of primary keratinocytes with arsenic at concentrations lower than 1.0 μmol/L induced cell proliferation, along with enhanced mitochondrial biogenesis. Furthermore, we observed that the mitochondrial oxygen consumption rate and intracellular ATP level were increased in arsenic-treated keratinocytes. Blocking of mitochondrial function by oligomycin A (Complex V inhibitor) or knockdown of mtTFA by RNA interference abrogated arsenic-induced cell proliferation without affecting cyclin D1 expression. We concluded that mtTFA up-regulation, augmented mitochondrial biogenesis, and enhanced mitochondrial functions may contribute to arsenic-induced cell proliferation. Targeting mitochondrial biogenesis may help treat arsenical cancers at the stage of cell proliferation.
Collapse
Affiliation(s)
- Chih-Hung Lee
- Department of Dermatology, Graduate Institute of Medicine, Center of Excellence for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Flora SJS. Arsenic-induced oxidative stress and its reversibility. Free Radic Biol Med 2011; 51:257-281. [PMID: 21554949 DOI: 10.1016/j.freeradbiomed.2011.04.008] [Citation(s) in RCA: 551] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2010] [Revised: 03/18/2011] [Accepted: 04/04/2011] [Indexed: 12/12/2022]
Abstract
This review summarizes the literature describing the molecular mechanisms of arsenic-induced oxidative stress, its relevant biomarkers, and its relation to various diseases, including preventive and therapeutic strategies. Arsenic alters multiple cellular pathways including expression of growth factors, suppression of cell cycle checkpoint proteins, promotion of and resistance to apoptosis, inhibition of DNA repair, alterations in DNA methylation, decreased immunosurveillance, and increased oxidative stress, by disturbing the pro/antioxidant balance. These alterations play prominent roles in disease manifestation, such as carcinogenicity, genotoxicity, diabetes, cardiovascular and nervous systems disorders. The exact molecular and cellular mechanisms involved in arsenic toxicity are rather unrevealed. Arsenic alters cellular glutathione levels either by utilizing this electron donor for the conversion of pentavalent to trivalent arsenicals or directly binding with it or by oxidizing glutathione via arsenic-induced free radical generation. Arsenic forms oxygen-based radicals (OH(•), O(2)(•-)) under physiological conditions by directly binding with critical thiols. As a carcinogen, it acts through epigenetic mechanisms rather than as a classical mutagen. The carcinogenic potential of arsenic may be attributed to activation of redox-sensitive transcription factors and other signaling pathways involving nuclear factor κB, activator protein-1, and p53. Modulation of cellular thiols for protection against reactive oxygen species has been used as a therapeutic strategy against arsenic. N-acetylcysteine, α-lipoic acid, vitamin E, quercetin, and a few herbal extracts show prophylactic activity against the majority of arsenic-mediated injuries in both in vitro and in vivo models. This review also updates the reader on recent advances in chelation therapy and newer therapeutic strategies suggested to treat arsenic-induced oxidative damage.
Collapse
Affiliation(s)
- Swaran J S Flora
- Division of Pharmacology & Toxicology, Defence Research and Development Establishment, Jhansi Road, Gwalior 474002, India.
| |
Collapse
|
32
|
Abstract
Arsenic is a metalloid that is considered to be a paradox in terms of its role both as a carcinogen and as a therapeutic agent. Chronic exposure to arsenic in drinking water has been linked with the development of various pathological conditions including cancer. Nevertheless, the therapeutic potential of arsenic and its derivatives in a variety of diseases have been exploited in the past. However, its role and mechanism of action as a therapeutic agent still remain an active area of research and investigation. Our ongoing work also suggests varied responses in cancer cells exposed to lower versus higher concentrations of arsenic. Furthermore, the arsenic combinations with chemopreventive or anticancer agents have been observed to sensitize the cell for cell-cycle arrest and cell death. Here, we have provided the account of recent updates on the mechanism of action of arsenic and its derivatives that lead to various disorders, and its role as a therapeutic agent both as a single agent as well as in combination chemotherapy.
Collapse
|
33
|
Datta P, Mukhopadhyay AP, Manna P, Tiekink ERT, Sil PC, Sinha C. Structure, photophysics, electrochemistry, DFT calculation, and in-vitro antioxidant activity of coumarin Schiff base complexes of Group 6 metal carbonyls. J Inorg Biochem 2011; 105:577-588. [PMID: 21419093 DOI: 10.1016/j.jinorgbio.2010.04.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Revised: 04/24/2010] [Accepted: 04/30/2010] [Indexed: 11/23/2022]
Abstract
N-[(2-Pyridyl)methyliden]-6-coumarin (L) is synthesized by the condensation of 6-aminocoumarin and pyridine-2-carboxaldehyde. Group-6 tetracarbonyl complexes, [M(CO)(4)(L)] (M=Cr, Mo, and W) are synthesized and characterized by mass spectrometry and NMR, FT-IR and UV-visible spectroscopy. X-ray crystal structure of [Cr(CO)(4)(L)] shows N(pyridine), N(imine) chelation to chromium(0). A supramolecular chain is formed by C-H⋯O and π⋯π interactions. The ligand and the complexes are fluorescent. Cyclic voltammetry of the complexes exhibit quasireversible M(I)/M(0) redox couple. The complexes exhibit potential antioxidant property both in cell free and in-vitro studies and highest activity is observed to [W(CO)(4)(L)]. Density functional theory (DFT) computation has been performed to correlate with the electronic configuration, composition of wave functions with the UV-visible spectra and redox properties.
Collapse
Affiliation(s)
- Papia Datta
- Department of Chemistry, Inorganic Chemistry Section, Jadavpur University, Kolkata, India
| | | | | | | | | | | |
Collapse
|
34
|
Potential molecular mechanisms for combined toxicity of arsenic and alcohol. J Inorg Biochem 2010; 104:1229-33. [PMID: 20817264 DOI: 10.1016/j.jinorgbio.2010.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Revised: 07/30/2010] [Accepted: 08/06/2010] [Indexed: 12/26/2022]
Abstract
Arsenic is a ubiquitous environmental factor that has been identified as a risk factor for a wide range of human diseases. Alcohol is clearly a toxic substance when consumed in excess. Alcohol abuse results in a variety of pathological effects, including damages to liver, heart, and brain, as well as other organs, and is associated with an increased risk of certain types of cancers. In history, arsenic-contaminated beers caused severe diseases. There are populations who are exposed to relatively high levels of arsenic in their drinking water and consume alcohol at the same time. In this focused review, we aim to discuss important molecular mechanisms responsible for arsenic toxicity and potential combined toxic effects of alcohol and arsenic.
Collapse
|
35
|
Kozul CD, Hampton TH, Davey JC, Gosse JA, Nomikos AP, Eisenhauer PL, Weiss DJ, Thorpe JE, Ihnat MA, Hamilton JW. Chronic exposure to arsenic in the drinking water alters the expression of immune response genes in mouse lung. ENVIRONMENTAL HEALTH PERSPECTIVES 2009; 117:1108-15. [PMID: 19654921 PMCID: PMC2717138 DOI: 10.1289/ehp.0800199] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Accepted: 03/04/2009] [Indexed: 05/02/2023]
Abstract
BACKGROUND Chronic exposure to drinking water arsenic is a significant worldwide environmental health concern. Exposure to As is associated with an increased risk of lung disease, which may make it a unique toxicant, because lung toxicity is usually associated with inhalation rather than ingestion. OBJECTIVES The goal of this study was to examine mRNA and protein expression changes in the lungs of mice exposed chronically to environmentally relevant concentrations of As in the food or drinking water, specifically examining the hypothesis that As may preferentially affect gene and protein expression related to immune function as part of its mechanism of toxicant action. METHODS C57BL/6J mice fed a casein-based AIN-76A defined diet were exposed to 10 or 100 ppb As in drinking water or food for 5-6 weeks. RESULTS Whole genome transcriptome profiling of animal lungs revealed significant alterations in the expression of many genes with functions in cell adhesion and migration, channels, receptors, differentiation and proliferation, and, most strikingly, aspects of the innate immune response. Confirmation of mRNA and protein expression changes in key genes of this response revealed that genes for interleukin 1beta, interleukin 1 receptor, a number of toll-like receptors, and several cytokines and cytokine receptors were significantly altered in the lungs of As-exposed mice. CONCLUSIONS These findings indicate that chronic low-dose As exposure at the current U.S. drinking-water standard can elicit effects on the regulation of innate immunity, which may contribute to altered disease risk, particularly in lung.
Collapse
Affiliation(s)
- Courtney D. Kozul
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, New Hampshire, USA
- Center for Environmental Health Sciences, Dartmouth College, Hanover, New Hampshire, USA
| | - Thomas H. Hampton
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, New Hampshire, USA
- Center for Environmental Health Sciences, Dartmouth College, Hanover, New Hampshire, USA
| | - Jennifer C. Davey
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, New Hampshire, USA
- Center for Environmental Health Sciences, Dartmouth College, Hanover, New Hampshire, USA
| | - Julie A. Gosse
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, New Hampshire, USA
- Center for Environmental Health Sciences, Dartmouth College, Hanover, New Hampshire, USA
| | - Athena P. Nomikos
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, New Hampshire, USA
- Center for Environmental Health Sciences, Dartmouth College, Hanover, New Hampshire, USA
| | - Phillip L. Eisenhauer
- Vermont Lung Center, University of Vermont College of Medicine, Burlington, Vermont, USA
| | - Daniel J. Weiss
- Vermont Lung Center, University of Vermont College of Medicine, Burlington, Vermont, USA
| | - Jessica E. Thorpe
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Michael A. Ihnat
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Joshua W. Hamilton
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, New Hampshire, USA
- Center for Environmental Health Sciences, Dartmouth College, Hanover, New Hampshire, USA
- Address correspondence to J.W. Hamilton, Bay Paul Center in Comparative Molecular Biology and Evolution, Marine Biological Laboratory, 7 MBL St., Woods Hole, MA 02543 USA. Telephone: (508) 289-7300. Fax: (508) 289-7934. E-mail:
| |
Collapse
|
36
|
Wu JZ, Ho PC. Comparing the Relative Oxidative DNA Damage Caused by Various Arsenic Species by Quantifying Urinary Levels of 8-Hydroxy-2′-Deoxyguanosine with Isotope-Dilution Liquid Chromatography/Mass Spectrometry. Pharm Res 2009; 26:1525-33. [DOI: 10.1007/s11095-009-9865-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2008] [Accepted: 02/23/2009] [Indexed: 11/28/2022]
|
37
|
Fukuyama T, Ueda H, Hayashi K, Tajima Y, Shuto Y, Kosaka T, Harada T. Sensitizing Potential of Chromated Copper Arsenate in Local Lymph Node Assays Differs with the Solvent Used. J Immunotoxicol 2008; 5:99-106. [DOI: 10.1080/15476910802085715] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
38
|
Manna P, Sinha M, Sil PC. Arsenic-induced oxidative myocardial injury: protective role of arjunolic acid. Arch Toxicol 2008; 82:137-149. [PMID: 18197399 DOI: 10.1007/s00204-007-0272-8] [Citation(s) in RCA: 149] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2007] [Accepted: 12/06/2007] [Indexed: 11/24/2022]
Abstract
Arsenic, one of the most harmful metalloids, is ubiquitous in the environment. The present study has been carried out to investigate the protective role of a triterpenoid saponin, arjunolic acid (AA) against arsenic-induced cardiac oxidative damage. In the study, NaAsO2 was chosen as the source of arsenic. The free radical scavenging activity and the effect of AA on the intracellular antioxidant power were determined from its 2,2-diphenyl-1-picryl hydrazyl radical scavenging ability and ferric reducing/antioxidant power assay, respectively. Oral administration of NaAsO2 at a dose of 10 mg/kg body weight for 2 days caused significant accumulation of arsenic in cardiac tissues of the experimental mice in association with the reduction in cardiac antioxidant enzymes activities, namely superoxide dismutase, catalase, glutathione-S-transferase, glutathione reductase and glutathione peroxidase. Arsenic intoxication also decreased the cardiac glutathione (GSH) and total thiol contents and increased the levels of oxidized glutathione (GSSG), lipid peroxidation end products and protein carbonyl content. Treatment with AA at a dose of 20 mg/kg body weight for 4 days prior to NaAsO2 intoxication protected the cardiac tissue from arsenic-induced oxidative impairment. In addition to oxidative stress, arsenic administration increased total cholesterol level as well as the reduced high-density lipoprotein cholesterol level in the sera of the experimental mice. AA pretreatment, however, could prevent this hyperlipidemia. Histological studies on the ultrastructural changes in cardiac tissue supported the protective activity of AA also. Combining all, results suggest that AA could protect cardiac tissues against arsenic-induced oxidative stress probably due to its antioxidant property.
Collapse
Affiliation(s)
- Prasenjit Manna
- Department of Chemistry, Bose Institute, 93/1, Acharya Prafulla Chandra Road, Kolkata 700009, West Bengal, India
| | | | | |
Collapse
|
39
|
Vujcic M, Shroff M, Singh KK. Genetic determinants of mitochondrial response to arsenic in yeast Saccharomyces cerevisiae. Cancer Res 2007; 67:9740-9. [PMID: 17942904 DOI: 10.1158/0008-5472.can-07-1962] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have used yeast Saccharomyces cerevisiae as a tool to identify the importance of mitochondrial processes involved in arsenic-induced carcinogenicity in humans. We screened 466 single-gene knockout strains of yeast S. cerevisiae known to be involved in biogenesis of mitochondria for sodium arsenite (AsIII) and sodium arsenate (AsV) sensitivity. We identified 72 arsenite-sensitive and 81 arsenate-sensitive mutants. We categorized the identified mutants based on the various mitochondrial processes, including nucleic acid metabolism, oxidative phosphorylation, protein synthesis, and vacuolar acidification. We have identified 65 human orthologues to proteins involved in arsenite sensitivity and 3 human orthologues to arsenite resistance. Furthermore, 23 human orthologues to arsenate sensitivity and 20 human orthologues to arsenate-resistant proteins, including MSH3, COX10, GCSH, PPOX, and MTHFD1, were also identified. Using PathwayAssist software, we did cellular network analysis between identified mitochondrial proteins. Three types of interactions, (a) protein-protein interactions, (b) common transcriptional regulators, and (c) common target genes, were identified. We found that RTG (retrograde) genes involved in mitochondria-to-nucleus signaling regulate both arsenite sensitivity and resistance. Furthermore, our study revealed that ABF1, a multifunctional transcriptional factor, regulates genes involved in both arsenite and arsenate sensitivity and resistance. However, REB1 and RAP1 transcriptional regulators were common to only arsenate- and arsenite-sensitive genes, respectively. These studies indicate that multiple pathways involved in mitochondrial biogenesis protect yeast S. cerevisiae from arsenic-induced toxicity. Together, our studies suggest that evolutionary conserved mitochondrial networks identified in yeast S. cerevisiae must play an important role in arsenic-induced carcinogenesis in humans.
Collapse
Affiliation(s)
- Marija Vujcic
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | | | | |
Collapse
|
40
|
Hwang BJ, Utti C, Steinberg M. Induction of cyclin D1 by submicromolar concentrations of arsenite in human epidermal keratinocytes. Toxicol Appl Pharmacol 2006; 217:161-7. [PMID: 17005224 DOI: 10.1016/j.taap.2006.08.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Revised: 08/08/2006] [Accepted: 08/09/2006] [Indexed: 10/24/2022]
Abstract
Arsenic is a prevalent environmental carcinogen but arsenic is not directly mutagenic and the mechanism by which arsenite brings about oncogenic transformation is poorly understood. To gain insight into the oncogenic properties of arsenic, we studied the expression of cyclin D1 in cultured human epidermal keratinocytes treated with submicromolar concentrations of sodium arsenite. Arsenite at concentrations between 200 and 800 nM over a 3-day period brought about an increase in cell growth rate. Uptake of the vital stain, neutral red, arsenite at 200 and 400 nM concentrations brought about a parallel increase in cell viability over the same treatment period. Analysis of cell cycle parameters by flow cytometry showed that the growth stimulation was accompanied by a concomitant shift from the G1 into the S/G2 cell cycle compartment in the arsenite-treated cells. Real-time PCR analysis of cyclin D1 transcription showed that there was an induction of more than three-fold in cells exposed to 400 nM arsenite for 3 days. Quantitation of cyclin D levels in Western blots showed that arsenite treatment caused a time-dependent induction of cyclin D proteins representing an induction of about 2.0-fold after a 7 day treatment period. Electrophoretic mobility shift assays (EMSA) showed that arsenite also stimulated binding of the transcription factors, AP1 and CREBP to their respective binding motifs within 3 days. This supports a mechanism of oncogenesis based on persistent upregulation of D type cyclins leading to a concomitant loss of G1/S checkpoint control.
Collapse
Affiliation(s)
- Bor-Jang Hwang
- Department of Chemistry, The City College of the City University of New York, Convent Avenue and 138th Street, New York, NY 10031, USA
| | | | | |
Collapse
|
41
|
Tsou TC, Yeh SC, Tsai EM, Tsai FY, Chao HR, Chang LW. Arsenite enhances tumor necrosis factor-α-induced expression of vascular cell adhesion molecule-1. Toxicol Appl Pharmacol 2005; 209:10-8. [PMID: 16271621 DOI: 10.1016/j.taap.2005.03.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2004] [Revised: 01/29/2005] [Accepted: 03/14/2005] [Indexed: 11/20/2022]
Abstract
Epidemiological studies demonstrated a high association of vascular diseases with arsenite exposure. We hypothesize that arsenite potentiates the effect of proinflammatory cytokines on vascular endothelial cells, and hence contributes to atherosclerosis. In this study, we investigated the effect of arsenite and its induction of glutathione (GSH) on vascular cell adhesion molecule-1 (VCAM-1) protein expression in human umbilical vein endothelial cells (HUVECs) in response to tumor necrosis factor-alpha (TNF-alpha), a typical proinflammatory cytokine. Our study demonstrated that arsenite pretreatment potentiated the TNF-alpha-induced VCAM-1 expression with up-regulations of both activator protein-1 (AP-1) and nuclear factor-kappaB (NF-kappaB). To elucidate the role of GSH in regulation of AP-1, NF-kappaB, and VCAM-1 expression, we employed l-buthionine (S,R)-sulfoximine (BSO), a specific gamma-glutamylcysteine synthetase (gamma-GCS) inhibitor, to block intracellular GSH synthesis. Our investigation revealed that, by depleting GSH, arsenite attenuated the TNF-alpha-induced VCAM-1 expression as well as a potentiation of AP-1 and an attenuation of NF-kappaB activations by TNF-alpha. Moreover, we found that depletion of GSH would also attenuate the TNF-alpha-induced VCAM-1 expression with a down-regulation of the TNF-alpha-induced NF-kappaB activation and without significant effect on AP-1. On the other hand, the TNF-alpha-induced VCAM-1 expression could be completely abolished by inhibition of AP-1 or NF-kappaB activity, suggesting that activation of both AP-1 and NF-kappaB was necessary for VCAM-1 expression. In summary, we demonstrate that arsenite enhances the TNF-alpha-induced VCAM-1 expression in HUVECs via regulation of AP-1 and NF-kappaB activities in a GSH-sensitive manner. Our present study suggested a potential mechanism for arsenite in the induction of vascular inflammation and vascular diseases via modulating the actions of proinflammatory cytokines.
Collapse
Affiliation(s)
- Tsui-Chun Tsou
- Division of Environmental Health and Occupational Medicine, National Health Research Institutes, 100 Shih-Chuan 1st Road, Kaohsiung 807, Taiwan, ROC.
| | | | | | | | | | | |
Collapse
|
42
|
Hermann AC, Kim CH. Effects of arsenic on zebrafish innate immune system. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2005; 7:494-505. [PMID: 16007375 DOI: 10.1007/s10126-004-4109-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2004] [Accepted: 12/23/2004] [Indexed: 05/02/2023]
Abstract
The innate immune response, the first line of defense against invading pathogens, can be perturbed by environmental toxicants such as arsenic. This study reports the effects of arsenic on innate immunity of zebrafish. Respiratory burst activity, messenger RNA expression of tumor necrosis factor alpha (TNF-alpha), a primer of the respiratory burst response, and mRNA expression of the antiviral cytokines interferon (IFN) and MX, : before and after viral infection, were examined in arsenic-exposed zebrafish larvae. Respiratory burst activity and TNF-alpha expression were decreased upon arsenic exposure, indicating inhibition of TNF-alpha priming of the respiratory burst response. Arsenic enhanced IFN expression slightly over time, but reduced MX : expression. In zebrafish infected with snakehead rhabdovirus, arsenic decreased induction and altered the kinetics of IFN and MX : upon infection. Differences in IFN and MX : expression in arsenic-exposed larvae point toward an interruption of the Janus kinase-signal transducer and activator of transcription (JAK/STAT) pathway.
Collapse
Affiliation(s)
- Andrea C Hermann
- Department of Biochemistry, Microbiology, and Molecular Biology, University of Maine, 5735 Hitchner Hall, Orono, ME 04469, USA
| | | |
Collapse
|
43
|
Yang C, Wu J, Zhang R, Zhang P, Eckard J, Yusuf R, Huang X, Rossman TG, Frenkel K. Caffeic acid phenethyl ester (CAPE) prevents transformation of human cells by arsenite (As) and suppresses growth of As-transformed cells. Toxicology 2005; 213:81-96. [PMID: 16085347 DOI: 10.1016/j.tox.2005.05.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2005] [Revised: 04/14/2005] [Accepted: 05/18/2005] [Indexed: 10/25/2022]
Abstract
Recent evidence suggests that inflammatory cytokines and growth factors contribute to arsenite (As)-induced human carcinogenesis. We investigated the expression of inflammatory cytokine mRNAs during the transformation process induced by chronic As exposure in non-tumorigenic human osteogenic sarcoma (N-HOS) cells using gene arrays, and results were confirmed by RT-PCR and protein arrays. Caffeic acid phenethyl ester (CAPE), a naturally occurring immunomodulating agent, was used to evaluate the role of inflammatory factors in the process of As-mediated N-HOS cell transformation and in As-transformed HOS (AsT-HOS) cells. We found that an 8-week continuous exposure of N-HOS to 0.3 microM arsenite resulted in HOS cell transformation. That exposure also caused substantial decreases in inflammatory cytokine mRNAs, such as interleukin (IL) IL-1alpha, IL-2, IL-8, IL-18, MCP-1, TGF-beta2, and TNF-alpha, while it increased c-jun mRNA in a time-dependent manner. Co-incubation of N-HOS with As and CAPE (0.5-2.5 microM) prevented As-mediated declines in cytokine mRNAs in the co-treated cells, as well as their transformation to anchorage independence, while it caused decreases in c-jun mRNA. CAPE (up to 10 microM) had no effect on growth of N-HOS cells. However, CAPE (1-10 microM) treatment of AsT-HOS cells inhibited cell growth, induced cell cycle G2/M arrest, and triggered apoptosis, accompanied by changes in cytokine gene expression, as well as decreases in cyclin B1 and cdc2 abundance. Resveratrol (RV) and (-)(.) epigallocatechin gallate (EGCG), preventive agents present in grapes and green tea, respectively, induced similar changes in AsT-HOS cell growth but required much higher doses than CAPE to cause 50% growth arrest (<2.5 microM CAPE versus 25 microM RV or 50 microM EGCG). Overall, our findings suggest that inflammatory cytokines play an important role in the suppressive effects of CAPE on As-induced cell transformation and in the selective cytotoxicity of CAPE to As-transformed HOS cells.
Collapse
Affiliation(s)
- Chengfeng Yang
- Department of Environmental Medicine and NYU Cancer Institute, NYU School of Medicine, New York, NY 10016, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Felix K, Manna SK, Wise K, Barr J, Ramesh GT. Low levels of arsenite activates nuclear factor-kappaB and activator protein-1 in immortalized mesencephalic cells. J Biochem Mol Toxicol 2005; 19:67-77. [PMID: 15849723 PMCID: PMC2743884 DOI: 10.1002/jbt.20062] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Degeneration of dopaminergic neurons is one of the major features of Parkinson's disease. Many redox-active metals such as iron and manganese have been implicated in neuronal degeneration characterized by symptoms resembling Parkinson's disease. Even though, arsenic, which is another redox-active metal, has been shown to affect the central monoaminergic systems, but its potential in causing dopaminergic cell degeneration has not been fully known. Hence, the present study was designed to investigate arsenic signaling especially that is mediated by reactive oxygen species and its effect on early transcription factors in dopamine producing mesencephalic cell line 1RB3AN27. These mesencephalic cells were treated with low concentrations of sodium arsenite (0.1, 0.5, 1, 5, and 10 microM) and incubated for different periods of time (0-4 h). Arsenite was cytotoxic at 5 and 10 microM concentrations only after 72-h incubation period. Arsenite, in a dose-dependent manner, induced generation of reactive oxygen species (ROS) and activation of early transcription factors such as nuclear factor-kappa B (NF-kappaB) and activator protein-1 (AP-1) as shown by electro mobility shift assay. Incubation of antioxidants, either N-acetyl-L-cysteine (50 microM) or alpha-tocopherol (50 microM) with 1 microM arsenite, suppressed ROS generation. Arsenite at 1 microM concentration was sufficient for maximal activation of NF-kappaB and AP-1 activation. Time kinetics studies showed maximal activation of NF-kappaB by 1 microM concentration of arsenite was seen at 120 min and correlated with complete degradation of Ikappa Balpha at 60 min. Similarly, maximal activation of AP-1 by 1 microM concentration of arsenite occurred at 120 min. N-acetyl-L-cysteine at 50 microM concentration inhibited arsenite-induced NF-kappa B and AP-1. In addition, arsenite was shown to induce phosphorylation of extracellular signal regulated kinase (ERK) 1/2 at concentrations of 1 microM and above. These results suggest that arsenite, at low and subcytoxic concentrations, appears to induce oxidative stress leading to activation of early transcription factors whereas addition of antioxidant inhibited the activation of these factors.
Collapse
Affiliation(s)
- Kumar Felix
- Molecular Neurotoxicology Laboratory, Department of Biology, Texas Southern University, Houston, TX 77004, USA
| | | | | | | | | |
Collapse
|
45
|
Lee C, Lee YM, Rice RH. Human epidermal cell protein responses to arsenite treatment in culture. Chem Biol Interact 2005; 155:43-54. [PMID: 15899475 DOI: 10.1016/j.cbi.2005.04.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2004] [Revised: 04/14/2005] [Accepted: 04/14/2005] [Indexed: 11/16/2022]
Abstract
Study of the responses of target cells in culture is anticipated to help understand the mechanisms by which inorganic arsenic causes pathological effects in vivo. Treatment of human epidermal cells with arsenic has been shown to produce a myriad of changes in gene transcription. Present work focused on finding the extent of arsenite-induced changes in the protein pattern and whether global effects on protein sulfhydryls were evident. First, examining the profile of protein expression by two-dimensional gel electrophoresis indicated that approximately 40% of the 300 distinct protein spots that were monitored changed by at least two-fold in amount all through a 9-day exposure period. Second, examining soluble extracts of the treated cells by Activated Thiol Sepharose column chromatography gave little indication of change in the overall protein thiol content. Finally, among the 10 proteins identified that showed prominent changes in amount as a result of treatment for 1 or 4 days, enzymes of the glycolytic pathway were seen to be substantially elevated as a result of treatment, suggesting decreased utilization by the cells of oxidative phosphorylation. Since these changes were more conspicuous at the protein level than in previous transcriptional studies, the results emphasize the importance of proteomic analysis to complement transcriptional analysis of cell responses to perturbation by arsenic.
Collapse
Affiliation(s)
- Chan Lee
- Department of Food Science and Technology, Chung-Ang University, Ansung, Republic of Korea
| | | | | |
Collapse
|
46
|
Qian Y, Liu KJ, Chen Y, Flynn DC, Castranova V, Shi X. Cdc42 Regulates Arsenic-induced NADPH Oxidase Activation and Cell Migration through Actin Filament Reorganization. J Biol Chem 2005; 280:3875-84. [PMID: 15492012 DOI: 10.1074/jbc.m403788200] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although arsenic is a human carcinogen, the molecular mechanisms of its action remain to be understood. The present study reports that exposure to arsenic induced actin filament reorganization, resulting in lamellipodia and filopodia structures through the activation of Cdc42 in SVEC4-10 endothelial cells. It was also found that arsenic induced the formation of the superoxide anion (O2*) in SVEC4-10 cells. Immunoprecipitation and Western blotting analysis demonstrated that arsenic stimulation induced serine phosphorylation of p47phox, a key component of NADPH oxidase, indicating that arsenic induces O2* formation through NADPH oxidase activation. Inhibition of arsenic-induced actin filament reorganization by either overexpression of a dominant negative Cdc42 or pretreatment of an actin filament stabilizing regent, jasplakinolide, abrogated arsenic-induced NADPH oxidase activation, showing that the activation of NADPH oxidase was regulated by Cdc42-mediated actin filament reorganization. This study also showed that overexpression of a dominant negative Rac1 was sufficient to abolish arsenic-induced O2*- production, implying that Rac1 activities are required for Cdc42-mediated NADPH oxidase activation in response to arsenic stimulation. Furthermore, arsenic stimulation induced cell migration, which can be inhibited by the inactivation of either Cdc42 or NADPH oxidase. Taken together, the results indicate that arsenic is able to activate NADPH oxidase through Cdc42-mediated actin filament reorganization, leading to the induction of an increase in cell migration in SVEC4-10 endothelial cells.
Collapse
Affiliation(s)
- Yong Qian
- Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia 26505, USA.
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
Arsenic is a metalloid compound that is widely distributed in the environment. Human exposure of this compound has been associated with increased cancer incidence. Although the exact mechanisms remain to be investigated, numerous carcinogenic pathways have been proposed. Potential carcinogenic actions for arsenic include oxidative stress, genotoxic damage, DNA repair inhibition, epigenetic events, and activation of certain signal transduction pathways leading to abberrant gene expression. In this article, we summarize current knowledge on the molecular mechanisms of arsenic carcinogenesis with an emphasis on ROS and signal transduction pathways.
Collapse
Affiliation(s)
- Chuanshu Huang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA.
| | | | | | | |
Collapse
|
48
|
Abstract
Arsenic is a known toxin and carcinogen that is present in industrial settings and in the environment. The mechanisms of disease initiation and progression are not fully understood. In the last a few years, there has been increasing evidence of the correlation between the generation of reactive oxygen species (ROS), DNA damage, tumor promotion, and arsenic exposure. This article summarizes the current literature on the arsenic mediated generation of ROS and reactive nitrogen species (RNS) in various biological systems. This article also discusses the role of ROS and RNS in arsenic-induced DNA damage and activation of oxidative sensitive gene expression.
Collapse
Affiliation(s)
- Honglian Shi
- College of Pharmacy, University of New Mexico, Albuquerque, NM 87131, USA
| | | | | |
Collapse
|
49
|
Shi H, Hudson LG, Liu KJ. Oxidative stress and apoptosis in metal ion-induced carcinogenesis. Free Radic Biol Med 2004; 37:582-93. [PMID: 15288116 DOI: 10.1016/j.freeradbiomed.2004.03.012] [Citation(s) in RCA: 172] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2003] [Accepted: 03/19/2004] [Indexed: 12/25/2022]
Abstract
Epidemiological evidence suggests that exposure to certain metals causes carcinogenesis. The mechanisms of metal-induced carcinogenesis have been pursued in chemical, biochemical, cellular, and animal models. Significant evidence has accumulated that oxidative stress may be a common pathway in cellular responses to exposure to different metals. For example, in the last few years evidence in support of a correlation between the generation of reactive oxygen species, DNA damage, tumor promotion, and arsenic exposure has strengthened. This article summarizes the current literature on metal-mediated oxidative stress, apoptosis, and their relation to metal-mediated carcinogenesis, concentrating on arsenic and chromium.
Collapse
Affiliation(s)
- Honglian Shi
- College of Pharmacy, University of New Mexico, Albuquerque, NM 87131, USA
| | | | | |
Collapse
|
50
|
Shi H, Hudson LG, Ding W, Wang S, Cooper KL, Liu S, Chen Y, Shi X, Liu KJ. Arsenite Causes DNA Damage in Keratinocytes Via Generation of Hydroxyl Radicals. Chem Res Toxicol 2004; 17:871-8. [PMID: 15257611 DOI: 10.1021/tx049939e] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Arsenic is an environmental and occupational toxin. Dermatologic toxicities due to arsenic exposure are well-documented and include basal cell and squamous cell carcinomas. However, the mechanism of arsenic-induced skin cancer is not well-understood. Recent studies indicate that arsenic exposure results in the generation of reactive oxygen species (ROS) and oxidative stress. Here, we examined the chemical nature of the specific ROS, studied the interrelationship among these species, and identified the specific species that is responsible for the subsequent DNA damage in a spontaneously immortalized keratinocyte cell line. We detected the formation of O(2)(*)(-) and H(2)O(2) in keratinocytes incubated with arsenite [As(III)] but not with arsenate. As(III)-induced DNA damage was detected in a concentration-dependent manner and evident at low micromolar concentrations. Catalase, an H(2)O(2) scavenger, eliminated H(2)O(2) and reduced the As(III)-mediated DNA damage. Superoxide dismutase, by enhancing the production of H(2)O(2) and (*)OH, significantly increased the As(III)-mediated DNA damage. Sodium formate, a competitive scavenger for (*)OH, and deferoxamine, a metal chelator, both reduced the DNA damage. These results suggest that exposure to arsenite generates O(2)(*)(-) and H(2)O(2), and (*)OH, derived from H(2)O(2), is responsible, at least in part, for the observed DNA damage. These findings demonstrate arsenic-induced formation of specific ROS and provide the direct evidence of (*)OH-mediated DNA damage in keratinocytes, which may play an important role in the mechanism for arsenic-induced skin carcinogenicity.
Collapse
Affiliation(s)
- Honglian Shi
- Program in Toxicology, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, USA
| | | | | | | | | | | | | | | | | |
Collapse
|