1
|
Andrade-Feraud CM, Acanda de la Rocha AM, Berlow NE, Duque S, Velazco A, Castillo D, Holcomb B, Coats ER, Ghurani YR, Lucey CM, Pearson B, Guilarte TR, Azzam DJ. Chronic arsenic exposure of ovarian surface and fallopian tube cultures induces giant and/or multinucleated cells with phagocytosis-like properties and an inflammatory phenotype. Toxicol Appl Pharmacol 2025; 500:117394. [PMID: 40368219 DOI: 10.1016/j.taap.2025.117394] [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: 01/31/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
Chronic exposure to arsenic, a toxic metalloid frequently found in groundwater and food, represents a significant environmental health risk and has been implicated in the etiology of several cancers, including ovarian cancer. However, the precise pathways through which arsenic exerts its toxic impact on the ovary are not fully understood. This study investigates the impact of chronic arsenic exposure at environmentally relevant concentrations (75 ppb or μg/L) on primary human ovarian surface (OCE1) and fallopian tube (FNE1) cultures derived from the same donor. These heterogeneous cultures provide a unique, human-relevant platform to investigate how chronic arsenic exposure influences distinct cell types within a shared microenvironment. Prolonged arsenic exposure induced significant cytotoxicity and promoted the formation of giant and/or multinucleated cells in both cultures. These cells exhibited phagocytosis-like properties, actively engulfing apoptotic debris. Transcriptomic analyses and pathway enrichment revealed robust activation of pro-inflammatory signaling, notably the canonical NF-κB pathway. This was marked by nuclear translocation of the NF-κB p65 subunit and elevated expression and secretion of pro-inflammatory cytokines, including TNFα, IL-6, and IL-8, driving a sustained inflammatory response. Moreover, arsenic-exposed cells displayed persistent DNA damage, as indicated by increased γ-H2AX foci, accompanied by nuclear structural alterations and elevated expression of cancer stem cell markers, including OCT2, CD133, and ALDH1. These findings suggest that arsenic-induced inflammation and genomic instability converge to promote a tumor-supportive microenvironment, highlighting the potential role of chronic arsenic exposure in ovarian carcinogenesis, particularly in the context of inflammation-driven carcinogenesis.
Collapse
Affiliation(s)
- Cristina M Andrade-Feraud
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America
| | - Arlet M Acanda de la Rocha
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America
| | - Noah E Berlow
- First Ascent Biomedical, Inc., United States of America
| | - Santiago Duque
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America
| | - Alexander Velazco
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America
| | - Diego Castillo
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America
| | - Baylee Holcomb
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America
| | - Ebony R Coats
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America
| | - Yasmin R Ghurani
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America
| | - Catherine M Lucey
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, United States of America
| | - Brandon Pearson
- Environmental and Molecular Toxicology, Oregon State University, OR, United States of America
| | - Tomás R Guilarte
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America
| | - Diana J Azzam
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America.
| |
Collapse
|
2
|
Kim J, Kim HJ, Choi E, Park JJ, Cho M, Choi S, Kim H, Lee JS, Park H. Genome-wide identification of Tegillarca granosa ATP-binding cassette (ABC) transporter family related to arsenic toxicity. Genomics 2025; 117:111024. [PMID: 40015574 DOI: 10.1016/j.ygeno.2025.111024] [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: 09/23/2024] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
Arsenic is a widespread environmental contaminant recognized for its high mobility and potential toxicity. Arsenic levels at Suncheon Bay, one of the primary Tegillarca granosa culturation sites in South Korea, were identified as higher than the habitat's threshold effect level (TEL). After 12 and 48 h of arsenic exposure, a total of 939 and 842 DEGs were identified in the gill and mantle, respectively. Detoxification genes were identified based on DEG analysis, and out of 10 ABCA3 genes in T. granosa, seven ABCA3 genes in total were up- and/or downregulated in two tissues. The metabolic and the cell adhesion molecules KEGG pathways were the most enriched among the commonly identified up- and downregulated genes. The 'metabolic process' gene ontology term was highly enriched with upregulated DEGs. We then identified 74 ATP-binding cassette (ABC) genes in the T. granosa genome, which has seven subfamilies (A to G), with gene expansion found in the ABCC and ABCA subfamilies. Although the precise mechanisms of arsenic-induced gene dysregulation remain unknown, our findings suggest that ABCA3 genes might participate in arsenic active transport and play an important role in arsenic detoxification.
Collapse
Affiliation(s)
- Jinmu Kim
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea; Department of Evolution, Ecology, and Behavior, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States
| | - Hyeon Jin Kim
- Department of Aqualife Medicine, Chonnam National University, Yeosu 59626, Republic of Korea
| | - Eunkyung Choi
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Jung Jun Park
- Aquaculture Industry Research Division, East Sea Fisheries Research Institute, National Institute of Fisheries Science, Gangneung 25435, Republic of Korea
| | - Minjoo Cho
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Soyun Choi
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Hyejin Kim
- Fisheries Seed and Breeding Research Institute, National Institute of Fisheries Science, Haenam 59002, Republic of Korea
| | - Jung Sick Lee
- Department of Aqualife Medicine, Chonnam National University, Yeosu 59626, Republic of Korea.
| | - Hyun Park
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
3
|
González-Alfonso WL, Petrosyan P, Del Razo LM, Sánchez-Peña LC, Tapia-Rodríguez M, Hernández-Muñoz R, Gonsebatt ME. Chronic Exposure to Arsenic and Fluoride Starting at Gestation Alters Liver Mitochondrial Protein Expression and Induces Early Onset of Liver Fibrosis in Male Mouse Offspring. Biol Trace Elem Res 2025; 203:930-943. [PMID: 38676876 PMCID: PMC11750905 DOI: 10.1007/s12011-024-04198-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 04/21/2024] [Indexed: 04/29/2024]
Abstract
The presence of arsenic (As) and fluoride (F-) in drinking water is of concern due to the enormous number of individuals exposed to this condition worldwide. Studies in cultured cells and animal models have shown that As- or F-induced hepatotoxicity is primarily associated with redox disturbance and altered mitochondrial homeostasis. To explore the hepatotoxic effects of chronic combined exposure to As and F- in drinking water, pregnant CD-1 mice were exposed to 2 mg/L As (sodium arsenite) and/or 25 mg/L F- (sodium fluoride). The male offspring continued the exposure treatment up to 30 (P30) or 90 (P90) postnatal days. GSH levels, cysteine synthesis enzyme activities, and cysteine transporter levels were investigated in liver homogenates, as well as the expression of biomarkers of ferroptosis and mitochondrial biogenesis-related proteins. Serum transaminase levels and Hematoxylin-Eosin and Masson trichrome-stained liver tissue slices were examined. Combined exposure at P30 significantly reduced GSH levels and the mitochondrial transcription factor A (TFAM) expression while increasing lipid peroxidation, free Fe 2+, p53 expression, and serum ALT activity. At P90, the upregulation of cysteine uptake and synthesis was associated with a recovery of GSH levels. Nevertheless, the downregulation of TFAM continued and was now associated with a downstream inhibition of the expression of MT-CO2 and reduced levels of mtDNA and fibrotic liver damage. Our experimental approach using human-relevant doses gives evidence of the increased risk for early liver damage associated with elevated levels of As and F- in the diet during intrauterine and postnatal period.
Collapse
Affiliation(s)
- Wendy L González-Alfonso
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, 04510, México
| | - Pavel Petrosyan
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, 04510, México
| | - Luz M Del Razo
- Departamento de Toxicología, Centro de Investigación y Estudios Avanzados, 07360, Mexico City, Mexico
| | - Luz C Sánchez-Peña
- Departamento de Toxicología, Centro de Investigación y Estudios Avanzados, 07360, Mexico City, Mexico
| | - Miguel Tapia-Rodríguez
- Unidad de Microscopia, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Rolando Hernández-Muñoz
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, 04510, México
| | - María E Gonsebatt
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, 04510, México.
| |
Collapse
|
4
|
Chung FFL, Khoueiry R, Sallé A, Cuenin C, Bošković M, Herceg Z. Sodium arsenite-induced DNA methylation alterations exacerbated by p53 knockout in MCF7 cells. Heliyon 2024; 10:e39548. [PMID: 39512451 PMCID: PMC11539298 DOI: 10.1016/j.heliyon.2024.e39548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/24/2024] [Accepted: 10/16/2024] [Indexed: 11/15/2024] Open
Abstract
Epigenetic alterations are ubiquitous across human malignancies. Thus, functional characterization of epigenetic events deregulated by environmental pollutants should enhance our understanding of the mechanisms of carcinogenesis and inform preventive strategies. Recent reports showing the presence of known cancer-driving mutations in normal tissues have sparked debate on the importance of non-mutational stressors potentially acting as cancer promoters. Here, we aimed to test the hypothesis that the presence of mutations in p53, a commonly mutated gene in human malignancies, may influence cellular response to an environmental non-mutagenic agent, potentially involving epigenetic mechanism. We used the CRISPR-Cas9 system to generate knockouts of p53 in MCF7 and T47D breast cancer cell lines and characterized DNA methylome changes by targeted pyrosequencing and methylome-wide Infinium MethylationEPIC BeadChip arrays after exposure to sodium arsenite, a well-established human carcinogen with documented effects on the epigenome. We found that the knockout of p53 alone was associated with extensive alterations in DNA methylation content, with predominant CpG hypermethylation concurrent with global demethylation, as determined by LINE-1 repetitive element pyrosequencing. While exposure to sodium arsenite induced little to no effects in parental cell lines, mutant cells, upon treatment with sodium arsenite, exhibited a markedly altered response in comparison to their wild-type counterparts. We further performed genome regional analyses and found that differentially methylated regions (DMRs) associated with exposure to sodium arsenite map to genes involved in chromatin remodeling and cancer development. Reconstitution of wild-type p53 only partially restored p53-mutant-specific differential methylation states in response to sodium arsenite exposure, which may be due to the insufficient reconstitution of p53 function, or suggestive of a potential exposure-specific epigenetic memory. Together, our results revealed wide-spread epigenetic alterations associated with p53 mutation that influence cellular response to sodium arsenite exposure, which may constate an important epigenetic mechanism by which tumour promoting agents synergize with driver mutations in cancer promotion.
Collapse
Affiliation(s)
- Felicia Fei-Lei Chung
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer (IARC), 25 Av. Tony Garnier, 69007, Lyon, France
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Jalan University, Bandar Sunway, Subang Jaya, 47500, Malaysia
| | - Rita Khoueiry
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer (IARC), 25 Av. Tony Garnier, 69007, Lyon, France
| | - Aurélie Sallé
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer (IARC), 25 Av. Tony Garnier, 69007, Lyon, France
| | - Cyrille Cuenin
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer (IARC), 25 Av. Tony Garnier, 69007, Lyon, France
| | - Maria Bošković
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer (IARC), 25 Av. Tony Garnier, 69007, Lyon, France
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt Am Main, Germany
| | - Zdenko Herceg
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer (IARC), 25 Av. Tony Garnier, 69007, Lyon, France
| |
Collapse
|
5
|
Mukherjee AG, Gopalakrishnan AV. Arsenic-induced prostate cancer: an enigma. Med Oncol 2024; 41:50. [PMID: 38184511 DOI: 10.1007/s12032-023-02266-5] [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: 10/25/2023] [Accepted: 11/21/2023] [Indexed: 01/08/2024]
Abstract
Arsenic exhibits varying degrees of toxicity depending on its many chemical forms. The carcinogenic properties of arsenic have already been established. However, the precise processes underlying the development of diseases following acute or chronic exposure to arsenic remain poorly known. Most of the existing investigation has focused on studying the occurrence of cancer following significant exposure to elevated levels of arsenic. Nevertheless, multiple investigations have documented diverse health consequences from prolonged exposure to low levels of arsenic. Inorganic arsenic commonly causes lung, bladder, and skin cancer. Some investigations have shown an association between arsenic in drinking water and prostate cancer, but few investigations have focused on exploring this connection. There is currently a lack of relevant animal models demonstrating a clear link between inorganic arsenic exposure and the development of prostate cancer. Nevertheless, studies using cellular model systems have demonstrated that arsenic can potentially promote the malignant transformation of human prostate epithelial cells in vitro. The administration of elevated levels of arsenic has been demonstrated to elicit cell death in instances of acute experimental exposure. Conversely, in cases of chronic exposure, arsenic prompts cellular proliferation and sustains cellular viability, thereby circumventing the constraints imposed by telomere shortening and apoptosis. Furthermore, cells consistently exposed to the stimulus exhibit an augmented ability to invade surrounding tissues and an enhanced potential to form tumors. This review aims to portray mechanistic insights into arsenic-induced prostate cancer.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
6
|
EFSA Panel on Contaminants in the Food Chain (CONTAM), Schrenk D, Bignami M, Bodin L, Chipman JK, del Mazo J, Grasl‐Kraupp B, Hogstrand C, Hoogenboom L(R, Leblanc J, Nebbia CS, Nielsen E, Ntzani E, Petersen A, Sand S, Vleminckx C, Wallace H, Barregård L, Benford D, Broberg K, Dogliotti E, Fletcher T, Rylander L, Abrahantes JC, Gómez Ruiz JÁ, Steinkellner H, Tauriainen T, Schwerdtle T. Update of the risk assessment of inorganic arsenic in food. EFSA J 2024; 22:e8488. [PMID: 38239496 PMCID: PMC10794945 DOI: 10.2903/j.efsa.2024.8488] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2024] Open
Abstract
The European Commission asked EFSA to update its 2009 risk assessment on arsenic in food carrying out a hazard assessment of inorganic arsenic (iAs) and using the revised exposure assessment issued by EFSA in 2021. Epidemiological studies show that the chronic intake of iAs via diet and/or drinking water is associated with increased risk of several adverse outcomes including cancers of the skin, bladder and lung. The CONTAM Panel used the benchmark dose lower confidence limit based on a benchmark response (BMR) of 5% (relative increase of the background incidence after adjustment for confounders, BMDL05) of 0.06 μg iAs/kg bw per day obtained from a study on skin cancer as a Reference Point (RP). Inorganic As is a genotoxic carcinogen with additional epigenetic effects and the CONTAM Panel applied a margin of exposure (MOE) approach for the risk characterisation. In adults, the MOEs are low (range between 2 and 0.4 for mean consumers and between 0.9 and 0.2 at the 95th percentile exposure, respectively) and as such raise a health concern despite the uncertainties.
Collapse
|
7
|
Wysocki R, Rodrigues JI, Litwin I, Tamás MJ. Mechanisms of genotoxicity and proteotoxicity induced by the metalloids arsenic and antimony. Cell Mol Life Sci 2023; 80:342. [PMID: 37904059 PMCID: PMC10616229 DOI: 10.1007/s00018-023-04992-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/12/2023] [Accepted: 09/29/2023] [Indexed: 11/01/2023]
Abstract
Arsenic and antimony are metalloids with profound effects on biological systems and human health. Both elements are toxic to cells and organisms, and exposure is associated with several pathological conditions including cancer and neurodegenerative disorders. At the same time, arsenic- and antimony-containing compounds are used in the treatment of multiple diseases. Although these metalloids can both cause and cure disease, their modes of molecular action are incompletely understood. The past decades have seen major advances in our understanding of arsenic and antimony toxicity, emphasizing genotoxicity and proteotoxicity as key contributors to pathogenesis. In this review, we highlight mechanisms by which arsenic and antimony cause toxicity, focusing on their genotoxic and proteotoxic effects. The mechanisms used by cells to maintain proteostasis during metalloid exposure are also described. Furthermore, we address how metalloid-induced proteotoxicity may promote neurodegenerative disease and how genotoxicity and proteotoxicity may be interrelated and together contribute to proteinopathies. A deeper understanding of cellular toxicity and response mechanisms and their links to pathogenesis may promote the development of strategies for both disease prevention and treatment.
Collapse
Affiliation(s)
- Robert Wysocki
- Department of Genetics and Cell Physiology, Faculty of Biological Sciences, University of Wroclaw, 50-328, Wroclaw, Poland.
| | - Joana I Rodrigues
- Department of Chemistry and Molecular Biology, University of Gothenburg, Box 462, 405 30, Göteborg, Sweden
| | - Ireneusz Litwin
- Academic Excellence Hub - Research Centre for DNA Repair and Replication, Faculty of Biological Sciences, University of Wroclaw, 50-328, Wroclaw, Poland
| | - Markus J Tamás
- Department of Chemistry and Molecular Biology, University of Gothenburg, Box 462, 405 30, Göteborg, Sweden.
| |
Collapse
|
8
|
Yang X, Weber AA, Mennillo E, Secrest P, Chang M, Wong S, Le S, Liu J, Benner CW, Karin M, Gordts PL, Tukey RH, Chen S. Effects of Early Life Oral Arsenic Exposure on Intestinal Tract Development and Lipid Homeostasis in Neonatal Mice: Implications for NAFLD Development. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:97001. [PMID: 37668303 PMCID: PMC10478510 DOI: 10.1289/ehp12381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 05/01/2023] [Accepted: 07/11/2023] [Indexed: 09/06/2023]
Abstract
BACKGROUND Newborns can be exposed to inorganic arsenic (iAs) through contaminated drinking water, formula, and other infant foods. Epidemiological studies have demonstrated a positive association between urinary iAs levels and the risk of developing nonalcoholic fatty liver disease (NAFLD) among U.S. adolescents and adults. OBJECTIVES The present study examined how oral iAs administration to neonatal mice impacts the intestinal tract, which acts as an early mediator for NAFLD. METHODS Neonatal mice were treated with a single dose of iAs via oral gavage. Effects on the small intestine were determined by histological examination, RNA sequencing, and biochemical analysis. Serum lipid profiling was analyzed by fast protein liquid chromatography (FPLC), and hepatosteatosis was characterized histologically and biochemically. Liver X receptor-alpha (LXR α ) knockout (L x r α - / - ) mice and liver-specific activating transcription factor 4 (ATF4)-deficient (A t f 4 Δ H e p ) mice were used to define their roles in iAs-induced effects during the neonatal stage. RESULTS Neonatal mice exposed to iAs via oral gavage exhibited accumulation of dietary fat in enterocytes, with higher levels of enterocyte triglycerides and free fatty acids. These mice also showed accelerated enterocyte maturation and a longer small intestine. This was accompanied by higher levels of liver-derived very low-density lipoprotein and low-density lipoprotein triglycerides, and a lower level of high-density lipoprotein cholesterol in the serum. Mice exposed during the neonatal period to oral iAs also developed hepatosteatosis. Compared with the control group, iAs-induced fat accumulation in enterocytes became more significant in neonatal L x r α - / - mice, accompanied by accelerated intestinal growth, hypertriglyceridemia, and hepatosteatosis. In contrast, regardless of enterocyte fat accumulation, hepatosteatosis was largely reduced in iAs-treated neonatal A t f 4 Δ H e p mice. CONCLUSION Exposure to iAs in neonatal mice resulted in excessive accumulation of fat in enterocytes, disrupting lipid homeostasis in the serum and liver, revealing the importance of the gut-liver axis and endoplasmic reticulum stress in mediating iAs-induced NAFLD at an early age. https://doi.org/10.1289/EHP12381.
Collapse
Affiliation(s)
- Xiaojing Yang
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego (UC San Diego), La Jolla, California, USA
| | - André A. Weber
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego (UC San Diego), La Jolla, California, USA
| | - Elvira Mennillo
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego (UC San Diego), La Jolla, California, USA
| | - Patrick Secrest
- Department of Medicine, Division of Endocrinology and Metabolism, UC San Diego, La Jolla, California, USA
| | - Max Chang
- Department of Medicine, School of Medicine, UC San Diego, La Jolla, California, USA
| | - Samantha Wong
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego (UC San Diego), La Jolla, California, USA
| | - Sabrina Le
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego (UC San Diego), La Jolla, California, USA
| | - Junlai Liu
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, UC San Diego, La Jolla, California, USA
| | | | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, UC San Diego, La Jolla, California, USA
| | - Philip L.S.M. Gordts
- Department of Medicine, Division of Endocrinology and Metabolism, UC San Diego, La Jolla, California, USA
| | - Robert H. Tukey
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego (UC San Diego), La Jolla, California, USA
| | - Shujuan Chen
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego (UC San Diego), La Jolla, California, USA
| |
Collapse
|
9
|
Sun J, Wu L, Wu M, Liu Q, Cao H. Non-coding RNA therapeutics: Towards a new candidate for arsenic-induced liver disease. Chem Biol Interact 2023; 382:110626. [PMID: 37442288 DOI: 10.1016/j.cbi.2023.110626] [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: 03/26/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 07/15/2023]
Abstract
Arsenic, a metalloid toxicant, has caused serious environmental pollution and is presently a global health issue. Long-term exposure to arsenic causes diverse organ and system dysfunctions, including liver disease. Arsenic-induced liver disease comprises a spectrum of liver pathologies, ranging from hepatocyte damage, steatosis, fibrosis, to hepatocellular carcinoma. Various mechanisms, including an imbalance in redox reactions, mitochondrial dysfunction and epigenetic changes, participate in the pathogenesis of arsenic-induced liver disease. Altered epigenetic processes involved in its initiation and progression. Dysregulated modulations of non-coding RNAs (ncRNAs), including miRNAs, lncRNAs and circRNAs, exert regulating effects on these processes. Here, we have reviewed the underlying pathogenic mechanisms that lead to progressive arsenic-induced liver disease, and we provide a discussion focusing on the effects of ncRNAs on dysfunctions in intercellular communication and on the activation of hepatic stellate cells and malignant transformation of hepatocytes. Further, we have discussed the roles of ncRNAs in intercellular communication via extracellular vesicles and cytokines, and have provided a perspective for the application of ncRNAs as biomarkers in the early diagnosis and evaluation of the pathogenesis of arsenic-induced liver disease. Further investigations of ncRNAs will help us to understand the nature of arsenic-induced liver disease and to identify biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Jing Sun
- Department of Nutrition, Functional Food Clinical Evaluation Center, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China
| | - Lu Wu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Meng Wu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Qizhan Liu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China.
| | - Hong Cao
- Department of Nutrition, Functional Food Clinical Evaluation Center, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China.
| |
Collapse
|
10
|
Kumar N, Mathur A, Bunker SK, John PJ. Cell Cycle dysregulation on prenatal and postnatal Arsenic exposure in skin of Wistar rat neonates. Xenobiotica 2023:1-15. [PMID: 37449383 DOI: 10.1080/00498254.2023.2237102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
This study explores the effects of prenatal and postnatal (until weaning period) arsenic exposure given via pregnant females on Wistar rat neonates. Pregnant female rats were divided in four groups - control, low dose, moderate dose and high dose groups of sodium arsenite exposure during gestation and weaning period. Half of the neonates were sacrificed at day 1 of birth and other half at day 21 of birth. Cell cycle analysis in epidermal keratinocytes using flowcytometer revealed that there was a consistent increase in number of cells in G2/M phase from 0.04% in control group to 0.88%, 1.59% and 2.77% in low, moderate and high dose groups respectively for neonates sacrificed at day-1. Whereas, the increase in number of cells with increasing doses in G2/M phase of neonates sacrificed at day-21 was from 3.44% to 5.1%, 6.82%, and 9.17%. At postnatal day 21, mRNA expression of Cyclin A and B1, p53, Caspases 3, 7 and 9, and Bax were found to be up-regulated. Whereas that of Cyclin E, CDK 1 and 2 and Bcl2 were down regulated consistently in skin tissues of arsenic exposed groups.
Collapse
Affiliation(s)
- Navneet Kumar
- Centre for advanced studies, Department of Zoology, University of Rajasthan, Jaipur, India - 302004
| | - Astha Mathur
- Centre for advanced studies, Department of Zoology, University of Rajasthan, Jaipur, India - 302004
| | - Suresh Kumar Bunker
- Centre for advanced studies, Department of Zoology, University of Rajasthan, Jaipur, India - 302004
| | - Placheril J John
- Centre for advanced studies, Department of Zoology, University of Rajasthan, Jaipur, India - 302004
| |
Collapse
|
11
|
Chen X, Wu R, Wu H, Hu Y, Wang H, Fu J, Pi J, Xu Y. Integrated miRNA-mRNA analysis reveals the dysregulation of lipid metabolism in mouse liver induced by developmental arsenic exposure. JOURNAL OF HAZARDOUS MATERIALS 2023; 445:130459. [PMID: 36463740 DOI: 10.1016/j.jhazmat.2022.130459] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 06/17/2023]
Abstract
Developmental arsenic exposure leads to increased susceptibility to liver diseases including nonalcoholic fatty liver diseases, but the mechanism is incompletely understood. In this study, C57BL/6J mice were used to establish a lifetime arsenic exposure model covering developmental stage. We found that arsenic-exposed offspring in later life showed hepatic lipid deposition and increased triglyceride content. Despite no significant hepatic pathological changes in the offspring at weaning, 86 miRNAs and 136 mRNAs were differentially expressed according to miRNA array and mRNA sequencing. The differentially expressed genes (DEGs) were crossed with the target genes predicted by differentially expressed miRNAs (DEMs), and 47 differentially expressed target genes (DETGs) were obtained. Functional annotation suggested that lipid metabolism related pathways were significantly enriched. The pivotal regulator in the four major pathways to maintain liver lipid homeostasis were further determined, with significant alterations found in FABP5, SREBP1, ACOX1 and EHHADH. Of note, miRNA-mRNA integration analysis revealed that miR-7118-5p, miR-7050-5p, miR-27a/b-3p, and miR-103-3p acted as key regulators of fatty acid metabolism genes. Taken together, miRNA-mRNA integration analysis indicates that the lipid metabolism pathway in the liver of weaned mice was dysregulated by developmental arsenic exposure, which may contribute to the development of NAFLD in later life.
Collapse
Affiliation(s)
- Xin Chen
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Ruirui Wu
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Hengchao Wu
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Yuxin Hu
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Huihui Wang
- School of Public Health, China Medical University, Shenyang, Liaoning, China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang, Liaoning, China
| | - Jingqi Fu
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Jingbo Pi
- School of Public Health, China Medical University, Shenyang, Liaoning, China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang, Liaoning, China
| | - Yuanyuan Xu
- School of Public Health, China Medical University, Shenyang, Liaoning, China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
12
|
Wu H, Wu R, Chen X, Geng H, Hu Y, Gao L, Fu J, Pi J, Xu Y. Developmental arsenic exposure induces dysbiosis of gut microbiota and disruption of plasma metabolites in mice. Toxicol Appl Pharmacol 2022; 450:116174. [PMID: 35878798 DOI: 10.1016/j.taap.2022.116174] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/19/2022] [Accepted: 07/19/2022] [Indexed: 10/16/2022]
Abstract
Arsenic is a notorious environmental pollutant. Of note, developmental arsenic exposure has been found to increase the risk of developing a variety of ailments later in life, but the underlying mechanism is not well understood. Many elements of host health have been connected to the gut microbiota. It is still unclear whether and how developmental arsenic exposure affects the gut microbiota. In the present study, we found that developmental arsenic exposure changed intestinal morphology and increased intestinal permeability and inflammation in mouse pups at weaning. These alterations were accompanied by a significant change in gut microbiota, as evidenced by considerably reduced gut microbial richness and diversity. In developmentally arsenic-exposed pups, the relative abundance of Muribaculaceae was significantly decreased, while the relative abundance of Akkermansia and Bacteroides was significantly enhanced at the genus level. Metabolome and pathway enrichment analyses indicated that amino acid and purine metabolism was promoted, while glycerophospholipid metabolism was inhibited. Interestingly, the relative abundance of Muribaculaceae and Akkermansia showed a strong correlation with most plasma metabolites significantly altered by developmental arsenic exposure. These data indicate that gut microbiota dysbiosis may be a critical link between developmental arsenic exposure and metabolic disorders and shed light on the mechanisms underlying increased susceptibility to diseases due to developmental arsenic exposure.
Collapse
Affiliation(s)
- Hengchao Wu
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Ruirui Wu
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Xin Chen
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Huamin Geng
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Yuxin Hu
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Lanyue Gao
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Jingqi Fu
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Jingbo Pi
- School of Public Health, China Medical University, Shenyang, Liaoning, China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang, Liaoning, China
| | - Yuanyuan Xu
- School of Public Health, China Medical University, Shenyang, Liaoning, China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
13
|
Wu R, Chen X, Wu H, Hu Y, Wang G, Wang H, Yang B, Fu J, Gao Y, Pi J, Xu Y. Nrf2 activation contributes to hepatic tumor-augmenting effects of developmental arsenic exposure. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 837:155685. [PMID: 35523338 DOI: 10.1016/j.scitotenv.2022.155685] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 06/14/2023]
Abstract
Developmental arsenic exposure increases cancer risk in later life with the mechanism elusive. Oxidative stress is a dominant determinant in arsenic toxicity. However, the role of Nrf2, a key regulator in antioxidative response, in tumor-augmenting effects by developmental arsenic exposure is unclear. In the present study, wild-type C57BL/6J and Nrf2-konckout (Nrf2-KO) were developmentally exposed to inorganic arsenic via drinking water. For hepatic tumorigenesis analysis, mice were intraperitoneally injected with diethylnitrosamine (DEN) at two weeks of age. Developmental arsenic exposure aggravated tumor multiplicity and burden, and expression of PCNA and AFP in hepatic tumors induced by DEN. Nrf2 activation as indicated by over-expression of Nrf2 and its downstream genes, including Gss, Gsr, p62, Gclc and Gclm, was found in liver tumors, as well as in the livers in developmentally arsenic-exposed pups at weaning. Notably, Nrf2 deficiency attenuated tumor-augmenting effects and over-expression of Nrf2 downstream genes due to developmental arsenic exposure. Furthermore, the levels of urinary DEN metabolite (acetaldehyde) and hepatic DNA damage markers (O6-ethyl-2-deoxyguanosine adducts and γ-histone H2AX) after DEN treatment were elevated by Nrf2 agonist, 2-Cyano-3,12-dioxooleana-1,9-dien-28-imidazolide. Collectively, our data suggest that augmentation of DEN-induced hepatic tumorigenesis by developmental arsenic exposure is dependent on Nrf2 activation, which may be related to the role of Nrf2 in DEN metabolic activation. Our findings reveal, at least in part, the mechanism underlying increased susceptibility to developing cancer due to developmental arsenic exposure.
Collapse
Affiliation(s)
- Ruirui Wu
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Xin Chen
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Hengchao Wu
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Yuxin Hu
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Gang Wang
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Huihui Wang
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Bei Yang
- College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Jingqi Fu
- School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Yanhui Gao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jingbo Pi
- School of Public Health, China Medical University, Shenyang, Liaoning, China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang, Liaoning, China
| | - Yuanyuan Xu
- School of Public Health, China Medical University, Shenyang, Liaoning, China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
14
|
Raeeszadeh M, Gravandi H, Akbari A. Determination of some heavy metals levels in the meat of animal species (sheep, beef, turkey, and ostrich) and carcinogenic health risk assessment in Kurdistan province in the west of Iran. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:62248-62258. [PMID: 35277826 DOI: 10.1007/s11356-022-19589-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 03/02/2022] [Indexed: 06/14/2023]
Abstract
Heavy metals accumulation in food products as a result of industrialization is one of the main potential threats to public health. This study was aimed to evaluate the concentrations of heavy metals in the meat of some prevalent farm animal species including sheep, beef, turkeys, and ostriches in Sanandaj (one of the strategic cities in Kurdistan province, Iran). In this study, the contents of some heavy metals (selenium, lead, cadmium, arsenic, cobalt, zinc, nickel, copper, and chromium) were assessed in 170 meat samples collected from meat distribution centers in Sanandaj, Kurdistan province. The ICP-MS method was used to assess the levels of these elements in the meat of beef, sheep, turkey, and ostrich as the main consumed meats in this region. Statistical analysis was performed by ANOVA and one-sample t-test, and the correlation coefficient of the concentrations of metals was also calculated. The results showed that there were no significant differences in the average contents of selenium, nickel, cobalt, and chromium among various meats (P > 0.05). However, the amounts of lead, cadmium, arsenic, zinc, copper, chromium, and nickel were meaningfully different to maximum permissible limits (MPL) (P < 0.05). In addition, a significant negative correlation was found between arsenic and selenium in meats. Except for cobalt, the concentrations of other metals were higher than MPL in the evaluated meats. Despite the target hazard quotient (THQ) for cadmium in sheep and beef meats being higher than their MPL (> 1), its target risk of cancer (TR) was acceptable. It can be concluded that monitoring and elimination of heavy metals pollution are essential because of their high accumulation in the meat of main farm animal species in Sanandaj, Kurdistan province.
Collapse
Affiliation(s)
- Mahdieh Raeeszadeh
- Department of Basic Sciences, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran.
| | - Hamed Gravandi
- Graduate of Faculty of Veterinary Sciences, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| | - Abolfazl Akbari
- Department of Physiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
15
|
Kim C, Chen J, Ceresa BP. Chronic arsenic increases cell migration in BEAS-2B cells by increasing cell speed, cell persistence, and cell protrusion length. Exp Cell Res 2021; 408:112852. [PMID: 34599931 DOI: 10.1016/j.yexcr.2021.112852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 10/20/2022]
Abstract
There is a strong association between arsenic exposure and lung cancer development, however, the mechanism by which arsenic exposure leads to carcinogenesis is not clear. In our previous study, we observed that when BEAS-2B cells are chronically exposed to arsenic, there is an increase in secreted TGFα, as well as an increase in EGFR expression and activity. Further, these changes were broadly accompanied with an increase in cell migration. The overarching goal of this study was to acquire finer resolution of the arsenic-dependent changes in cell migration, as well as to understand the role of increased EGFR expression and activity levels in the underlying mechanisms of cell migration. To do this, we used a combination of biochemical and single cell assays, and observed chronic arsenic treatment enhancing cell migration by increasing cell speed, cell persistence and cell protrusion length. All three parameters were further increased by the addition of TGFα, indicating EGFR activity is sufficient to enhance those aspects of cell migration. In contrast, EGFR activity was necessary for the increase in cell speed, as it was reversed with an EGFR inhibitor, AG1478, but was not necessary to enhance persistence and protrusion length. From these data, we were able to isolate both EGFR-dependent and -independent features of cell migration that were enhanced by chronic arsenic exposure.
Collapse
Affiliation(s)
- Christine Kim
- Department of Pharmacology and Toxicology, University of Louisville, USA
| | - Joseph Chen
- Department of Pharmacology and Toxicology, University of Louisville, USA; Department of Bioengineering, University of Louisville, USA
| | - Brian P Ceresa
- Department of Pharmacology and Toxicology, University of Louisville, USA.
| |
Collapse
|
16
|
Wang Z. Mechanisms of the synergistic lung tumorigenic effect of arsenic and benzo(a)pyrene combined- exposure. Semin Cancer Biol 2021; 76:156-162. [PMID: 33971262 PMCID: PMC9000133 DOI: 10.1016/j.semcancer.2021.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/01/2021] [Indexed: 12/20/2022]
Abstract
Humans are often exposed to mixtures of environmental pollutants especially environmental chemical carcinogens, representing a significant environmental health issue. However, our understanding on the carcinogenic effects and mechanisms of environmental carcinogen mixture exposures is limited and mostly relies on the findings from studying individual chemical carcinogens. Both arsenic and benzo(a)pyrene (BaP) are among the most common environmental carcinogens causing lung cancer and other types of cancer in humans. Millions of people are exposed to arsenic via consuming arsenic-contaminated drinking water and even more people are exposed to BaP via cigarette smoking and consuming BaP-contaminated food. Thus arsenic and BaP combined-exposure in humans is common. Previous epidemiology studies indicated that arsenic-exposed people who were cigarette smokers had significantly higher lung cancer risk than those who were non-smokers. Since BaP is one of the major carcinogens in cigarette smoke, it has been speculated that arsenic and BaP combined-exposure may play important roles in the increased lung cancer risk observed in arsenic-exposed cigarette smokers. In this review, we summarize important findings and inconsistencies about the co-carcinogenic effects and underlying mechanisms of arsenic and BaP combined-exposure and propose new areas for future studies. A clear understanding on the mechanism of co-carcinogenic effects of arsenic and BaP combined exposure may identify novel targets to more efficiently treat and prevent lung cancer resulting from arsenic and BaP combined-exposure.
Collapse
Affiliation(s)
- Zhishan Wang
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44109, USA.
| |
Collapse
|
17
|
Efremenko A, Balbuena P, Clewell RA, Black M, Pluta L, Andersen ME, Gentry PR, Yager JW, Clewell HJ. Time-dependent genomic response in primary human uroepithelial cells exposed to arsenite for up to 60 days. Toxicology 2021; 461:152893. [PMID: 34425169 DOI: 10.1016/j.tox.2021.152893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/02/2021] [Accepted: 08/05/2021] [Indexed: 11/16/2022]
Abstract
Evidence from both in vivo and in vitro studies suggests that gene expression changes from long-term exposure to arsenite evolve markedly over time, including reversals in the direction of expression change in key regulatory genes. In this study, human uroepithelial cells from the ureter segments of 4 kidney-donors were continuously treated in culture with arsenite at concentrations of 0.1 or 1 μM for 60 days. Gene expression at 10, 20, 30, 40, and 60 days was determined using Affymetrix human genome microarrays and signal pathway analysis was performed using GeneGo Metacore. Arsenic treated cells continued to proliferate for the full 60-day period, whereas untreated cells ceased proliferating after approximately 30 days. A peak in the number of gene changes in the treated cells compared to untreated controls was observed between 30 and 40 days of exposure, with substantially fewer changes at 10 and 60 days, suggesting remodeling of the cells over time. Consistent with this possibility, the direction of expression change for a number of key genes was reversed between 20 and 30 days, including CFOS and MDM2. While the progression of gene changes was different for each subject, a common pattern was observed in arsenic treated cells over time, with early upregulation of oxidative stress responses (HMOX1, NQ01, TXN, TXNRD1) and down-regulation of immune/inflammatory responses (IKKα). At around 30 days, there was a transition to increased inflammatory and proliferative signaling (AKT, CFOS), evidence of epithelial-to-mesenchymal transition (EMT), and alterations in DNA damage responses (MDM2, ATM). A common element in the changing response of cells to arsenite over time appears to involve up-regulation of MDM2 by inflammatory signaling (through AP-1 and NF-κB), leading to inhibition of P53 function.
Collapse
Affiliation(s)
- Alina Efremenko
- The Hamner Institutes for Health Sciences, RTP, NC, United States
| | | | | | - Michael Black
- The Hamner Institutes for Health Sciences, RTP, NC, United States
| | - Linda Pluta
- The Hamner Institutes for Health Sciences, RTP, NC, United States
| | | | | | - Janice W Yager
- Ramboll US Corporation, Emeryville, CA, United States(1)
| | - Harvey J Clewell
- The Hamner Institutes for Health Sciences, RTP, NC, United States.
| |
Collapse
|
18
|
Kim C, Ceresa BP. Using In Vitro Models to Dissect the Molecular Effects of Arsenic Exposure in Skin and Lung Cell Lines. APPLIED IN VITRO TOXICOLOGY 2021; 7:71-88. [DOI: 10.1089/aivt.2020.0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Affiliation(s)
- Christine Kim
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| | - Brian P. Ceresa
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
19
|
Whitlock B. Telomere Length and Arsenic: Improving Animal Models of Toxicity by Choosing Mice With Shorter Telomeres. Int J Toxicol 2021; 40:211-217. [PMID: 34008434 DOI: 10.1177/10915818211009844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Arsenic is both a chemotherapeutic drug and an environmental toxicant that affects hundreds of millions of people each year. Arsenic exposure in drinking water has been called the worst poisoning in human history. How arsenic is handled in the body is frequently studied using rodent models to investigate how arsenic both causes and treats disease. These models, used in a variety of arsenic-related testing, from tumor formation to drug toxicity monitoring, have virtually always been developed from animals with telomeres that are unnaturally long, likely because of accidental artificial selective pressures. Mice that have been bred in captivity in laboratory conditions, often for over 100 years, are the standard in creating animal models for this research. Using these mice introduces challenges to any work that can be affected by the length of telomeres and the related capacities for tissue repair and cancer resistance. However, arsenic research is particularly susceptible to the misuse of such animal models due to the multiple and various interactions between arsenic and telomeres. Researchers in the field commonly find mouse models and humans behaving very differently upon exposure to acute and chronic arsenic, including drug therapies which seem safe in mice but are toxic in humans. Here, some complexities and apparent contradictions of the arsenic carcinogenicity and toxicity research are reconciled by an explanatory model that involves telomere length explained by the evolutionary pressures in laboratory mice. A low-risk hypothesis is proposed which has the power to determine whether researchers can easily develop more powerful and accurate mouse models by simply avoiding mouse lineages that are very old and have strangely long telomeres. Swapping in newer mouse lineages for the older, long-telomere mice may vastly improve our ability to research arsenic toxicity with virtually no increase in cost or difficulty of research.
Collapse
Affiliation(s)
- Brayden Whitlock
- University of Alberta Health Accelerator, Edmonton, Alberta, Canada.,Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
20
|
Inesta-Vaquera F, Navasumrit P, Henderson CJ, Frangova TG, Honda T, Dinkova-Kostova AT, Ruchirawat M, Wolf CR. Application of the in vivo oxidative stress reporter Hmox1 as mechanistic biomarker of arsenic toxicity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 270:116053. [PMID: 33213951 DOI: 10.1016/j.envpol.2020.116053] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/02/2020] [Accepted: 11/06/2020] [Indexed: 05/26/2023]
Abstract
Inorganic arsenic (iAs) is a naturally occurring metalloid present in drinking water and polluted air exposing millions of people globally. Epidemiological studies have linked iAs exposure to the development of numerous diseases including cognitive impairment, cardiovascular failure and cancer. Despite intense research, an effective therapy for chronic arsenicosis has yet to be developed. Laboratory studies have been of great benefit in establishing the pathways involved in iAs toxicity and providing insights into its mechanism of action. However, the in vivo analysis of arsenic toxicity mechanisms has been difficult by the lack of reliable in vivo biomarkers of iAs's effects. To address this issue we have applied the use of our recently developed stress reporter models to study iAs toxicity. The reporter mice Hmox1 (oxidative stress/inflammation; HOTT) and p21 (DNA damage) were exposed to iAs at acute and chronic, environmentally relevant, doses. We observed induction of the oxidative stress reporters in several cell types and tissues, which was largely dependent on the activation of transcription factor NRF2. We propose that our HOTT reporter model can be used as a surrogate biomarker of iAs-induced oxidative stress, and it constitutes a first-in-class platform to develop treatments aimed to counteract the role of oxidative stress in arsenicosis. Indeed, in a proof of concept experiment, the HOTT reporter mice were able to predict the therapeutic utility of the antioxidant N-acetyl cysteine in the prevention of iAs associated toxicity.
Collapse
Affiliation(s)
- Francisco Inesta-Vaquera
- Department of Systems Medicine. School of Medicine. University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK.
| | - Panida Navasumrit
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok, 10210, Thailand
| | - Colin J Henderson
- Department of Systems Medicine. School of Medicine. University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK
| | - Tanya G Frangova
- Department of Systems Medicine. School of Medicine. University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK
| | - Tadashi Honda
- Department of Chemistry and Institute of Chemical Biology & Drug Discovery, Stony Brook University, Stony Brook, NY, 11794-3400, USA
| | - Albena T Dinkova-Kostova
- Department of Molecular Medicine. School of Medicine. University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK
| | - Mathuros Ruchirawat
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok, 10210, Thailand
| | - C Roland Wolf
- Department of Systems Medicine. School of Medicine. University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK
| |
Collapse
|
21
|
Xiao T, Zou Z, Xue J, Syed BM, Sun J, Dai X, Shi M, Li J, Wei S, Tang H, Zhang A, Liu Q. LncRNA H19-mediated M2 polarization of macrophages promotes myofibroblast differentiation in pulmonary fibrosis induced by arsenic exposure. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 268:115810. [PMID: 33162208 DOI: 10.1016/j.envpol.2020.115810] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/02/2020] [Accepted: 10/09/2020] [Indexed: 06/11/2023]
Abstract
Arsenic is a potent toxicant, and long-term exposure to inorganic arsenic causes lung damage. M2 macrophages play an important role in the pathogenesis of pulmonary fibrosis. However, the potential connections between arsenic and M2 macrophages in the development of pulmonary fibrosis are elusive. C57BL/6 mice were fed with drinking water containing 0, 10 and 20 ppm arsenite for 12 months. We have found that, in lung tissues of mice, arsenite, a biologically active form of arsenic, elevated H19, c-Myc, and Arg1; decreased let-7a; and caused pulmonary fibrosis. For THP-1 macrophages (THP-M) and bone-marrow-derived macrophages (BMDMs), 8 μM arsenite increased H19, c-Myc, and Arg1; decreased let-7a; and induced M2 polarization of macrophages, which caused secretion of the fibrogenic cytokine, TGF-β1. Down-regulation of H19 or up-regulation of let-7a reversed the arsenite-induced M2 polarization of macrophages. Arsenite-treated THP-M and BMDMs co-cultured with MRC-5 cells or primary lung fibroblasts (PLFs) elevated levels of p-SMAD2/3, SMAD4, α-SMA, and collagen I in lung fibroblasts and resulted in the activation of lung fibroblasts. Knockout of H19 or up-regulation of let-7a in macrophages reversed the effects. The results indicated that H19 functioned as an miRNA sponge for let-7a, which was involved in arsenite-induced M2 polarization of macrophages and induced the myofibroblast differentiation phenotype by regulation of c-Myc. In the sera of arseniasis patients, levels of hydroxyproline and H19 were higher, and levels of let-7a were lower than levels in the controls. These observations elucidate a possible mechanism for arsenic exposure-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Tian Xiao
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Zhonglan Zou
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, People's Republic of China
| | - Junchao Xue
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Binafsha Manzoor Syed
- Medical Research Centre, Liaquat University of Medical & Health Sciences, Jamshoro, 76090, Sindh, Pakistan
| | - Jing Sun
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Xiangyu Dai
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Ming Shi
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, 523808, Guangdong, People's Republic of China
| | - Junjie Li
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Shaofeng Wei
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, People's Republic of China
| | - Huanwen Tang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, 523808, Guangdong, People's Republic of China
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, People's Republic of China
| | - Qizhan Liu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China.
| |
Collapse
|
22
|
Kim C, States JC, Ceresa BP. Chronic and acute arsenic exposure enhance EGFR expression via distinct molecular mechanisms. Toxicol In Vitro 2020; 67:104925. [PMID: 32599262 DOI: 10.1016/j.tiv.2020.104925] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/02/2020] [Accepted: 06/22/2020] [Indexed: 01/06/2023]
Abstract
The impacts of acute arsenic exposure (i.e. vomiting, diarrhea, and renal failure) are distinct from those brought about by sustained, low level exposure from environmental sources or drinking of contaminated well water. Chronic arsenic exposure is a risk factor for the development of pulmonary diseases, including lung cancer. How arsenic exposure leads to pulmonary disease is not fully understood. Both acute versus chronic arsenic exposure increase EGFR expression, but do so via distinct molecular mechanisms. BEAS-2B cells were exposed to either acute sodium arsenite (5 μM for 24 h) or chronic sodium arsenite (100 nM for 24 weeks). Cells treated with acute arsenic exhibited a decrease in viability, changes in morphology, and increased mRNA level of BTC. In contrast, during 24 weeks of arsenic exposure, the cells had increased EGFR expression and activity, and increased mRNA and protein levels of TGFα. Further, chronic arsenic treatment caused an increase in cell migration in the absence of exogenous ligand. Elevated TGFα and EGFR expression are features of many non-small cell lung cancers. We propose that lung epithelial cells chronically exposed to low level arsenic increases EGFR signaling via TGFα production to enhance ligand-independent cell migration.
Collapse
Affiliation(s)
- Christine Kim
- Department of Pharmacology and Toxicology, University of Louisville, USA
| | | | - Brian P Ceresa
- Department of Pharmacology and Toxicology, University of Louisville, USA.
| |
Collapse
|
23
|
Rodriguez KF, Mellouk N, Ungewitter EK, Nicol B, Liu C, Brown PR, Willson CJ, Yao HHC. In utero exposure to arsenite contributes to metabolic and reproductive dysfunction in male offspring of CD-1 mice. Reprod Toxicol 2020; 95:95-103. [PMID: 32428649 DOI: 10.1016/j.reprotox.2020.05.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 01/13/2023]
Abstract
In utero exposure to arsenite (iAs) is known to increase disease risks later in life. We investigated the effect of in utero exposure to iAs in the drinking water on metabolic and reproductive parameters in male mouse offspring at postnatal and adult stages. Pregnant CD-1 mice were exposed to iAs (as sodium arsenite) in the drinking water at 0 (control), 10 ppb (EPA standard for drinking water), and 42.5 ppm (tumor-inducing dose in mice) from embryonic day (E) 10-18. At birth, pups were fostered to unexposed females. Male offspring exposed to 10 ppb in utero exhibited increase in body weight at birth when compared to controls. Male offspring exposed to 42.5 ppm in utero showed a tendency for increased body weight and a smaller anogenital distance. The body weight in iAs-exposed pups continued to increase significantly compared to control at 3 weeks and 11 weeks of age. At 5 months of age, iAs-exposed males exhibited greater body fat content and glucose intolerance. Male offspring exposed to 10 ppb in utero had higher circulating levels of leptin compared to control. In addition, males exposed to 42.5 ppm in utero exhibited decreased total number of pups born compared to controls and lower average number of litters sired over a six-month period. These results indicate that in utero exposure to iAs at either human relevant concentration or tumor-inducing concentration is a potential cause of developmental origin of metabolic and reproductive dysfunction in adult male mice.
Collapse
Affiliation(s)
- Karina F Rodriguez
- Reproductive Developmental Biology Group, Reproduction and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Namya Mellouk
- Reproductive Developmental Biology Group, Reproduction and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Erica K Ungewitter
- Reproductive Developmental Biology Group, Reproduction and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Barbara Nicol
- Reproductive Developmental Biology Group, Reproduction and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Chang Liu
- Reproductive Developmental Biology Group, Reproduction and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Paula R Brown
- Reproductive Developmental Biology Group, Reproduction and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Cynthia J Willson
- Integrated Laboratory Systems, Inc., Research Triangle Park, NC, United States
| | - Humphrey H-C Yao
- Reproductive Developmental Biology Group, Reproduction and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States.
| |
Collapse
|
24
|
Liu J, Gunewardena S, Yue Cui J, Klaassen CD, Chorley BN, Corton JC. Transplacental arsenic exposure produced 5-methylcytosine methylation changes and aberrant microRNA expressions in livers of male fetal mice. Toxicology 2020; 435:152409. [PMID: 32068019 PMCID: PMC10546472 DOI: 10.1016/j.tox.2020.152409] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 02/11/2020] [Accepted: 02/13/2020] [Indexed: 10/25/2022]
Abstract
Arsenic is a known human carcinogen. Early-life exposure to inorganic arsenic induces tumors in humans and in C3H mice. We hypothesized that arsenic exposure in utero may induce epigenetic changes at the level of DNA methylation and miRNA alterations that could lead to greater postnatal susceptibility to cancer. To test this hypothesis, pregnant C3H mice were given sodium arsenite at doses known to cause liver cancer (42.5 and 85 ppm in the drinking water) from gestation day 8-19, and the livers from male fetal mice were collected for analysis. The antibody against 5-methylcytosine was used to perform chromatin-immunoprecipitation coupled with sequencing (ChIP-Seq) to determine genome-wide methylation alterations. In utero arsenic exposure produced global DNA hypomethylation and an array of gene-specific DNA methylation changes, including hypomethylation of Cyclin D1 and hypermethylation of Tp53. Illumina Correlation Engine analysis revealed 260 methylation alterations that would affect 143 microRNAs. MicroRNA array further revealed 140 aberrantly expressed miRNAs out of the 718 miRNAs. The increased expression of miR-205, miR-203, miR-215, miR-34a, and decreased expression of miR-217 were confirmed by qPCR. Comparison of the methylation changes to those of microarray analyses indicates little if any correspondence between gene methylation and gene expression. The increased expression of Xist, Prrc2, Krit1, Nish, and decreased expression of Prss2, Spp1, Col1a2, and Lox were confirmed by qPCR. In summary, in utero arsenic exposure induced global alterations in DNA methylation and aberrant miRNA expression that might contribute to adult adverse outcomes including liver cancer.
Collapse
Affiliation(s)
- Jie Liu
- University of Kansas Medical Center, Kansas City, KS 66160, United States; USEPA, Office of Research and Development, National Health and Environmental Effect Research Laboratory, Research Triangle Park, NC 27711, United States; Key Lab of Pharmacology of Ministry of Education, Zunyi Medical College, Zunyi, China.
| | | | - Julia Yue Cui
- University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Curtis D Klaassen
- University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Brian N Chorley
- USEPA, Office of Research and Development, National Health and Environmental Effect Research Laboratory, Research Triangle Park, NC 27711, United States
| | - J Christopher Corton
- USEPA, Office of Research and Development, National Health and Environmental Effect Research Laboratory, Research Triangle Park, NC 27711, United States.
| |
Collapse
|
25
|
Wang Z, Yang P, Xie J, Lin HP, Kumagai K, Harkema J, Yang C. Arsenic and benzo[a]pyrene co-exposure acts synergistically in inducing cancer stem cell-like property and tumorigenesis by epigenetically down-regulating SOCS3 expression. ENVIRONMENT INTERNATIONAL 2020; 137:105560. [PMID: 32062438 PMCID: PMC7099608 DOI: 10.1016/j.envint.2020.105560] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 02/05/2020] [Accepted: 02/06/2020] [Indexed: 05/23/2023]
Abstract
Arsenic and benzo[a]pyrene (BaP) are among the most common environmental carcinogens causing lung cancer. Millions of people are exposed to arsenic through consuming arsenic-contaminated drinking water. High levels of BaP are found in well-done barbecued meat and other food in addition to cigarette smoke. Hence, arsenic and BaP co-exposure in humans is common. However, the combined health effect and the underlying mechanism of arsenic and BaP co-exposure have not been well-understood. In this study we investigate the combined tumorigenic effect of arsenic and BaP co-exposure and the mechanism using both cell culture and mouse models. It was found that arsenic (sodium arsenite, 1.0 µM) and BaP (2.5 µM) co-exposure for 30 weeks synergizes in inducing malignant transformation of immortalized non-tumorigenic human bronchial epithelial cells and cancer stem cell (CSC)-like property to enhance their tumorigenicity. In animal studies, A/J mice were exposed to arsenic in drinking water (sodium arsenite, 20 ppm) starting from gestation day 18. After birth, the dams continuously received arsenic water throughout lactation. At weaning (3 weeks of age), male offspring were exposed to either arsenic alone via drinking the same arsenic water or exposed to arsenic plus BaP. BaP was administered via oral gavage (3 µmol per mouse per week) once a week starting from 3 weeks of age for 8 weeks. All mice were euthanized 34-weeks after the first BaP exposure. It was found that mice in control and arsenic exposure alone group did not develop lung tumors. All mice in BaP exposure alone group developed lung adenomas. However, arsenic and BaP co-exposure synergized in increasing lung tumor multiplicity and tumor burden. Furthermore, 30% of mice in arsenic and BaP co-exposure group also developed lung adenocarcinomas. Mechanistic studies revealed that arsenic and BaP co-exposure does not produce more BPDE-DNA adducts than BaP exposure alone; but acts synergistically in activating aryl hydrocarbon receptor (AhR) to up-regulate the expression of a histone H3 lysine 9 methyltransferase SUV39H1 and increase the level of suppressive H3 lysine 9 dimethylation (H3K9me2), which down-regulates the expression of tumor suppressive SOCS3 leading to enhanced activation of Akt and Erk1/2 to promote cell transformation, CSC-like property and tumorigenesis. Together, these findings suggest that arsenic and BaP co-exposure synergizes in causing epigenetic dysregulation to enhance cell transformation, CSC-like property and tumorigenesis.
Collapse
Affiliation(s)
- Zhishan Wang
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, USA.
| | - Ping Yang
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, USA; School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Jie Xie
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, USA; School of Health Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Hsuan-Pei Lin
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Kazuyoshi Kumagai
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Jack Harkema
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Chengfeng Yang
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
26
|
Chen QY, Shen S, Sun H, Wu F, Kluz T, Kibriya MG, Chen Y, Ahsan H, Costa M. PBMC gene expression profiles of female Bangladeshi adults chronically exposed to arsenic-contaminated drinking water. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 259:113672. [PMID: 31918125 PMCID: PMC11062206 DOI: 10.1016/j.envpol.2019.113672] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/06/2019] [Accepted: 11/22/2019] [Indexed: 06/10/2023]
Abstract
Arsenic, a class I human carcinogen, is ubiquitously found throughout the environment and around the globe, posing a great public health concern. Notably, Bangladesh and regions of West Bengal have been found to have high levels (0.5-4600 μg/L) of arsenic drinking water contamination, and approximately 50 million of the world's 200 million people chronically exposed to arsenic in Bangladesh alone. This study was carried out to examine genome-wide gene expression changes in individuals chronically exposed to arsenic-contaminated drinking water. Our study population includes twenty-nine Bangladeshi female participants with urinary arsenic levels ranging from 22.32 to 1828.12 μg/g creatinine. RNA extracted from peripheral blood mononuclear cells (PBMCs) were evaluated using RNA-Sequencing analysis. Our results indicate that a total of 1,054 genes were significantly associated with increasing urinary arsenic levels (FDR p < 0.05), which include 418 down-regulated and 636 up-regulated genes. Further Ingenuity Pathway Analysis revealed potential target genes (DAPK1, EGR2, APP), microRNAs (miR-155, -338, -210) and pathways (NOTCH signaling pathway) related to arsenic carcinogenesis. The selection of female-only participants provides a homogenous study population since arsenic has significant sex dependent effects, and the wide exposure range provides new insight for key gene expression changes that correlate with increasing urinary arsenic levels.
Collapse
Affiliation(s)
- Qiao Yi Chen
- Department of Environmental Medicine, New York University School of Medicine, 10010, New York, NY, USA.
| | - Steven Shen
- Institute of Health Informatics, University of Minnesota, 55455, Minneapolis, MN, USA
| | - Hong Sun
- Department of Environmental Medicine, New York University School of Medicine, 10010, New York, NY, USA
| | - Fen Wu
- Department of Population Health and Environmental Medicine, 10016, New York University School of Medicine, New York, NY, USA
| | - Thomas Kluz
- Department of Environmental Medicine, New York University School of Medicine, 10010, New York, NY, USA
| | - Muhammad G Kibriya
- Institute for Population and Precision Health, Department of Public Health Sciences, The University of Chicago, Chicago, IL, 60637, USA
| | - Yu Chen
- Department of Population Health and Environmental Medicine, 10016, New York University School of Medicine, New York, NY, USA
| | - Habibul Ahsan
- Institute for Population and Precision Health, Department of Public Health Sciences, The University of Chicago, Chicago, IL, 60637, USA
| | - Max Costa
- Department of Environmental Medicine, New York University School of Medicine, 10010, New York, NY, USA.
| |
Collapse
|
27
|
Ahn J, Boroje IJ, Ferdosi H, Kramer ZJ, Lamm SH. Prostate Cancer Incidence in U.S. Counties and Low Levels of Arsenic in Drinking Water. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17030960. [PMID: 32033184 PMCID: PMC7036874 DOI: 10.3390/ijerph17030960] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/27/2020] [Accepted: 01/30/2020] [Indexed: 12/20/2022]
Abstract
Background: Although inorganic arsenic in drinking water at high levels (100s–1000s μg/L [ppb]) increases cancer risk (skin, bladder, lung, and possibly prostate), the evidence at lower levels is limited. Methods: We conducted an ecologic analysis of the dose-response relationship between prostate cancer incidence and low arsenic levels in drinking water in a large study of U.S. counties (N = 710). County arsenic levels were <200 ug/L with median <100 ug/L and dependency greater than 10%. Groundwater well usage, water arsenic levels, prostate cancer incidence rates (2009–2013), and co-variate data were obtained from various U.S. governmental agencies. Poisson and negative-binomial regression analyses and stratified analysis were performed. Results: The best fitting polynomial analysis yielded a J-shaped linear-quadratic model. Linear and quadratic terms were significant (p < 0.001) in the Poisson model, and the quadratic term was significant (p < 0.05) in the negative binomial model. This model indicated a decreasing risk of prostate cancer with increasing arsenic level in the low range and increasing risk above. Conclusions: This study of prostate cancer incidence in US counties with low levels of arsenic in their well-water arsenic levels finds a j-shaped model with decreasing risk at very low levels and increasing risk at higher levels.
Collapse
Affiliation(s)
- Jaeil Ahn
- Department of Biostatistics, Bioinformatics, and Biomathematics, Georgetown University School of Medicine, Washington, DC 20007, USA;
| | - Isabella J. Boroje
- Center for Epidemiology and Environmental Health (CEOH, LLC), Washington, DC 20016, USA; (I.J.B.); (H.F.); (Z.J.K.)
- Department of Epidemiology and Biostatistics, Milken Institute School of Public Health, George Washington University, Washington, DC 20052, USA
- Department of Global Health, Milken Institute School of Public Health, George Washington University, Washington, DC 20052, USA
| | - Hamid Ferdosi
- Center for Epidemiology and Environmental Health (CEOH, LLC), Washington, DC 20016, USA; (I.J.B.); (H.F.); (Z.J.K.)
- Department of Epidemiology and Biostatistics, Milken Institute School of Public Health, George Washington University, Washington, DC 20052, USA
| | - Zachary J. Kramer
- Center for Epidemiology and Environmental Health (CEOH, LLC), Washington, DC 20016, USA; (I.J.B.); (H.F.); (Z.J.K.)
| | - Steven H. Lamm
- Center for Epidemiology and Environmental Health (CEOH, LLC), Washington, DC 20016, USA; (I.J.B.); (H.F.); (Z.J.K.)
- Department of Health Policy and Management, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Pediatrics, Georgetown University School of Medicine, Washington, DC 20007, USA
- Correspondence:
| |
Collapse
|
28
|
Coryell M, Roggenbeck BA, Walk ST. The Human Gut Microbiome's Influence on Arsenic Toxicity. CURRENT PHARMACOLOGY REPORTS 2019; 5:491-504. [PMID: 31929964 PMCID: PMC6953987 DOI: 10.1007/s40495-019-00206-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Arsenic exposure is a public health concern of global proportions with a high degree of interindividual variability in pathologic outcomes. Arsenic metabolism is a key factor underlying toxicity, and the primary purpose of this review is to summarize recent discoveries concerning the influence of the human gut microbiome on the metabolism, bioavailability, and toxicity of ingested arsenic. We review and discuss the current state of knowledge along with relevant methodologies for studying these phenomena. RECENT FINDINGS Bacteria in the human gut can biochemically transform arsenic-containing compounds (arsenicals). Recent publications utilizing culture-based approaches combined with analytical biochemistry and molecular genetics have helped identify several arsenical transformations by bacteria that are at least possible in the human gut and are likely to mediate arsenic toxicity to the host. Other studies that directly incubate stool samples in vitro also demonstrate the gut microbiome's potential to alter arsenic speciation and bioavailability. In vivo disruption or elimination of the microbiome has been shown to influence toxicity and body burden of arsenic through altered excretion and biotransformation of arsenicals. Currently, few clinical or epidemiological studies have investigated relationships between the gut microbiome and arsenic-related health outcomes in humans, although current evidence provides strong rationale for this research in the future. SUMMARY The human gut microbiome can metabolize arsenic and influence arsenical oxidation state, methylation status, thiolation status, bioavailability, and excretion. We discuss the strength of current evidence and propose that the microbiome be considered in future epidemiologic and toxicologic studies of human arsenic exposure.
Collapse
Affiliation(s)
- Michael Coryell
- Department of Microbiology and Immunology, Montana State University, 109 Lewis Hall, Bozeman, MT 59717, USA
| | - Barbara A. Roggenbeck
- Department of Microbiology and Immunology, Montana State University, 109 Lewis Hall, Bozeman, MT 59717, USA
| | - Seth T. Walk
- Department of Microbiology and Immunology, Montana State University, 109 Lewis Hall, Bozeman, MT 59717, USA
| |
Collapse
|
29
|
Grosse Y, Lajoie P, Billard M, Krewski D, Rice J, Baan RA, Cogliano V, Bird M, Zielinski JM. Development of a database on tumors and tumor sites in humans and in experimental animals for 'Group 1 agents identified through volume 109 of the IARC Monographs. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2019; 22:237-243. [PMID: 31612803 DOI: 10.1080/10937404.2019.1642601] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Volume 100 in the series of IARC Monographs on the Evaluation of Carcinogenic Risks to Humans comprises an update and review of relevant information on all agents determined to induce cancer in humans. These Group 1 agents are categorized in 6 Monographs (Volumes 100A-F) published in 2012. This paper describes the methodology and stringent criteria used in the creation of a comprehensive database on tumors noted in animals and humans for the carcinogens reviewed in Volume 100, and for additional Group 1 agents that were identified in subsequent Monographs through Volume 109. The development of this database involved the systematic collection of relevant data on tumors detected in humans and experimental animals identified by the Working Groups that conducted evaluations reported in the IARC Monographs. The database includes all human tumor sites identified by the Working Groups, along with all tumor sites for which there was sufficient evidence in experimental animals. This database provides a basis for assessing the degree of concordance between tumor sites observed in humans and experimental animals for Group 1 agents identified through Volume 109.
Collapse
Affiliation(s)
- Yann Grosse
- IARC Monographs Programme, International Agency for Research on Cancer, Lyon, France
| | - Pascale Lajoie
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, Canada
- Division of Cancer Care and Epidemiology, Queen's University Cancer Research Institute, Kingston, Canada
| | - Mélissa Billard
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, Canada
| | - Daniel Krewski
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, Canada
- Risk Sciences International, Ottawa, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada
| | - Jerry Rice
- School of Medicine, Georgetown University, Washington, D.C., USA
| | - Robert A Baan
- International Agency for Research on Cancer (retired), Lyon, France
| | - Vincent Cogliano
- Integrated Risk Information System, US Environmental Protection Agency, Washington, D.C., USA
| | - Michael Bird
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, Canada
| | - Jan M Zielinski
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, Canada
| |
Collapse
|
30
|
Lipid Metabolism Alterations in a Rat Model of Chronic and Intergenerational Exposure to Arsenic. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4978018. [PMID: 31737665 PMCID: PMC6815581 DOI: 10.1155/2019/4978018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/17/2019] [Accepted: 08/20/2019] [Indexed: 12/26/2022]
Abstract
Chronic exposure to arsenic (As), whether directly through the consumption of contaminated drinking water or indirectly through the daily intake of As-contaminated food, is a health threat for more than 150 million people worldwide. Epidemiological studies found an association between chronic consumption of As and several pathologies, the most common being cancer-related disorders. However, As consumption has also been associated with metabolic disorders that could lead to diverse pathologies, such as type 2 diabetes mellitus, nonalcoholic fatty liver disease, and obesity. Here, we used ultra-performance liquid chromatography (UPLC) coupled to electrospray ionization/quadrupole time-of-flight mass spectrometry (ESI-QToF) to assess the effect of chronic intergenerational As exposure on the lipid metabolism profiles of serum from 4-month-old Wistar rats exposed to As prenatally and also during early life in drinking water (3 ppm). Significant differences in the levels of certain identified lysophospholipids, phosphatidylcholines, and triglycerides were found between the exposed rats and the control groups, as well as between the sexes. Significantly increased lipid oxidation determined by the malondialdehyde (MDA) method was found in exposed rats compared with controls. Chronic intergenerational As exposure alters the rat lipidome, increases lipid oxidation, and dysregulates metabolic pathways, the factors associated with the chronic inflammation present in different diseases associated with chronic exposure to As (i.e., keratosis, Bowen's disease, and kidney, liver, bladder, and lung cancer).
Collapse
|
31
|
Barajas-Olmos FM, Ortiz-Sánchez E, Imaz-Rosshandler I, Córdova-Alarcón EJ, Martínez-Tovar A, Villanueva-Toledo J, Morales-Marín ME, Cruz-Colín JL, Rangel C, Orozco L, Centeno F. Analysis of the dynamic aberrant landscape of DNA methylation and gene expression during arsenic-induced cell transformation. Gene 2019; 711:143941. [PMID: 31242453 DOI: 10.1016/j.gene.2019.143941] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/30/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023]
Abstract
Inorganic arsenic is a well-known carcinogen associated with several types of cancer, but the mechanisms involved in arsenic-induced carcinogenesis are not fully understood. Recent evidence points to epigenetic dysregulation as an important mechanism in this process; however, the effects of epigenetic alterations in gene expression have not been explored in depth. Using microarray data and applying a multivariate clustering analysis in a Gaussian mixture model, we describe the alterations in DNA methylation around the promoter region and the impact on gene expression in HaCaT cells during the transformation process caused by chronic exposure to arsenic. Using this clustering approach, the genes were grouped according to their methylation and expression status in the epigenetic landscape, and the changes that occurred during the cellular transformation were identified adequately. Thus, we present a valuable method for identifying epigenomic dysregulation.
Collapse
Affiliation(s)
- Francisco M Barajas-Olmos
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, Instituto Nacional de Medicina Genómica, Ciudad de México, Mexico
| | - Elizabeth Ortiz-Sánchez
- Subdireccion de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México, Mexico
| | - Ivan Imaz-Rosshandler
- DAMTP, Centre for Mathematical Sciences, University of Cambridge, Cambridge CB3 OWA, UK
| | | | - Adolfo Martínez-Tovar
- Laboratorio de Biología Molecular, Servicio de Hematología, Hospital General de México "Dr. Eduardo Liceaga", Ciudad de México, Mexico
| | - Jairo Villanueva-Toledo
- Centro de Investigación en Salud "Dr. Jesús Kumate Rodríguez", Instituto Mexicano del Seguro Social, Mérida, Yucatán, Mexico; Cátedras CONACYT - Fundación IMSS AC, CONACYT, Ciudad de México, Mexico
| | - Mirna E Morales-Marín
- Laboratorio de Genómica de Enfermedades Psiquiátricas y Neurodegenerativas, Instituto Nacional de Medicina Genómica, Ciudad de México, Mexico
| | - José L Cruz-Colín
- Subdirección de Investigación Básica, Instituto Nacional de Medicina Genómica, Ciudad de México, Mexico
| | - Claudia Rangel
- Computational Genomics Consortium, Instituto Nacional de Medicina Genómica, Ciudad de México, Mexico
| | - Lorena Orozco
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, Instituto Nacional de Medicina Genómica, Ciudad de México, Mexico
| | - Federico Centeno
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, Instituto Nacional de Medicina Genómica, Ciudad de México, Mexico.
| |
Collapse
|
32
|
Twaddle NC, Beland FA, Doerge DR. Metabolism and disposition of arsenic species from controlled dosing with sodium arsenite in adult and neonatal rhesus monkeys. VI. Toxicokinetic studies following oral administration. Food Chem Toxicol 2019; 133:110760. [PMID: 31421213 DOI: 10.1016/j.fct.2019.110760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/09/2019] [Accepted: 08/12/2019] [Indexed: 12/25/2022]
Abstract
Arsenic is a common toxic contaminant in food and drinking water. Metabolic activation of arsenic species produces reactive trivalent intermediates that can disrupt cellular regulatory systems by covalent binding to thiol groups. Arsenic exposures have been associated with human diseases including cancer, diabetes, lung and cardiovascular disorders and there is accumulating evidence that early life exposures are important in the etiology. Previous toxicokinetic studies of arsenite ingestion in neonatal CD-1 mice showed consistent evidence for metabolic and physiologic immaturity that led to elevated internal exposures to trivalent arsenic species in the youngest mice, relative to adults. The current study in rhesus monkeys showed that metabolism and binding of trivalent intermediates after arsenite ingestion were similar between adult monkeys and CD-1 mice. Unlike neonatal mice, monkeys from the age of 5-70 days showed similar metabolism and binding profiles, which were also similar to those in adults. The absence of evidence for metabolic immaturity in monkeys suggests that toxicological effects observed in mice from early postnatal exposures to arsenic could over-predict those possible in primates, based on significantly higher internal exposures.
Collapse
Affiliation(s)
- Nathan C Twaddle
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Frederick A Beland
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Daniel R Doerge
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA.
| |
Collapse
|
33
|
Wang Z, Yang C. Metal carcinogen exposure induces cancer stem cell-like property through epigenetic reprograming: A novel mechanism of metal carcinogenesis. Semin Cancer Biol 2019; 57:95-104. [PMID: 30641125 PMCID: PMC6625953 DOI: 10.1016/j.semcancer.2019.01.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/08/2019] [Accepted: 01/10/2019] [Indexed: 12/13/2022]
Abstract
Arsenic, cadmium, nickel and hexavalent chromium are among the most common environmental pollutants and potent carcinogens. Chronic exposure to these metals causes various types of cancer in humans, representing a significant environmental health issue. Although under active investigation, the mechanisms of metal carcinogenesis have not been clearly defined. One common feature of these metal carcinogens is that they are all able to cause various epigenetic dysregulations, which are believed to play important roles in their carcinogenicity. However, how metal carcinogen-caused epigenetic dysregulation contributes to metal carcinogenesis remains largely unknown. The evolution of cancer stem cell (CSC) theory has opened exciting new avenues for studying the mechanism of metal carcinogenesis. Increasing evidence indicates that chronic metal carcinogen exposure produces CSC-like cells through dysregulated epigenetic mechanisms. This review will first provide some brief introductions about CSC, epigenetics and epigenetic regulation of CSCs; then summarize progresses in recent studies on metal carcinogen-induced CSC-like property through epigenetic reprograming as a novel mechanism of metal carcinogenesis. Some perspectives for future studies in this field are also presented.
Collapse
Affiliation(s)
- Zhishan Wang
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY, United States.
| | - Chengfeng Yang
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY, United States; Center for Research on Environment Disease, College of Medicine, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
34
|
Twaddle NC, Vanlandingham M, Beland FA, Doerge DR. Metabolism and disposition of arsenic species from controlled dosing with dimethylarsinic acid (DMAV) in adult female CD-1 mice. V. Toxicokinetic studies following oral and intravenous administration. Food Chem Toxicol 2019; 130:22-31. [DOI: 10.1016/j.fct.2019.04.045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/23/2019] [Accepted: 04/24/2019] [Indexed: 12/26/2022]
|
35
|
Arsenic induces gender difference of estrogen receptor in AECII cells from ICR fetal mice. Toxicol In Vitro 2019; 56:133-140. [DOI: 10.1016/j.tiv.2019.01.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 01/10/2019] [Accepted: 01/21/2019] [Indexed: 11/30/2022]
|
36
|
Tsuji JS, Chang ET, Gentry PR, Clewell HJ, Boffetta P, Cohen SM. Dose-response for assessing the cancer risk of inorganic arsenic in drinking water: the scientific basis for use of a threshold approach. Crit Rev Toxicol 2019; 49:36-84. [DOI: 10.1080/10408444.2019.1573804] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
| | - Ellen T. Chang
- Exponent, Inc., Menlo Park, CA and Stanford Cancer Institute, Stanford, CA, USA
| | | | | | - Paolo Boffetta
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samuel M. Cohen
- Havlik-Wall Professor of Oncology, Department of Pathology and Microbiology and the Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
37
|
|
38
|
Long-Term Health Effects and Underlying Biological Mechanisms of Developmental Exposure to Arsenic. Curr Environ Health Rep 2019; 5:134-144. [PMID: 29411302 DOI: 10.1007/s40572-018-0184-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE OF REVIEW Exposure to inorganic arsenic (iAs) via drinking water represents a significant global public health threat with chronic exposure associated with cancer, skin lesions, neurological impairment, and cardiovascular diseases. Particularly susceptible populations include the developing fetus and young children. This review summarizes some of the critical studies of the long-term health effects and underlying biological mechanisms related to developmental exposure to arsenic. It also highlights the complex factors, such as the sex of the exposed individual, that contribute to susceptibility to the later life health effects of iAs. RECENT FINDINGS Studies in animal models, as well as human population-based studies, have established that prenatal and early life iAs exposures are associated with long-term effects, and many of these effects display sexually dimorphic responses. As an underlying molecular basis, recent epidemiologic and toxicologic studies have demonstrated that changes to the epigenome may play a key mechanistic role underlying many of the iAs-associated health outcomes. Developmental exposure to iAs results in early and later life health effects. Mechanisms underlying these outcomes are likely complex, and include disrupted key biological pathways with ties to the epigenome. This highlights the importance of continued research, particularly in animal models, to elucidate the important underpinnings (e.g., timing of exposure, metabolism, dose) of these complex health outcomes and to identify the biological mechanisms underlying sexual dimorphism in iAs-associated diseases. Future research should investigate preventative strategies for the protection from the detrimental health endpoints associated with early life exposure to iAs. Such strategies could include potential interventions focused on dietary supplementation for example the adoption of a folate-rich diet.
Collapse
|
39
|
Dreval K, Tryndyak V, Kindrat I, Twaddle NC, Orisakwe OE, Mudalige TK, Beland FA, Doerge DR, Pogribny IP. Cellular and Molecular Effects of Prolonged Low-Level Sodium Arsenite Exposure on Human Hepatic HepaRG Cells. Toxicol Sci 2019; 162:676-687. [PMID: 29301061 DOI: 10.1093/toxsci/kfx290] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Inorganic arsenic is a human carcinogen associated with several types of cancers, including liver cancer. Inorganic arsenic has been postulated to target stem cells, causing their oncogenic transformation. This is proposed to be one of the key events in arsenic-associated carcinogenesis; however, the underlying mechanisms for this process remain largely unknown. To address this question, human hepatic HepaRG cells, at progenitor and differentiated states, were continuously treated with a noncytotoxic concentration of 1 μM sodium arsenite (NaAsO2). The HepaRG cells demonstrated active intracellular arsenite metabolism that shared important characteristic with primary human hepatocytes. Treatment of proliferating progenitor-like HepaRG cells with NaAsO2 inhibited their differentiation into mature hepatocyte-like cells, up-regulated genes involved in cell growth, proliferation, and survival, and down-regulated genes involved in cell death. In contrast, treatment of differentiated hepatocyte-like HepaRG cells with NaAsO2 resulted in enhanced cell death of mature hepatocyte-like cells, overexpression of cell death-related genes, and down-regulation of genes in the cell proliferation pathway, while biliary-like cells remained largely unaffected. Mechanistically, the cytotoxic effect of arsenic on mature hepatocyte-like HepaRG cells may be attributed to arsenic-induced dysregulation of cellular iron metabolism. The inhibitory effect of NaAsO2 on the differentiation of progenitor cells, the resistance of biliary-like cells to cell death, and the enhanced cell death of functional hepatocyte-like cells resulted in stem-cell activation. These effects favored the proliferation of liver progenitor cells that can serve as a source of initiation and driving force of arsenic-mediated liver carcinogenesis.
Collapse
Affiliation(s)
- Kostiantyn Dreval
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, Arkansas 72079
| | - Volodymyr Tryndyak
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, Arkansas 72079
| | - Iryna Kindrat
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, Arkansas 72079.,Department of Biological and Medical Chemistry, Ivano-Frankivsk National Medical University, Ivano-Frankivsk, Ukraine
| | - Nathan C Twaddle
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, Arkansas 72079
| | - Orish Ebere Orisakwe
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, Arkansas 72079.,Department of Experimental Pharmacology and Toxicology, University of Port-Harcourt, Rivers State, Nigeria
| | - Thilak K Mudalige
- Office of Regulatory Affairs, Arkansas Regional Laboratory, U.S. Food and Drug Administration, Jefferson, Arkansas 72079
| | - Frederick A Beland
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, Arkansas 72079
| | - Daniel R Doerge
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, Arkansas 72079
| | - Igor P Pogribny
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, Arkansas 72079
| |
Collapse
|
40
|
Genome-wide epigenetic signatures of childhood adversity in early life: Opportunities and challenges. J Dev Orig Health Dis 2019; 10:65-72. [PMID: 30744719 DOI: 10.1017/s2040174418000843] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Maternal adversity and fetal glucocorticoid exposure has long-term effects on cardiovascular, metabolic and behavioral systems in offspring that can persist throughout the lifespan. These data, along with other environmental exposure data, implicate epigenetic modifications as potential mechanisms for long-term effects of maternal exposures on adverse health outcomes in offspring. Advances in microarray, sequencing and bioinformatic approaches have enabled recent studies to examine the genome-wide epigenetic response to maternal adversity. Studies of maternal exposures to xenobiotics such as arsenic and smoking have been performed at birth to examine fetal epigenomic signatures in cord blood relating to adult health outcomes. However, there have been no epigenomic studies examining these effects in animal models. On the other hand, to date, only a few studies of the effects of maternal psychosocial stress have been performed in human infants, and the majority of animal studies have examined epigenomic outcomes in adulthood. In terms of maternal exposure to excess glucocorticoids by synthetic glucocorticoid treatment, there has been no epigenetic study performed in humans and only a few studies undertaken in animal models. This review emphasizes the importance of examining biomarkers of exposure to adversity throughout development to identify individuals at risk and to target interventions. Thus, research performed at birth will be reviewed. In addition, potential subject characteristics associated with epigenetic modifications, technical considerations, the selection of target tissues and combining human studies with animal models will be discussed in relation to the design of experiments in this field of study.
Collapse
|
41
|
Selmin OI, Donovan MG, Skovan B, Paine-Murieta GD, Romagnolo DF. Arsenic‑induced BRCA1 CpG promoter methylation is associated with the downregulation of ERα and resistance to tamoxifen in MCF7 breast cancer cells and mouse mammary tumor xenografts. Int J Oncol 2019; 54:869-878. [PMID: 30664189 PMCID: PMC6365020 DOI: 10.3892/ijo.2019.4687] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/03/2018] [Indexed: 02/07/2023] Open
Abstract
A significant percentage (~30%) of estrogen receptor-α (ERα)-positive tumors become refractory to endocrine therapies; however, the mechanisms responsible for this resistance remain largely unknown. Chronic exposure to arsenic through foods and contaminated water has been linked to an increased incidence of several tumors and long-term health complications. Preclinical and population studies have indicated that arsenic exposure may interfere with endocrine regulation and increase the risk of breast tumorigenesis. In this study, we examined the effects of sodium arsenite (NaAsIII) exposure in ERα-positive breast cancer cells in vitro and in mammary tumor xenografts. The results revealed that acute (within 4 days) and long-term (10 days to 7 weeks) in vitro exposure to environmentally relevant doses reduced breast cancer 1 (BRCA1) and ERα expression associated with the gain of cyclin D1 (CCND1) and folate receptor 1 (FOLR1), and the loss of methylenetetrahydrofolate reductase (MTHFR) expression. Furthermore, long-term exposure to NaAsIII induced the proliferation and compromised the response of MCF7 cells to tamoxifen (TAM). The in vitro exposure to NaAsIII induced BRCA1 CpG methylation associated with the increased recruitment of DNA methyltransferase 1 (DNMT1) and the loss of RNA polymerase II (PolII) at the BRCA1 gene. Xenografts of NaAsIII-preconditioned MCF7 cells (MCF7NaAsIII) into the mammary fat pads of nude mice produced a larger tumor volume compared to tumors from control MCF7 cells and were more refractory to TAM in association with the reduced expression of BRCA1 and ERα, CpG hypermethylation of estrogen receptor 1 (ESR1) and BRCA1, and the increased expression of FOLR1. These cumulative data support the hypothesis that exposure to AsIII may contribute to reducing the efficacy of endocrine therapy against ERα-positive breast tumors by hampering the expression of ERα and BRCA1 via CpG methylation, respectively of ESR1 and BRCA1.
Collapse
Affiliation(s)
- Ornella I Selmin
- The University of Arizona Cancer Center, The University of Arizona, Tucson, AZ 85724, USA
| | - Micah G Donovan
- Cancer Biology Graduate Interdisciplinary Program, The University of Arizona, Tucson, AZ 85724, USA
| | - Bethany Skovan
- The University of Arizona Cancer Center, The University of Arizona, Tucson, AZ 85724, USA
| | | | - Donato F Romagnolo
- The University of Arizona Cancer Center, The University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
42
|
Krewski D, Rice JM, Bird M, Milton B, Collins B, Lajoie P, Billard M, Grosse Y, Cogliano VJ, Caldwell JC, Rusyn II, Portier CJ, Melnick RL, Baan RA, Little J, Zielinski JM. Concordance between sites of tumor development in humans and in experimental animals for 111 agents that are carcinogenic to humans. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2019; 22:203-236. [PMID: 31795923 PMCID: PMC7139235 DOI: 10.1080/10937404.2019.1642586] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Since the inception of the IARC Monographs Programme in the early 1970s, this Programme has developed 119 Monograph Volumes on more than 1000 agents for which there exists some evidence of cancer risk to humans. Of these, 120 agents were found to meet the criteria for classification as carcinogenic to humans (Group 1). Volume 100 of the IARC Monographs, compiled in 2008-2009 and published in 2012, provided a review and update of the 107 Group 1 agents identified as of 2009. These agents were divided into six broad categories: (I) pharmaceuticals; (II) biological agents; (III) arsenic, metals, fibers and dusts; (IV) radiation; (V) personal habits and indoor combustions; and (VI) chemical agents and related occupations. The Group I agents reviewed in Volume 100, as well as five additional Group 1 agents defined in subsequent Volumes of the Monographs, were used to assess the degree of concordance between sites where tumors originate in humans and experimental animals including mice, rats, hamsters, dogs, and non-human primates using an anatomically based tumor nomenclature system, representing 39 tumor sites and 14 organ and tissue systems. This evaluation identified 91 Group 1 agents with sufficient evidence (82 agents) or limited evidence (9 agents) of carcinogenicity in animals. The most common tumors observed in both humans and animals were those of the respiratory system including larynx, lung, and lower respiratory tract. In humans, respiratory system tumors were noted for 31 of the 111 distinct Group 1 carcinogens identified up to and including Volume 109 of the IARC Monographs, comprising predominantly 14 chemical agents and related occupations in category VI; seven arsenic, metals, fibers, and dusts in category III, and five personal habits and indoor combustions in category V. Subsequent to respiratory system tumors, those in lymphoid and hematopoietic tissues (26 agents), the urothelium (18 agents), and the upper aerodigestive tract (16 agents) were most often seen in humans, while tumors in digestive organs (19 agents), skin (18 agents), and connective tissues (17 agents) were frequently seen in animals. Exposures to radiation, particularly X- and γ-radiation, and tobacco smoke were associated with tumors at multiple sites in humans. Although the IARC Monographs did not emphasize tumor site concordance between animals and humans, substantial concordance was detected for several organ and tissue systems, even under the stringent criteria for sufficient evidence of carcinogenicity used by IARC. Of the 60 agents for which at least one tumor site was identified in both humans and animals, 52 (87%) exhibited tumors in at least one of the same organ and tissue systems in humans and animals. It should be noted that some caution is needed in interpreting concordance at sites where sample size is particularly small. Although perfect (100%) concordance was noted for agents that induce tumors of the mesothelium, only two Group 1 agents that met the criteria for inclusion in the concordance analysis caused tumors at this site. Although the present analysis demonstrates good concordance between animals and humans for many, but not all, tumor sites, limitations of available data may result in underestimation of concordance.
Collapse
Affiliation(s)
- Daniel Krewski
- McLaughlin Centre for Population Health Risk Assessment, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Risk Sciences International, Ottawa, Canada
- School of Epidemiology, Public Health and Preventive Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Jerry M. Rice
- Department of Oncology, Georgetown University Medical Center, Georgetown University, Washington, DC, USA
| | - Michael Bird
- McLaughlin Centre for Population Health Risk Assessment, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | | | | | - Pascale Lajoie
- McLaughlin Centre for Population Health Risk Assessment, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Division of Cancer Care and Epidemiology, Queen’s University Cancer Research Institute, Kingston, Canada
| | - Mélissa Billard
- McLaughlin Centre for Population Health Risk Assessment, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Yann Grosse
- IARC Monographs Programme, International Agency for Research on Cancer, Lyon, France
| | - Vincent J. Cogliano
- National Center for Environmental Assessment, U.S. Environmental Protection Agency, Washington, DC, USA
| | - Jane C. Caldwell
- National Center for Environmental Assessment, U.S. Environmental Protection Agency, Washington, DC, USA
| | - Ivan I. Rusyn
- Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Christopher J. Portier
- National Center for Environmental Health, Agency for Toxic Substances and Disease Registry, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Robert A. Baan
- International Agency for Research on Cancer (retired), Lyon, France
| | - Julian Little
- School of Epidemiology, Public Health and Preventive Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Jan M. Zielinski
- McLaughlin Centre for Population Health Risk Assessment, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- School of Epidemiology, Public Health and Preventive Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
43
|
Young JL, Cai L, States JC. Impact of prenatal arsenic exposure on chronic adult diseases. Syst Biol Reprod Med 2018; 64:469-483. [PMID: 29873257 PMCID: PMC6291241 DOI: 10.1080/19396368.2018.1480076] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/03/2018] [Accepted: 05/14/2018] [Indexed: 02/08/2023]
Abstract
Exposure to environmental stressors during susceptible windows of development can result in negative health outcomes later in life, a concept known as the Developmental Origins of Health and Disease (DOHaD). There is a growing body of evidence that exposures to metals early in life (in utero and postnatal) increase the risk of developing adult diseases such as cancer, cardiovascular disease, non-alcoholic fatty liver disease, and diabetes. Of particular concern is exposure to the metalloid arsenic, a drinking water contaminant and worldwide health concern. Epidemiological studies of areas with high levels of arsenic in the drinking water, such as some regions in Chile and Bangladesh, indicate an association between in utero arsenic exposure and the development of adult diseases. Therefore, the need for experimental models to address the mechanism underlining early onset of adult diseases have emerged including the in utero and whole-life exposure models. This review will highlight the epidemiological events and subsequent novel experimental models implemented to study the impact of early life exposure to arsenic on the development of adult diseases. In addition, current research using these models will be discussed as well as possible underlying mechanism for the early onset of disease. Abbreviations: ALT: alanine aminotransferase; AMI: acute myocardial infarction; AST: aspartate aminotransferase; ATSDR: Agency for Toxic Substances and Disease Registry; CVD: cardiovascular disease; DMA: dimethylarsinate; DOHaD: Developmental Origins of Health and Disease; EPA: U.S. Environmental Protection Agency; ER-α: estrogen receptor alpha; HDL: high-density lipoprotein; HOMA-IR: homeostatic model assessment of insulin resistance; iAs: inorganic arsenic; LDL: low-density lipoprotein; MetS: metabolic syndrome; MMA: monomethylarsonate; NAFLD: non-alcoholic fatty liver disease; PND: postnatal day; ppb: parts per billion; ppm: parts per million; SAM: S-adenosylmethionine; USFDA: United States Food and Drug Administration.
Collapse
Affiliation(s)
- Jamie L. Young
- Department of Pharmacology and Toxicology, University of Louisville, 505 S. Hancock St, Louisville, KY., 40202. USA Tel.: +1 502 852 2631.
| | - Lu Cai
- Pediatric Research Institute, Departments of Pediatrics, Radiation Oncology and pharmacology and Toxicology, University of Louisville, 570 S. Preston St, Room 304F, Louisville, KY., 40202. USA Tel,: +1 502 852 2214.
| | - J. Christopher States
- Department of Pharmacology and Toxicology, University of Louisville, 505 S. Hancock St, Room 304, Louisville, KY., 40202. USA Tel.: +1 502 852 5347.
| |
Collapse
|
44
|
Metabolism and disposition of arsenic species from controlled dosing with sodium arsenite in adult female CD-1 mice. III. Toxicokinetic studies following oral and intravenous administration. Food Chem Toxicol 2018; 121:676-686. [DOI: 10.1016/j.fct.2018.09.068] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 09/11/2018] [Accepted: 09/28/2018] [Indexed: 12/20/2022]
|
45
|
Twaddle NC, Vanlandingham M, Beland FA, Fisher JW, Doerge DR. Metabolism and disposition of arsenic species from oral dosing with sodium arsenite in neonatal CD-1 mice. IV. Toxicokinetics following gavage administration and lactational transfer. Food Chem Toxicol 2018; 123:28-41. [PMID: 30342114 DOI: 10.1016/j.fct.2018.10.046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 09/27/2018] [Accepted: 10/16/2018] [Indexed: 12/18/2022]
Abstract
Arsenic is a ubiquitous contaminant, with typical human dietary intake below 1 μg/kg bw/d and extreme drinking water exposures up to ∼50 μg/kg bw/d. The formation and binding of trivalent metabolites are central to arsenic toxicity and strong human evidence suggests special concern for early life exposures in the etiology of adult diseases, especially cancer. This study measured the metabolism and disposition of arsenite in neonatal mice to understand the role of maturation in metabolic activation and detoxification of arsenic. Many age-related differences were observed after gavage administration of arsenite, with consistent evidence in blood and tissues for higher exposures to trivalent arsenic species in neonatal mice related to the immaturity of metabolic and/or excretory functions. The evidence for greater tissue binding of arsenic species in young mice is consistent with enhanced susceptibility to toxicity based on metabolic and toxicokinetic differences alone. Lactational transfer from arsenite-dosed dams to suckling mice was minimal, based on no dosing-related changes in the levels of arsenic species in pup blood or milk collected from the dams. Animal models evaluating whole-life exposure to inorganic arsenic must use direct dosing in early neonatal life to predict accurately potential toxicity from early life exposures in children.
Collapse
Affiliation(s)
- Nathan C Twaddle
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Michelle Vanlandingham
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Frederick A Beland
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Jeffrey W Fisher
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Daniel R Doerge
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA.
| |
Collapse
|
46
|
Clewell HJ, Yager JW, Greene TB, Gentry PR. Application of the adverse outcome pathway (AOP) approach to inform mode of action (MOA): A case study with inorganic arsenic. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2018; 81:893-912. [PMID: 30230972 DOI: 10.1080/15287394.2018.1500326] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/10/2018] [Accepted: 07/10/2018] [Indexed: 06/08/2023]
Abstract
The aim of this study was to establish a process for deriving a chemical-specific mode of action (MOA) from chemical-agnostic adverse outcome pathway (AOPs), using inorganic arsenic (iAs) as a case study. The AOP developed for this case study are related to disruption of cellular signaling by chemicals that strongly bind to vicinal dithiols in cellular proteins, leading to disruption of inflammatory and oxidative stress signaling along with inhibition of the DNA damage responses. The proposed MOA for iAs incorporates this AOP, overlaid on a background of increasing oxidative stress and/or co-exposure to mutagenic chemicals or radiation. The most challenging aspect of developing a MOA from AOP is the incorporation of metabolism and dose-response, neither of which may be considered in the development of an AOP. The cellular responses to relatively low concentrations (below 100 parts per billion) of iAs in drinking water appear to be secondary to binding of trivalent arsenite and its trivalent metabolite, monomethyl arsenous acid to key cellular vicinal dithiols in target tissues, resulting in a co-carcinogenic MOA. The proposed AOP may also be applied to non-cancer endpoints, enabling an integrated approach to conducting a risk assessment for iAs.
Collapse
|
47
|
Yu S, Liao WT, Lee CH, Chai CY, Yu CL, Yu HS. Immunological dysfunction in chronic arsenic exposure: From subclinical condition to skin cancer. J Dermatol 2018; 45:1271-1277. [DOI: 10.1111/1346-8138.14620] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 07/29/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Sebastian Yu
- Department of Dermatology; Kaohsiung Medical University Hospital; Kaohsiung Medical University; Kaohsiung Taiwan
- Department of Dermatology; College of Medicine; Kaohsiung Medical University; Kaohsiung Taiwan
- Department of Dermatology; University of California Davis School of Medicine; Sacramento California USA
| | - Wei-Ting Liao
- Department of Biotechnology; College of Life Science; Kaohsiung Medical University; Kaohsiung Taiwan
| | - Chih-Hung Lee
- Department of Dermatology; Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine; Kaohsiung Taiwan
| | - Chee-Yin Chai
- Department of Pathology; Kaohsiung Medical University Hospital; Kaohsiung Medical University; Kaohsiung Taiwan
| | - Chia-Li Yu
- Department of International Medicine; National Taiwan University Hospital; Taipei Taiwan
| | - Hsin-Su Yu
- Department of Dermatology; Kaohsiung Medical University Hospital; Kaohsiung Medical University; Kaohsiung Taiwan
- Department of Dermatology; College of Medicine; Kaohsiung Medical University; Kaohsiung Taiwan
- Graduate Institute of Clinical Medicine; College of Medicine; Kaohsiung Medical University; Kaohsiung Taiwan
| |
Collapse
|
48
|
Twaddle NC, Vanlandingham M, Beland FA, Doerge DR. Metabolism and disposition of arsenic species after repeated oral dosing with sodium arsenite in drinking water. II. Measurements in pregnant and fetal CD-1 mice. Food Chem Toxicol 2018. [PMID: 29530638 DOI: 10.1016/j.fct.2018.03.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Arsenic is ubiquitous in the earth's crust, and human diseases are linked with exposures that are similar to dietary intake estimates. Metabolic methylation of inorganic arsenic facilitates excretion of pentavalent metabolites and decreases acute toxicity; however, tissue binding of trivalent arsenic intermediates is evidence for concomitant metabolic activation. Pregnant and fetal CD-1 mice comprise a key animal model for arsenic carcinogenesis since adult-only exposures have minimal effects. This study evaluated inorganic arsenic and its metabolites in pentavalent and trivalent states in blood and tissues from maternal and fetal CD-1 mice after repeated administration of arsenite through drinking water. After 8 days of exposure, DMA species were ubiquitous in dams and fetuses. Despite the presence of MMAIII in dams, none was observed in any fetal sample. This difference may be important in assessing fetal susceptibility to arsenic toxicity because MMA production has been linked with human disease. Binding of DMAIII in fetal tissues provided evidence for metabolic activation, although the role for such binding in arsenic toxicity is unclear. This study provides links between administered dose, metabolism, and internal exposures from a key animal model of arsenic toxicity to better understand risks from human exposure to environmental arsenic.
Collapse
Affiliation(s)
- Nathan C Twaddle
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, United States
| | - Michelle Vanlandingham
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, United States
| | - Frederick A Beland
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, United States
| | - Daniel R Doerge
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, United States.
| |
Collapse
|
49
|
Murko M, Elek B, Styblo M, Thomas DJ, Francesconi KA. Dose and Diet - Sources of Arsenic Intake in Mouse in Utero Exposure Scenarios. Chem Res Toxicol 2018; 31:156-164. [PMID: 29244955 PMCID: PMC6611170 DOI: 10.1021/acs.chemrestox.7b00309] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
In humans, early life exposure to inorganic arsenic is associated with adverse health effects. Inorganic arsenic in utero or in early postnatal life also produces adverse health effects in offspring of pregnant mice that consumed drinking water containing low part per billion levels of inorganic arsenic. Because aggregate exposure of pregnant mice to inorganic arsenic from both drinking water and food has not been fully evaluated in experimental studies, quantifying arsenic exposure of the developing mouse is problematic. Here, we determined levels of total arsenic and arsenic species in natural ingredient rodent diets that are composed of many plant and animal-derived foodstuffs and in a purified ingredient rodent diet that is composed of a more restricted mixture of foodstuffs. In natural ingredient diets, total arsenic levels ranged from ∼60 to ∼400 parts per billion, and in the purified ingredient diet, total arsenic level was 13 parts per billion. Inorganic arsenic was the predominant arsenic species in trifluoroacetic acid extracts of each diet. Various exposure scenarios were evaluated using information on inorganic arsenic levels in diet and drinking water and on daily food and water consumption of pregnant mice. In a scenario in which pregnant mice consumed drinking water with 10 parts per billion of inorganic arsenic and a natural ingredient diet containing 89 parts per billion of inorganic arsenic, drinking water contributed only ∼20% of inorganic arsenic intake. Quantitation of arsenic species in diets used in studies in which drinking water is the nominal source of arsenic exposure provides more accurate dosimetry and improves understanding of dose-response relations. Use of purified ingredient diets will minimize the discrepancy between the target dosage level and the actual dosage level attained in utero exposure studies designed to evaluate effects of low level exposure to inorganic arsenic.
Collapse
Affiliation(s)
- Manuela Murko
- Institute of Chemistry, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Brittany Elek
- Pharmacokinetics Branch, Integrated Systems Toxicology Division, National Health and Environmental Effects Laboratory, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27709, United States
| | - Miroslav Styblo
- Department of Nutrition, Gillings School of Global Public Health, The University of North Carolina, Chapel Hill, North Carolina 27719, United States
| | - David J. Thomas
- Pharmacokinetics Branch, Integrated Systems Toxicology Division, National Health and Environmental Effects Laboratory, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27709, United States
| | | |
Collapse
|
50
|
|