1
|
Banerjee M, Lykoudi A, Hwang JY, Pan J, Rai SN, Park JW, States JC. Dysregulation of mRNA expression by hsa-miR-186 overexpression in arsenic-induced skin carcinogenesis. Toxicol Appl Pharmacol 2025; 495:117209. [PMID: 39719251 DOI: 10.1016/j.taap.2024.117209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 12/26/2024]
Abstract
Dysregulated miRNA expression contributes to development of arsenic-induced cutaneous squamous cell carcinoma (cSCC). hsa-miR-186 (miR-186) is overexpressed in arsenical cSCC tissues as well as in preclinical cell line model of arsenical cSCC. Simultaneous miR-186 overexpression and chronic inorganic trivalent arsenite (iAs; 100 nM) exposure transformed human HaCaT cell line preferentially over miR-186 overexpression or iAs exposure alone. Both iAs and miR-186 regulate the expression of wide range of mRNA targets. However, how their interaction impacts the transcriptome-wide mRNA expression landscape ushering in cancer is unknown. We performed longitudinal RNA-seq analysis in passage-matched HaCaT cell clones (±miR-186 overexpression) with simultaneous chronic iAs exposure (0/100 nM) at 12 and 29 weeks. We determined the impact of each factor and their interaction towards differential gene expression and pathway dysregulation employing two different statistical approaches (t-statistic and 2-factor ANOVA). We show that a core set of pathways are dysregulated deterministically irrespective of the statistical approach chosen, possibly representing necessary changes for transformation. The data suggest that each clonal line could take a unique route to dysregulate this core set of pathways necessary for transformation, highlighting the possible role of stochasticity in cancer development. Evidence is presented to sift the strengths and weaknesses of each statistical methodology in providing biological understanding of events that play crucial roles in carcinogenesis in large datasets with multiple contributing variables.
Collapse
Affiliation(s)
- Mayukh Banerjee
- Department of Pharmacology and Toxicology, University of Louisville, 505 S. Hancock Street, Louisville, KY 40202, USA; Center for Integrative Environmental Health Sciences, University of Louisville, 505 S. Hancock Street, Louisville, KY 40202, USA
| | - Angeliki Lykoudi
- Department of Pharmacology and Toxicology, University of Louisville, 505 S. Hancock Street, Louisville, KY 40202, USA
| | - Jae Y Hwang
- Center for Integrative Environmental Health Sciences, University of Louisville, 505 S. Hancock Street, Louisville, KY 40202, USA; Brown Cancer Center, University of Louisville, 505 S. Hancock Street, Louisville, KY 40202, USA; Department of Medicine, University of Louisville, 505 S. Hancock Street, Louisville, KY 40202, USA
| | - Jianmin Pan
- Center for Integrative Environmental Health Sciences, University of Louisville, 505 S. Hancock Street, Louisville, KY 40202, USA; Cancer Data Science Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Biostatistics and Informatics Shared Resources, University of Cincinnati Cancer Center, Cincinnati, OH, USA; Department of Biostatistics, Health Informatics and Data Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Department of Bioinformatics and Biostatistics, University of Louisville, 505 S. Hancock Street, Louisville, KY 40202, USA
| | - Shesh N Rai
- Center for Integrative Environmental Health Sciences, University of Louisville, 505 S. Hancock Street, Louisville, KY 40202, USA; Cancer Data Science Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Biostatistics and Informatics Shared Resources, University of Cincinnati Cancer Center, Cincinnati, OH, USA; Department of Biostatistics, Health Informatics and Data Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Department of Bioinformatics and Biostatistics, University of Louisville, 505 S. Hancock Street, Louisville, KY 40202, USA
| | - Juw W Park
- Center for Integrative Environmental Health Sciences, University of Louisville, 505 S. Hancock Street, Louisville, KY 40202, USA; Brown Cancer Center, University of Louisville, 505 S. Hancock Street, Louisville, KY 40202, USA; Department of Medicine, University of Louisville, 505 S. Hancock Street, Louisville, KY 40202, USA
| | - J Christopher States
- Department of Pharmacology and Toxicology, University of Louisville, 505 S. Hancock Street, Louisville, KY 40202, USA; Center for Integrative Environmental Health Sciences, University of Louisville, 505 S. Hancock Street, Louisville, KY 40202, USA.
| |
Collapse
|
2
|
Wang L, Qiu N, Tong S, Yu Y, Xi S, Wang F. Matrine Suppresses Arsenic-Induced Malignant Transformation of SV-HUC-1 Cells via NOX2. Int J Mol Sci 2024; 25:8878. [PMID: 39201564 PMCID: PMC11354282 DOI: 10.3390/ijms25168878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Arsenic (As) has been classified as a carcinogen for humans. There is abundant evidence indicating that arsenic increases the risk of bladder cancer among human populations. However, the underlying mechanisms have yet to be fully understood and elucidated. NADPH oxidases (NOXs) are the main enzymes for ROS production in the body. NADPH Oxidase 2 (NOX2), which is the most distinctive and ubiquitously expressed subunit of NOXs, can promote the formation and development of tumors. The utilization of NOX2 as a therapeutic target has been proposed to modulate diseases resulting from the activation of NOD-like receptor thermal protein domain associated protein 3 (NLRP3). Matrine has been reported to exhibit various pharmacological effects, including anti-inflammatory, antifibrotic, antitumor, and analgesic properties. However, it has not been reported whether matrine can inhibit malignant transformation induced by arsenic in uroepithelial cells through NOX2. We have conducted a series of experiments using both a sub-chronic NaAsO2 exposure rat model and a long-term NaAsO2 exposure cell model. Our findings indicate that arsenic significantly increases cell proliferation, migration, and angiogenesis in vivo and in vitro. Arsenic exposure resulted in an upregulation of reactive oxygen species (ROS), NOX2, and NLRP3 inflammasome expression. Remarkably, both in vivo and in vitro, the administration of matrine demonstrated a significant improvement in the detrimental impact of arsenic on bladder epithelial cells. This was evidenced by the downregulation of proliferation, migration, and angiogenesis, as well as the expression of the NOX2 and NLRP3 inflammasomes. Collectively, these findings indicate that matrine possesses the ability to reduce NOX2 levels and inhibit the transformation of bladder epithelial cells.
Collapse
Affiliation(s)
- Lanfei Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, China
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
- Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang 110122, China
| | - Nianfeng Qiu
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, China
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
- Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang 110122, China
| | - Suyuan Tong
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, China
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
- Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang 110122, China
| | - Yan Yu
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, China
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
- Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang 110122, China
| | - Shuhua Xi
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, China
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
- Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang 110122, China
| | - Fei Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, China
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
- Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang 110122, China
| |
Collapse
|
3
|
Shariat Razavi F, Kouchak M, Sistani Karampour N, Mahdavinia M, Nazari Khorasgani Z, Rezaie A, Rahbar N. AS1411aptamer conjugated liposomes for targeted delivery of arsenic trioxide in mouse xenograft model of melanoma cancer. J Liposome Res 2024; 34:288-302. [PMID: 37843918 DOI: 10.1080/08982104.2023.2271046] [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: 05/05/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023]
Abstract
Development of AS1411aptamer-conjugated liposomes for targeted delivery of arsenic trioxide is the primary goal of this study. AS1411aptamer was used as ligand to target nucleolin, which is highly expressed on the surface of melanoma cancer cells. The targeted liposomes were constructed by the thin film method, and arsenic trioxide was loaded as cobalt (II) hydrogen arsenite (CHA) to increase the loading efficiency and stability of the liposomes. The liposomal structure was characterized by Fourier Transform Infrared Spectroscopy (FT-IR) and field emission scanning electron microscopy (FESEM). In addition, particle sizes and zeta potential of the CHA-loaded liposomes (CHAL) and aptamer-functionalized CHA-loaded liposomes (AP-CHAL) were determined. In vitro cytotoxicity of CHAL and AP-CHAL were evaluated using MTT assay in murine melanoma (B16) and mouse embryonic fibroblast (MEF) cell lines. The encapsulation efficiency of CHAL and AP-CHAL was reported as 60.2 ± 6.5% and 58.7 ± 4.2%, respectively. In vivo antitumor activity of CHAL and AP-CHAL in the B16 tumor-xenograft mouse model was dramatically observed. All mice of both groups survived until the end of treatment and showed body weight gain. The tumor protrusion completely disappeared in 50% of the mice in these groups. Furthermore, histopathology studies demonstrated that CHAL and AP-CHAL did not induce significant toxicity in healthy mice tissues. However, unlike the CHAL group, which showed an initial increase in tumor volume, a specific antitumor effect was observed in the AP-CHAL group from the beginning of treatment. The results showed that AP-CHAL can be used as an effective drug delivery system with high potential in the treatment of solid tumors.
Collapse
Affiliation(s)
- Fatemeh Shariat Razavi
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Pharmaceutics, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Kouchak
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Pharmaceutics, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Neda Sistani Karampour
- Marine Pharmaceutical Science Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Pharmacology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masoud Mahdavinia
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Nazari Khorasgani
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Annahita Rezaie
- Department of Pathobiology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Nadereh Rahbar
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
4
|
Speer RM, Yu H, Zhou X, Nandi S, Alexandrov L, Guo Y, Hudson LG, Liu KJ. Arsenic and UVR co-exposure results in unique gene expression profile identifying key co-carcinogenic mechanisms. Toxicol Appl Pharmacol 2024; 482:116773. [PMID: 38036231 PMCID: PMC10883297 DOI: 10.1016/j.taap.2023.116773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
Changes in gene expression underlie many pathogenic endpoints including carcinogenesis. Metals, like arsenic, alter gene expression; however, the consequences of co-exposures of metals with other stressors are less understood. Although arsenic acts as a co-carcinogen by enhancing the development of UVR skin cancers, changes in gene expression in arsenic UVR co-carcinogenesis have not been investigated. We performed RNA-sequencing analysis to profile changes in gene expression distinct from arsenic or UVR exposures alone. A large number of differentially expressed genes (DEGs) were identified after arsenic exposure alone, while after UVR exposure alone fewer genes were changed. A distinct increase in the number of DEGs was identified after exposure to combined arsenic and UVR exposure that was synergistic rather than additive. In addition, a majority of these DEGs were unique from arsenic or UVR alone suggesting a distinct response to combined arsenic-UVR exposure. Globally, arsenic alone and arsenic plus UVR exposure caused a global downregulation of genes while fewer genes were upregulated. Gene Ontology analysis using the DEGs revealed cellular processes related to chromosome instability, cell cycle, cellular transformation, and signaling were targeted by combined arsenic and UVR exposure, distinct from UVR alone and arsenic alone, while others were related to epigenetic mechanisms such as the modification of histones. This result suggests the cellular functions we identified in this study may be key in understanding how arsenic enhances UVR carcinogenesis and that arsenic-enhanced gene expression changes may drive co-carcinogenesis of UVR exposure.
Collapse
Affiliation(s)
- Rachel M Speer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, USA.
| | - Hui Yu
- Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico, Albuquerque, NM, USA.
| | - Xixi Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, USA.
| | - Shuvro Nandi
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA; Moores Cancer Center, UC San Diego, La Jolla, CA 92037, USA.
| | - Ludmil Alexandrov
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA; Moores Cancer Center, UC San Diego, La Jolla, CA 92037, USA.
| | - Yan Guo
- Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico, Albuquerque, NM, USA.
| | - Laurie G Hudson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, USA.
| | - Ke Jian Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, USA; Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA; Department of Pathology, Stony Brook University School of Medicine, Stony Brook, NY 11794, USA.
| |
Collapse
|
5
|
Li C, Zhang A, Hu T, Yang Y, Tang S, Li J. Histone demethylase JHDM2A participates in the repair of arsenic-induced DNA damage in L-02 cells by regulating DDB2. Toxicol Ind Health 2022; 38:365-376. [PMID: 35579678 DOI: 10.1177/07482337221098319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Arsenic is widely present in nature and is a class I carcinogen confirmed by the World Health Organization and the International Agency for Research on Cancer. The liver is responsible for biotransformation in the body and is one of the major organs where arsenic accumulates in the body, but the mechanisms of arsenic-induced abnormal DNA damage repair pathways in the liver are still unclear. Recent studies have revealed that epigenetic mechanisms play an important role in arsenic-induced lesions. In this study, an in vitro model was established using human normal hepatocytes L-02 to investigate the mechanism of the specific demethylase JHDM2A of H3K9me2 in the repair of arsenic-induced DNA damage in L-02 cells. The results showed that with the increase of arsenic concentrations, the extent of DNA damage in L-02 cells showed an increasing trend and total intracellular H3K9me2 expression was downregulated. In addition, the enrichment level of H3K9me2 in the promoter region of DBB2, a key factor of nucleotide repair (NBR), increased, while protein and mRNA expression levels showed a decreasing trend. Thereafter, we overexpressed and repressed JHDM2A and found a close association between JHDM2A and arsenic-induced DNA damage. DDB2 protein and mRNA expression was downregulated with JHDM2A overexpression and upregulated with JHDM2A repression, while DBB2 promoter region H3K9me2 enrichment levels remained at a high level, although they were affected after JHDM2A overexpression or knockdown to some extent. These results suggest a potential mechanism by which JHDM2A may regulate DDB2 gene expression, participate in the NBR process, and play a role in arsenic-induced DNA damage in L-02 cells, which is not the result of JHDM2A exerting demethylation on H3K9me2 in the DDB2 promoter region. Our results provided an epigenetic mechanism for endemic arsenicosis, as well as a scientific basis for potential prevention and control measures.
Collapse
Affiliation(s)
- Changzhe Li
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education Department of Toxicology, School of Public Health, 74628Guizhou Medical University, Guiyang, China
| | - Anliu Zhang
- Guiyang Center for Disease Control and Prevention, Guiyang, China
| | - Ting Hu
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education Department of Toxicology, School of Public Health, 74628Guizhou Medical University, Guiyang, China
| | - Yue Yang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education Department of Toxicology, School of Public Health, 74628Guizhou Medical University, Guiyang, China
| | - Shunfang Tang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education Department of Toxicology, School of Public Health, 74628Guizhou Medical University, Guiyang, China
| | - Jun Li
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education Department of Toxicology, School of Public Health, 74628Guizhou Medical University, Guiyang, China
| |
Collapse
|
6
|
Ferragut Cardoso AP, Banerjee M, Al-Eryani L, Sayed M, Wilkey DW, Merchant ML, Park JW, States JC. Temporal Modulation of Differential Alternative Splicing in HaCaT Human Keratinocyte Cell Line Chronically Exposed to Arsenic for up to 28 Wk. ENVIRONMENTAL HEALTH PERSPECTIVES 2022; 130:17011. [PMID: 35072517 PMCID: PMC8785870 DOI: 10.1289/ehp9676] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 12/12/2021] [Accepted: 12/16/2021] [Indexed: 05/06/2023]
Abstract
BACKGROUND Chronic arsenic exposure via drinking water is associated with an increased risk of developing cancer and noncancer chronic diseases. Pre-mRNAs are often subject to alternative splicing, generating mRNA isoforms encoding functionally distinct protein isoforms. The resulting imbalance in isoform species can result in pathogenic changes in critical signaling pathways. Alternative splicing as a mechanism of arsenic-induced toxicity and carcinogenicity is understudied. OBJECTIVE This study aimed to accurately profile differential alternative splicing events in human keratinocytes induced by chronic arsenic exposure that might play a role in carcinogenesis. METHODS Independent quadruplicate cultures of immortalized human keratinocytes (HaCaT) were maintained continuously for 28 wk with 0 or 100 nM sodium arsenite. RNA-sequencing (RNA-Seq) was performed with poly(A) RNA isolated from cells harvested at 7, 19, and 28 wk with subsequent replicate multivariate analysis of transcript splicing (rMATS) analysis to detect and quantify differential alternative splicing events. Reverse transcriptase-polymerase chain reaction (RT-PCR) for selected alternative splicing events was performed to validate RNA-Seq predictions. Functional enrichment was performed by gene ontology (GO) analysis of the differential alternative splicing event data set at each time point. RESULTS At least 600 differential alternative splicing events were detected at each time point tested, comprising all the five main types of alternative splicing and occurring in both open reading frames (ORFs) and untranslated regions (UTRs). Based on functional relevance ELK4, SHC1, and XRRA1 were selected for validation of predicted alternative splicing events at 7 wk by RT-PCR. Densitometric analysis of RT-PCR data corroborated the rMATS predicted alternative splicing for all three events. Protein expression validation of the selected alternative splicing events was challenging given that very few isoform-specific antibodies are available. GO analysis demonstrated that the enriched terms in differential alternatively spliced mRNAs changed dynamically with the time of exposure. Notably, RNA metabolism and splicing regulation pathways were enriched at the 7-wk time point, when the greatest number of differentially alternatively spliced mRNAs are detected. Our preliminary proteomic analysis demonstrated that the expression of the canonical isoforms of the splice regulators DDX42, RMB25, and SRRM2 were induced upon chronic arsenic exposure, corroborating the splicing predictions. DISCUSSION These results using cultures of HaCaT cells suggest that arsenic exposure disrupted an alternative splice factor network and induced time-dependent genome-wide differential alternative splicing that likely contributed to the changing proteomic landscape in arsenic-induced carcinogenesis. However, significant challenges remain in corroborating alternative splicing data at the proteomic level. https://doi.org/10.1289/EHP9676.
Collapse
Affiliation(s)
- Ana P. Ferragut Cardoso
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| | - Mayukh Banerjee
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| | - Laila Al-Eryani
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| | - Mohammed Sayed
- Computer Science and Engineering, University of Louisville, Louisville, Kentucky, USA
| | - Daniel W. Wilkey
- Division of Nephrology & Hypertension, Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Michael L. Merchant
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
- Division of Nephrology & Hypertension, Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Juw W. Park
- Computer Science and Engineering, University of Louisville, Louisville, Kentucky, USA
- KY INBRE Bioinformatics Core, University of Louisville, Louisville, Kentucky, USA
| | - J. Christopher States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
7
|
Li S, Zhou Q, Liu W, Fu Z, Zhao H, Xi S. Targeting SLC1A5 blocks cell proliferation through inhibition of mTORC1 in arsenite-treated human uroepithelial cells. Toxicol Lett 2021; 345:1-11. [PMID: 33781819 DOI: 10.1016/j.toxlet.2021.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/14/2021] [Accepted: 03/24/2021] [Indexed: 10/21/2022]
Abstract
Arsenic is an environmental contaminant, which is widely distributed in soil, air, and water. There is sufficient evidence to indicate that arsenic increases the risk of bladder cancer in humans. However, its underlying mechanisms remain elusive. Glutamine (Gln) has multiple functions that promote carcinogenesis. Indeed, Gln transporters on cancer cells surface are often upregulated. Elevated expression levels of Alanine, serine, cysteine-preferring transporter 2 (ASCT2; SLC1A5) have been reported in many types of human tumors. This study characterized the role of SLC1A5 in cell proliferation in arsenite-treated cells. In short-term experiments, SV-40 immortalized human uroepithelial (SV-HUC-1) cells were treated with Sodium arsenite (NaAsO2) (0, 0.5, 1, 2, 4, 8 μM) for 24 h. In long-term experiments, SV-HUC-1 cells were exposed to 0.5 μM NaAsO2 for 40 weeks. In both short-term and long-term experiments, arsenite increased expression of SLC1A5 by 1.89-fold and 2.25-fold, respectively. Arsenite increased Gln consumption of SV-HUC-1 cells, and Gln starvation inhibited cell proliferation in long-term arsenite-treated cells. Importantly, inhibiting SLC1A5 blocked cell proliferation by downregulating mTORC1 in long-term arsenite-treated cells. Moreover, SLC1A5 regulated mTORC1 in an αKG-dependent manner. Our results suggest that SLC1A5 plays an important role in cell proliferation of arsenite-treated SV-HUC-1 cells.
Collapse
Affiliation(s)
- Sihao Li
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, China.
| | - Qing Zhou
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, China.
| | - Weijue Liu
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, China.
| | - Zhushan Fu
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, China.
| | - Hanqing Zhao
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, China.
| | - Shuhua Xi
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, China.
| |
Collapse
|
8
|
Hartwig A, Arand M, Epe B, Guth S, Jahnke G, Lampen A, Martus HJ, Monien B, Rietjens IMCM, Schmitz-Spanke S, Schriever-Schwemmer G, Steinberg P, Eisenbrand G. Mode of action-based risk assessment of genotoxic carcinogens. Arch Toxicol 2020; 94:1787-1877. [PMID: 32542409 PMCID: PMC7303094 DOI: 10.1007/s00204-020-02733-2] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 03/31/2020] [Indexed: 12/16/2022]
Abstract
The risk assessment of chemical carcinogens is one major task in toxicology. Even though exposure has been mitigated effectively during the last decades, low levels of carcinogenic substances in food and at the workplace are still present and often not completely avoidable. The distinction between genotoxic and non-genotoxic carcinogens has traditionally been regarded as particularly relevant for risk assessment, with the assumption of the existence of no-effect concentrations (threshold levels) in case of the latter group. In contrast, genotoxic carcinogens, their metabolic precursors and DNA reactive metabolites are considered to represent risk factors at all concentrations since even one or a few DNA lesions may in principle result in mutations and, thus, increase tumour risk. Within the current document, an updated risk evaluation for genotoxic carcinogens is proposed, based on mechanistic knowledge regarding the substance (group) under investigation, and taking into account recent improvements in analytical techniques used to quantify DNA lesions and mutations as well as "omics" approaches. Furthermore, wherever possible and appropriate, special attention is given to the integration of background levels of the same or comparable DNA lesions. Within part A, fundamental considerations highlight the terms hazard and risk with respect to DNA reactivity of genotoxic agents, as compared to non-genotoxic agents. Also, current methodologies used in genetic toxicology as well as in dosimetry of exposure are described. Special focus is given on the elucidation of modes of action (MOA) and on the relation between DNA damage and cancer risk. Part B addresses specific examples of genotoxic carcinogens, including those humans are exposed to exogenously and endogenously, such as formaldehyde, acetaldehyde and the corresponding alcohols as well as some alkylating agents, ethylene oxide, and acrylamide, but also examples resulting from exogenous sources like aflatoxin B1, allylalkoxybenzenes, 2-amino-3,8-dimethylimidazo[4,5-f] quinoxaline (MeIQx), benzo[a]pyrene and pyrrolizidine alkaloids. Additionally, special attention is given to some carcinogenic metal compounds, which are considered indirect genotoxins, by accelerating mutagenicity via interactions with the cellular response to DNA damage even at low exposure conditions. Part C finally encompasses conclusions and perspectives, suggesting a refined strategy for the assessment of the carcinogenic risk associated with an exposure to genotoxic compounds and addressing research needs.
Collapse
Affiliation(s)
- Andrea Hartwig
- Department of Food Chemistry and Toxicology, Institute of Applied Biosciences (IAB), Karlsruhe Institute of Technology (KIT), Adenauerring 20a, 76131, Karlsruhe, Germany.
| | - Michael Arand
- Institute of Pharmacology and Toxicology, University of Zurich, 8057, Zurich, Switzerland
| | - Bernd Epe
- Institute of Pharmacy and Biochemistry, University of Mainz, 55099, Mainz, Germany
| | - Sabine Guth
- Department of Toxicology, IfADo-Leibniz Research Centre for Working Environment and Human Factors, TU Dortmund, Ardeystr. 67, 44139, Dortmund, Germany
| | - Gunnar Jahnke
- Department of Food Chemistry and Toxicology, Institute of Applied Biosciences (IAB), Karlsruhe Institute of Technology (KIT), Adenauerring 20a, 76131, Karlsruhe, Germany
| | - Alfonso Lampen
- Department of Food Safety, German Federal Institute for Risk Assessment (BfR), 10589, Berlin, Germany
| | - Hans-Jörg Martus
- Novartis Institutes for BioMedical Research, 4002, Basel, Switzerland
| | - Bernhard Monien
- Department of Food Safety, German Federal Institute for Risk Assessment (BfR), 10589, Berlin, Germany
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Simone Schmitz-Spanke
- Institute and Outpatient Clinic of Occupational, Social and Environmental Medicine, University of Erlangen-Nuremberg, Henkestr. 9-11, 91054, Erlangen, Germany
| | - Gerlinde Schriever-Schwemmer
- Department of Food Chemistry and Toxicology, Institute of Applied Biosciences (IAB), Karlsruhe Institute of Technology (KIT), Adenauerring 20a, 76131, Karlsruhe, Germany
| | - Pablo Steinberg
- Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Haid-und-Neu-Str. 9, 76131, Karlsruhe, Germany
| | - Gerhard Eisenbrand
- Retired Senior Professor for Food Chemistry and Toxicology, Kühler Grund 48/1, 69126, Heidelberg, Germany.
| |
Collapse
|
9
|
Tchounwou PB, Yedjou CG, Udensi UK, Pacurari M, Stevens JJ, Patlolla AK, Noubissi F, Kumar S. State of the science review of the health effects of inorganic arsenic: Perspectives for future research. ENVIRONMENTAL TOXICOLOGY 2019; 34:188-202. [PMID: 30511785 PMCID: PMC6328315 DOI: 10.1002/tox.22673] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/06/2018] [Accepted: 10/09/2018] [Indexed: 05/06/2023]
Abstract
Human exposure to inorganic arsenic (iAs) is a global health issue. Although there is strong evidence for iAs-induced toxicity at higher levels of exposure, many epidemiological studies evaluating its effects at low exposure levels have reported mixed results. We comprehensively reviewed the literature and evaluated the scientific knowledge on human exposure to arsenic, mechanisms of action, systemic and carcinogenic effects, risk characterization, and regulatory guidelines. We identified areas where additional research is needed. These priority areas include: (1) further development of animal models of iAs carcinogenicity to identify molecular events involved in iAs carcinogenicity; (2) characterization of underlying mechanisms of iAs toxicity; (3) assessment of gender-specific susceptibilities and other factors that modulate arsenic metabolism; (4) sufficiently powered epidemiological studies to ascertain relationship between iAs exposure and reproductive/developmental effects; (5) evaluation of genetic/epigenetic determinants of iAs effects in children; and (6) epidemiological studies of people chronically exposed to low iAs concentrations.
Collapse
Affiliation(s)
- Paul B. Tchounwou
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD-RCMI Center for Environmental Health.Jackson State University, 1400 Lynch Street, Box18750, Jackson, Mississippi, MS 39217, USA
- Department of Biology, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box18750, Jackson, Mississippi, MS 39217, USA
| | - Clement G. Yedjou
- Department of Biology, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box18750, Jackson, Mississippi, MS 39217, USA
| | - Udensi K. Udensi
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD-RCMI Center for Environmental Health.Jackson State University, 1400 Lynch Street, Box18750, Jackson, Mississippi, MS 39217, USA
| | - Maricica Pacurari
- Department of Biology, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box18750, Jackson, Mississippi, MS 39217, USA
| | - Jacqueline J. Stevens
- Department of Biology, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box18750, Jackson, Mississippi, MS 39217, USA
| | - Anita K. Patlolla
- Department of Biology, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box18750, Jackson, Mississippi, MS 39217, USA
| | - Felicite Noubissi
- Department of Biology, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box18750, Jackson, Mississippi, MS 39217, USA
| | - Sanjay Kumar
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD-RCMI Center for Environmental Health.Jackson State University, 1400 Lynch Street, Box18750, Jackson, Mississippi, MS 39217, USA
| |
Collapse
|
10
|
Samavarchi Tehrani S, Mahmoodzadeh Hosseini H, Yousefi T, Abolghasemi M, Qujeq D, Maniati M, Amani J. The crosstalk between trace elements with DNA damage response, repair, and oxidative stress in cancer. J Cell Biochem 2019; 120:1080-1105. [PMID: 30378148 DOI: 10.1002/jcb.27617] [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] [Received: 07/24/2018] [Accepted: 08/14/2018] [Indexed: 01/24/2023]
Abstract
DNA damage response (DDR) is a regulatory system responsible for maintaining genome integrity and stability, which can sense and transduce DNA damage signals. The severity of damage appears to determine DDRs, which can include damage repair, cell-cycle arrest, and apoptosis. Furthermore, defective components in DNA damage and repair machinery are an underlying cause for the development and progression of various types of cancers. Increasing evidence indicates that there is an association between trace elements and DDR/repair mechanisms. In fact, trace elements seem to affect mediators of DDR. Besides, it has been revealed that oxidative stress (OS) and trace elements are associated with cancer development. In this review, we discuss the role of some critical trace elements in the risk of cancer. In addition, we provide a brief introduction on DDR and OS in cancer. Finally, we will further review the interactions between some important trace elements including selenium, zinc, chromium, cadmium, and arsenic, and DDR, and OS in cancer.
Collapse
Affiliation(s)
- Sadra Samavarchi Tehrani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hamideh Mahmoodzadeh Hosseini
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Tooba Yousefi
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Maryam Abolghasemi
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Durdi Qujeq
- Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
| | - Mahmood Maniati
- English Department, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Li Y, He M, Chen B, Hu B. Inhibition of arsenite methylation induces synergistic genotoxicity of arsenite and benzo(a)pyrene diol epoxide in SCC-7 cells. Metallomics 2019; 11:176-182. [DOI: 10.1039/c8mt00217g] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A comprehensive analytical method was developed to investigate the synergistic genotoxicity of BPDE and As(iii) in SCC-7 cells.
Collapse
Affiliation(s)
- Youxian Li
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education)
- Department of Chemistry
- Wuhan University
- Wuhan 430072
- P. R. China
| | - Man He
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education)
- Department of Chemistry
- Wuhan University
- Wuhan 430072
- P. R. China
| | - Beibei Chen
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education)
- Department of Chemistry
- Wuhan University
- Wuhan 430072
- P. R. China
| | - Bin Hu
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education)
- Department of Chemistry
- Wuhan University
- Wuhan 430072
- P. R. China
| |
Collapse
|
12
|
Koutros S, Baris D, Waddell R, Beane Freeman LE, Colt JS, Schwenn M, Johnson A, Ward MH, Hosain GM, Moore LE, Stolzenberg-Solomon R, Rothman N, Karagas MR, Silverman DT. Potential effect modifiers of the arsenic-bladder cancer risk relationship. Int J Cancer 2018; 143:2640-2646. [PMID: 29981168 PMCID: PMC6235710 DOI: 10.1002/ijc.31720] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 05/15/2018] [Accepted: 06/08/2018] [Indexed: 12/30/2022]
Abstract
Populations exposed to arsenic in drinking water have an increased bladder cancer risk and evidence suggests that several factors may modify arsenic metabolism, influencing disease risk. We evaluated whether the association between cumulative lifetime arsenic exposure from drinking water and bladder cancer risk was modified by factors that may impact arsenic metabolism in a population-based case-control study of 1,213 cases and 1,418 controls. Unconditional logistic regression was used to calculate odds ratios (ORs) and 95% confidence intervals (CIs) for the association between cumulative arsenic intake and bladder cancer stratified by age, sex, smoking status, body mass index (BMI), alcohol consumption and folate intake. P-values for interaction were computed using a likelihood ratio test. We observed no statistically significant multiplicative interactions although some variations in associations were notable across risk factors, particularly for smoking and BMI. Among former smokers and current smokers, those with the highest cumulative arsenic intake had elevated risks of bladder cancer (OR = 1.4, 95% CI: 0.96-2.0 and OR = 1.6, 95% CI: 0.91-3.0, respectively; while the OR among never smokers was 1.1, 95% CI: 0.6-1.9, p-interaction = 0.49). Among those classified as normal or overweight based on usual adult BMI, the highest level of cumulative arsenic intake was associated with elevated risks of bladder cancer (OR = 1.3, 95% CI: 0.89-2.0 and OR = 1.6, 95% CI: 1.1-2.4, respectively), while risk was not elevated among those who were obese (OR = 0.9, 95% CI: 0.4-1.8) (p-interaction = 0.14). Our study provides some limited evidence of modifying roles of age, sex, smoking, BMI, folate and alcohol on arsenic-related bladder cancer risk that requires confirmation in other, larger studies.
Collapse
Affiliation(s)
- Stella Koutros
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD
| | - Dalsu Baris
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD
| | - Richard Waddell
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Laura E Beane Freeman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD
| | - Joanne S Colt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD
| | | | | | - Mary H Ward
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD
| | | | - Lee E Moore
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD
| | - Rachael Stolzenberg-Solomon
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD
| | - Margaret R Karagas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Debra T Silverman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD
| |
Collapse
|
13
|
Zhou Q, Jin P, Liu J, Wang F, Xi S. HER2 and Src co-regulate proliferation, migration and transformation by downstream signaling pathways in arsenite-treated human uroepithelial cells. Metallomics 2018; 10:1141-1159. [PMID: 30066004 DOI: 10.1039/c8mt00131f] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
Epidemiological studies have established a strong association between arsenic exposure in drinking water and an increased incidence of bladder cancer in arseniasis-endemic areas. Increased expression of HER2 has been observed in various types of human malignancies including bladder cancer. This study investigated the role of HER2 in arsenite-induced transformation of uroepithelial cells SV-HUC-1 and the role of Src family kinases in HER2 signaling. We found that the expression HER2 and Src were increased following chronic arsenite exposure in a time-dependent fashion. Chronic arsenite exposure also led to an upregulation of proliferation factors such as cyclin D1, COX2, PCNA, VEGF, and HIF-1α. Furthermore, Ras/Raf/MAPK, PI3K/AKT, and JAK2/STAT3 signaling pathways were activated by arsenite treatment. Importantly, these changes were inhibited by HER2 inhibitors and in HER2 knocked down cells. In addition, downregulation of HER2 inhibited cell growth and migration properties of arsenite-treated cells. Inhibition of Src also inhibits activation of signaling pathways and malignant transformation of cells. And we obtained evidence of an interaction between HER2 and Src in SV-HUC-1 cell lines. These results suggest that HER2 and Src may play an important role in arsenite-induced transformation by multiple downstream signals pathways.
Collapse
Affiliation(s)
- Qing Zhou
- Department of Environmental and Occupational Health, Liaoning Provincial Key Laboratory of Arsenic Biological Effect and Poisoning, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, P. R. China.
| | | | | | | | | |
Collapse
|
14
|
Monomethylarsonous acid: Induction of DNA damage and oxidative stress in mouse natural killer cells at environmentally-relevant concentrations. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2018; 832-833:1-6. [DOI: 10.1016/j.mrgentox.2018.05.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/21/2018] [Accepted: 05/21/2018] [Indexed: 12/11/2022]
|
15
|
Genotoxicity evaluation of multi-component mixtures of polyaromatic hydrocarbons (PAHs), arsenic, cadmium, and lead using flow cytometry based micronucleus test in HepG2 cells. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2018; 827:9-18. [DOI: 10.1016/j.mrgentox.2018.01.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 12/16/2017] [Accepted: 01/09/2018] [Indexed: 01/09/2023]
|
16
|
Shi Q, Fijten RR, Spina D, Riffo Vasquez Y, Arlt VM, Godschalk RW, Van Schooten FJ. Altered gene expression profiles in the lungs of benzo[a]pyrene-exposed mice in the presence of lipopolysaccharide-induced pulmonary inflammation. Toxicol Appl Pharmacol 2017; 336:8-19. [PMID: 28987381 PMCID: PMC5703654 DOI: 10.1016/j.taap.2017.09.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 09/22/2017] [Accepted: 09/29/2017] [Indexed: 12/13/2022]
Abstract
Patients with inflammatory lung diseases are often additionally exposed to polycyclic aromatic hydrocarbons like B[a]P and B[a]P-induced alterations in gene expression in these patients may contribute to the development of lung cancer. Mice were intra-nasally treated with lipopolysaccharide (LPS, 20μg/mouse) to induce pulmonary inflammation and subsequently exposed to B[a]P (0.5mg/mouse) by intratracheal instillation. Gene expression changes were analyzed in mouse lungs by RNA microarrays. Analysis of genes that are known to be involved in the cellular response to B[a]P indicated that LPS significantly inhibited gene expression of various enzymes linked to B[a]P metabolism, which was confirmed by phenotypic analyses of enzyme activity. Ultimately, these changes resulted in higher levels of B[a]P-DNA adducts in the lungs of mice exposed to B[a]P with prior LPS treatment compared to the lungs of mice exposed to B[a]P alone. Using principle component analysis (PCA), we found that of all the genes that were significantly altered in their expression, those that were able to separate the different exposure conditions were predominantly related to immune-response. Moreover, an overall analysis of differentially expressed genes indicated that cell-cell adhesion and cell-cell communication was inhibited in lungs of mice that received both B[a]P and LPS. Our results indicate that pulmonary inflammation increased the genotoxicity of B[a]P via inhibition of both phase I and II metabolism. Therefore, inflammation could be a critical contributor to B[a]P-induced carcinogenesis in humans.
Collapse
Affiliation(s)
- Q Shi
- Department of Toxicology & Pharmacology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands
| | - R R Fijten
- Department of Toxicology & Pharmacology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands
| | - D Spina
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Y Riffo Vasquez
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - V M Arlt
- Analytical and Environmental Sciences Division, MRC-PHE Centre for Environmental & Health, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - R W Godschalk
- Department of Toxicology & Pharmacology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands.
| | - F J Van Schooten
- Department of Toxicology & Pharmacology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands
| |
Collapse
|
17
|
Impaired autophagic flux and p62-mediated EMT are involved in arsenite-induced transformation of L-02 cells. Toxicol Appl Pharmacol 2017; 334:75-87. [PMID: 28888487 DOI: 10.1016/j.taap.2017.09.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 08/29/2017] [Accepted: 09/05/2017] [Indexed: 12/30/2022]
Abstract
Autophagy is a catabolic process essential for preserving cellular homeostasis, and the epithelial-to-mesenchymal transition (EMT) is involved during tissue development and cancer progression. In arsenite-treated human hepatic epithelial (L-02) cells, arsenite reduced the autophagic flux, which caused accumulation of p62, an adaptor and receptor of autophagy. Further, in arsenite-transformed L-02 cells, the levels of E-cadherin were attenuated, but the levels of vimentin, which is expressed in mesenchymal cells, and Snail, a transcription regulator of the EMT, were up-regulated. Thus, after chronic exposure of L-02 cells to arsenite, the impaired autophagic flux induced the accumulation of p62, which up-regulated the expression of Snail, a protein involved in arsenite-induced EMT of these cells. Knockdown of p62 by siRNA reversed the arsenite-induced EMT and decreased the capacities of arsenite-transformed L-02 cells for colony formation and invasion and migration. Therefore, in arsenite-induced transformation of L-02 cells, the accumulation of p62, by impairing autophagic flux, mediates the EMT via Snail. These results provide a previously unknown mechanism underlying arsenic toxicity and carcinogenicity.
Collapse
|
18
|
Khairul I, Wang QQ, Jiang YH, Wang C, Naranmandura H. Metabolism, toxicity and anticancer activities of arsenic compounds. Oncotarget 2017; 8:23905-23926. [PMID: 28108741 PMCID: PMC5410354 DOI: 10.18632/oncotarget.14733] [Citation(s) in RCA: 191] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/11/2017] [Indexed: 01/17/2023] Open
Abstract
A variety of studies indicated that inorganic arsenic and its methylated metabolites have paradoxical effects, namely, carcinogenic and anticancer effects. Epidemiological studies have shown that long term exposure to arsenic can increase the risk of cancers of lung, skin or bladder in man, which is probably associated with the arsenic metabolism. In fact, the enzymatic conversion of inorganic arsenic by Arsenic (+3 oxidation state) methyltransferase (AS3MT) to mono- and dimethylated arsenic species has long been considered as a major route for detoxification. However, several studies have also indicated that biomethylation of inorganic arsenic, particularly the production of trivalent methylated metabolites, is a process that activates arsenic as a toxin and a carcinogen. On the other hand, arsenic trioxide (As2O3) has recently been recognized as one of the most effective drugs for the treatment of APL. However, elaboration of the cytotoxic mechanisms of arsenic and its methylated metabolites in eradicating cancer is sorely lacking. To provide a deeper understanding of the toxicity and carcinogenicity along with them use of arsenic in chemotherapy, caution is required considering the poor understanding of its various mechanisms of exerting toxicity. Thereby, in this review, we have focused on arsenic metabolic pathway, the roles of the methylated arsenic metabolites in toxicity and in the therapeutic efficacy for the treatments of solid tumors, APL and/or non-APL malignancies.
Collapse
Affiliation(s)
- Islam Khairul
- Department of Toxicology, School of Medicine and Public Health, Zhejiang University, Hangzhou, China
| | - Qian Qian Wang
- Department of Toxicology, School of Medicine and Public Health, Zhejiang University, Hangzhou, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yu Han Jiang
- Department of Toxicology, School of Medicine and Public Health, Zhejiang University, Hangzhou, China
- Ocean College, Zhejiang University, Hangzhou, China
| | - Chao Wang
- Department of Toxicology, School of Medicine and Public Health, Zhejiang University, Hangzhou, China
| | - Hua Naranmandura
- Department of Toxicology, School of Medicine and Public Health, Zhejiang University, Hangzhou, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Ocean College, Zhejiang University, Hangzhou, China
| |
Collapse
|
19
|
Holcomb N, Goswami M, Han SG, Scott T, D'Orazio J, Orren DK, Gairola CG, Mellon I. Inorganic arsenic inhibits the nucleotide excision repair pathway and reduces the expression of XPC. DNA Repair (Amst) 2017; 52:70-80. [PMID: 28237621 DOI: 10.1016/j.dnarep.2017.02.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 01/11/2017] [Accepted: 02/12/2017] [Indexed: 11/17/2022]
Abstract
Chronic exposure to arsenic, most often through contaminated drinking water, has been linked to several types of cancer in humans, including skin and lung cancer. However, the mechanisms underlying its role in causing cancer are not well understood. There is evidence that exposure to arsenic can enhance the carcinogenicity of UV light in inducing skin cancers and may enhance the carcinogenicity of tobacco smoke in inducing lung cancers. The nucleotide excision repair (NER) pathway removes different types of DNA damage including those produced by UV light and components of tobacco smoke. The aim of the present study was to investigate the effect of sodium arsenite on the NER pathway in human lung fibroblasts (IMR-90 cells) and primary mouse keratinocytes. To measure NER, we employed a slot-blot assay to quantify the introduction and removal of UV light-induced 6-4 photoproducts (6-4 PP) and cyclobutane pyrimidine dimers (CPDs). We find a concentration-dependent inhibition of the removal of 6-4 PPs and CPDs in both cell types treated with arsenite. Treatment of both cell types with arsenite resulted in a significant reduction in the abundance of XPC, a protein that is critical for DNA damage recognition in NER. The abundance of RNA expressed from several key NER genes was also significantly reduced by treatment of IMR-90 cells with arsenite. Finally, treatment of IMR-90 cells with MG-132 abrogated the reduction in XPC protein, suggesting an involvement of the proteasome in the reduction of XPC protein produced by treatment of cells with arsenic. The inhibition of NER by arsenic may reflect one mechanism underlying the role of arsenic exposure in enhancing cigarette smoke-induced lung carcinogenesis and UV light-induced skin cancer, and it may provide some insights into the emergence of arsenic trioxide as a chemotherapeutic agent.
Collapse
Affiliation(s)
- Nathaniel Holcomb
- Department of Toxicology and Cancer Biology, The Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | - Mamta Goswami
- Department of Toxicology and Cancer Biology, The Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | - Sung Gu Han
- Toxicology Laboratory, Department of Food Science and Biotechnology of Animal Resources, College of Animal Bioscience and Technology, Konkuk University, Seoul 05029, Republic of Korea
| | - Tim Scott
- Department of Toxicology and Cancer Biology, The Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | - John D'Orazio
- Department of Toxicology and Cancer Biology, The Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | - David K Orren
- Department of Toxicology and Cancer Biology, The Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | - C Gary Gairola
- Department of Toxicology and Cancer Biology, The Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | - Isabel Mellon
- Department of Toxicology and Cancer Biology, The Markey Cancer Center, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
20
|
Luo F, Sun B, Li H, Xu Y, Liu Y, Liu X, Lu L, Li J, Wang Q, Wei S, Shi L, Lu X, Liu Q, Zhang A. A MALAT1/HIF-2α feedback loop contributes to arsenite carcinogenesis. Oncotarget 2016; 7:5769-87. [PMID: 26735578 PMCID: PMC4868720 DOI: 10.18632/oncotarget.6806] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 12/05/2015] [Indexed: 02/07/2023] Open
Abstract
Arsenic is well established as a human carcinogen, but the molecular mechanisms leading to arsenic-induced carcinogenesis are complex and elusive. It is also not known if lncRNAs are involved in arsenic-induced liver carcinogenesis. We have found that MALAT1, a non-coding RNA, is over-expressed in the sera of people exposed to arsenite and in hepatocellular carcinomas (HCCs), and MALAT1 has a close relation with the clinicopathological characteristics of HCC. In addition, hypoxia-inducible factor (HIF)-2α is up-regulated in HCCs, and MALAT1 and HIF-2α have a positive correlation in HCC tissues. During the malignant transformation of human hepatic epithelial (L-02) cells induced by a low concentration (2.0 μM) of arsenite, MALAT1 and HIF-2α are increased. In addition, arsenite-induced MALAT1 causes disassociation of the von Hippel-Lindau (VHL) protein from HIF-2α, therefore, alleviating VHL-mediated HIF-2α ubiquitination, which causes HIF-2α accumulation. In turn, HIF-2α transcriptionally regulates MALAT1, thus forming a positive feedback loop to ensure expression of arsenite-induced MALAT1 and HIF-2α, which are involved in malignant transformation. Moreover, MALAT1 and HIF-2α promote the invasive and metastatic capacities of arsenite-induced transformed L-02 cells and in HCC-LM3 cells. The capacities of MALAT1 and HIF-2α to promote tumor growth are validated in mouse xenograft models. In mice, arsenite induces an inflammatory response, and MALAT1 and HIF-2α are over-expressed. Together, these findings suggest that the MALAT1/HIF-2α feedback loop is involved in regulation of arsenite-induced malignant transformation. Our results not only confirm a novel mechanism involving reciprocal regulation between MALAT1 and HIF-2α, but also expand the understanding of the carcinogenic potential of arsenite.
Collapse
Affiliation(s)
- Fei Luo
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China.,The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Baofei Sun
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guiyang Medical University, Guiyang 550025, Guizhou, People's Republic of China
| | - Huiqiao Li
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Yuan Xu
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China.,The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China.,Thoracic and GI Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Yi Liu
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China.,The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Xinlu Liu
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China.,The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Lu Lu
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China.,The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Jun Li
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guiyang Medical University, Guiyang 550025, Guizhou, People's Republic of China
| | - Qingling Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guiyang Medical University, Guiyang 550025, Guizhou, People's Republic of China
| | - Shaofeng Wei
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guiyang Medical University, Guiyang 550025, Guizhou, People's Republic of China
| | - Le Shi
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China.,The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Xiaolin Lu
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China.,The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Qizhan Liu
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China.,The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guiyang Medical University, Guiyang 550025, Guizhou, People's Republic of China
| |
Collapse
|
21
|
Ganapathy S, Li P, Fagman J, Yu T, Lafontant J, Zhang G, Chen C. Low doses of arsenic, via perturbing p53, promotes tumorigenesis. Toxicol Appl Pharmacol 2016; 306:98-104. [DOI: 10.1016/j.taap.2016.07.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 07/12/2016] [Accepted: 07/13/2016] [Indexed: 11/27/2022]
|
22
|
Zhang A, Li H, Xiao Y, Chen L, Zhu X, Li J, Ma L, Pan X, Chen W, He Z. Aberrant methylation of nucleotide excision repair genes is associated with chronic arsenic poisoning. Biomarkers 2016; 22:429-438. [PMID: 27685703 DOI: 10.1080/1354750x.2016.1217933] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
OBJECTIVE To define whether aberrant methylation of DNA repair genes is associated with chronic arsenic poisoning. METHODS Hundred and two endemic arsenicosis patients and 36 healthy subjects were recruited. Methylight and bisulfite sequencing (BSP) assays were used to examine the methylation status of ERCC1, ERCC2 and XPC genes in peripheral blood lymphocytes (PBLs) and skin lesions of arsenicosis patients and NaAsO2-treated HaCaT cells. RESULTS Hypermethylation of ERCC1 and ERCC2 and suppressed gene expression were found in PBLs and skin lesions of arsenicosis patients and was correlated with the level of arsenic exposure. Particularly, the expression of ERCC1 and ERCC2 was associated with the severity of skin lesions. In vitro studies revealed an induction of ERCC2 hypermethylation and decreased mRNA expression in response to NaAsO2 treatment. CONCLUSION Hypermethylation of ERCC1 and ERCC2 and concomitant suppression of gene expression might be served as the epigenetic marks associated with arsenic exposure and adverse health effects.
Collapse
Affiliation(s)
- Aihua Zhang
- a The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education Department of Toxicology , School of Public Health, Guizhou Medical University , Guiyang , China
| | - Huiyao Li
- b Department of Toxicology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment , School of Public Health, Sun Yat-Sen University , Guangzhou , China
| | - Yun Xiao
- a The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education Department of Toxicology , School of Public Health, Guizhou Medical University , Guiyang , China
| | - Liping Chen
- b Department of Toxicology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment , School of Public Health, Sun Yat-Sen University , Guangzhou , China
| | - Xiaonian Zhu
- c Department of Toxicology, School of Public Health , Guilin Medical University , Guilin , China
| | - Jun Li
- a The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education Department of Toxicology , School of Public Health, Guizhou Medical University , Guiyang , China
| | - Lu Ma
- a The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education Department of Toxicology , School of Public Health, Guizhou Medical University , Guiyang , China
| | - Xueli Pan
- a The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education Department of Toxicology , School of Public Health, Guizhou Medical University , Guiyang , China
| | - Wen Chen
- b Department of Toxicology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment , School of Public Health, Sun Yat-Sen University , Guangzhou , China
| | - Zhini He
- b Department of Toxicology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment , School of Public Health, Sun Yat-Sen University , Guangzhou , China
| |
Collapse
|
23
|
Rager JE, Tilley SK, Tulenko SE, Smeester L, Ray PD, Yosim A, Currier JM, Ishida MC, González-Horta MDC, Sánchez-Ramírez B, Ballinas-Casarrubias L, Gutiérrez-Torres DS, Drobná Z, Del Razo LM, García-Vargas GG, Kim WY, Zhou YH, Wright FA, Stýblo M, Fry RC. Identification of novel gene targets and putative regulators of arsenic-associated DNA methylation in human urothelial cells and bladder cancer. Chem Res Toxicol 2015; 28:1144-55. [PMID: 26039340 DOI: 10.1021/tx500393y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
There is strong epidemiologic evidence linking chronic exposure to inorganic arsenic (iAs) to myriad adverse health effects, including cancer of the bladder. We set out to identify DNA methylation patterns associated with arsenic and its metabolites in exfoliated urothelial cells (EUCs) that originate primarily from the urinary bladder, one of the targets of arsenic-induced carcinogenesis. Genome-wide, gene-specific promoter DNA methylation levels were assessed in EUCs from 46 residents of Chihuahua, Mexico, and the relationship was examined between promoter methylation profiles and the intracellular concentrations of total arsenic and arsenic species. A set of 49 differentially methylated genes was identified with increased promoter methylation associated with EUC tAs, iAs, and/or monomethylated As (MMAs) enriched for their roles in metabolic disease and cancer. Notably, no genes had differential methylation associated with EUC dimethylated As (DMAs), suggesting that DMAs may influence DNA methylation-mediated urothelial cell responses to a lesser extent than iAs or MMAs. Further analysis showed that 22 of the 49 arsenic-associated genes (45%) are also differentially methylated in bladder cancer tissue identified using The Cancer Genome Atlas repository. Both the arsenic- and cancer-associated genes are enriched for the binding sites of common transcription factors known to play roles in carcinogenesis, demonstrating a novel potential mechanistic link between iAs exposure and bladder cancer.
Collapse
Affiliation(s)
- Julia E Rager
- †Department of Environmental Sciences and Engineering, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27516, United States
| | - Sloane K Tilley
- †Department of Environmental Sciences and Engineering, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27516, United States
| | - Samantha E Tulenko
- †Department of Environmental Sciences and Engineering, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27516, United States
| | - Lisa Smeester
- †Department of Environmental Sciences and Engineering, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27516, United States
| | - Paul D Ray
- †Department of Environmental Sciences and Engineering, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27516, United States.,‡Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Andrew Yosim
- †Department of Environmental Sciences and Engineering, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27516, United States
| | - Jenna M Currier
- ‡Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - María C Ishida
- §Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua, Chihuahua 31125, México
| | | | - Blanca Sánchez-Ramírez
- §Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua, Chihuahua 31125, México
| | | | | | - Zuzana Drobná
- ∥Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Luz M Del Razo
- ⊥Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México, DF 07360, México
| | - Gonzalo G García-Vargas
- #Facultad de Medicina, Universidad Juárez del Estado de Durango, Gómez Palacio, Durango 34000, México
| | - William Y Kim
- ○Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | | | | | - Miroslav Stýblo
- ‡Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States.,∥Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rebecca C Fry
- †Department of Environmental Sciences and Engineering, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27516, United States.,‡Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
24
|
Álvarez-González I, Garcia-Melo F, Vásquez-Garzón VR, Villa-Treviño S, Madrigal-Santillán EO, Morales-González JA, Mendoza-Pérez JA, Madrigal-Bujaidar E. Evaluation of blueberry juice in mouse azoxymethane-induced aberrant crypts and oxidative damage. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2014; 2014:379890. [PMID: 25258642 PMCID: PMC4166644 DOI: 10.1155/2014/379890] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 08/07/2014] [Accepted: 08/09/2014] [Indexed: 02/07/2023]
Abstract
Blueberry is a plant with a number of nutritional and biomedical capabilities. In the present study we initially evaluated the capacity of its juice (BJ) to inhibit the number of aberrant crypts (AC) induced with azoxymethane (AOM) in mouse. BJ was administered daily by the oral route to three groups of animals during four weeks (1.6, 4.1, and 15.0 μL/g), respectively, while AOM (10 mg/kg) was intraperitoneally injected to the mentioned groups, twice a week, in weeks two and three of the assay. We also included two control groups of mice, one administered distilled water and the other the high dose of BJ. A significant increase of AC was observed in the AOM treated animals, and a mean protection of 75.6% was determined with the two low doses of BJ tested; however, the high dose of the juice administered together with AOM increased the number of crypts more than four times the value observed in animals administered only AOM. Furthermore, we determined the antioxidant potential of BJ with an ex vivo DPPH assay and found a dose-dependent decrease with a mean of 19.5%. We also determined the DNA oxidation/antioxidation by identifying 8-hydroxy-2'-deoxyguanosine adducts and found a mean decrease of 44.3% with the BJ administration with respect to the level induced by AOM. Our results show a complex differential effect of BJ related to the tested doses, opening the need to further evaluate a number of factors so as to determine the possibility of a cocarcinogenic potential.
Collapse
Affiliation(s)
- Isela Álvarez-González
- 1Laboratorio de Genética, Escuela Nacional de Ciencias Biológicas, IPN, Unidad Profesional A. López Mateos, Avenida Wilfredo Massieu s/n, Zacatenco, Colonia, Lindavista, CP 07738, México, DF, Mexico
| | - Fernando Garcia-Melo
- 1Laboratorio de Genética, Escuela Nacional de Ciencias Biológicas, IPN, Unidad Profesional A. López Mateos, Avenida Wilfredo Massieu s/n, Zacatenco, Colonia, Lindavista, CP 07738, México, DF, Mexico
| | - Verónica R. Vásquez-Garzón
- 2Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados, IPN, Avenida Instituto Politécnico 2508, Colonia San Pedro Zacatenco, Del. Gustavo A. Madero, CP 06360, México, DF, Mexico
| | - Saúl Villa-Treviño
- 2Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados, IPN, Avenida Instituto Politécnico 2508, Colonia San Pedro Zacatenco, Del. Gustavo A. Madero, CP 06360, México, DF, Mexico
| | - E. Osiris Madrigal-Santillán
- 3Laboratorio de Medicina de la Conservación, Escuela Superior de Medicina, IPN, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, Del. Miguel Hidalgo, CP 11340, México, DF, Mexico
| | - José A. Morales-González
- 3Laboratorio de Medicina de la Conservación, Escuela Superior de Medicina, IPN, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, Del. Miguel Hidalgo, CP 11340, México, DF, Mexico
| | - Jorge A. Mendoza-Pérez
- 4Laboratorio de Química Ambiental. Escuela Nacional de Ciencias Biológicas, IPN, Unidad Profesional A. López Mateos, Avenida Wilfredo Massieu s/n, Zacatenco, Colonia Lindavista, CP 07738, México, DF, Mexico
| | - Eduardo Madrigal-Bujaidar
- 1Laboratorio de Genética, Escuela Nacional de Ciencias Biológicas, IPN, Unidad Profesional A. López Mateos, Avenida Wilfredo Massieu s/n, Zacatenco, Colonia, Lindavista, CP 07738, México, DF, Mexico
- *Eduardo Madrigal-Bujaidar:
| |
Collapse
|
25
|
Affiliation(s)
- Shengwen Shen
- Department
of Laboratory Medicine
and Pathology, 10-102 Clinical Sciences Building, University
of Alberta, Edmonton, Alberta, Canada, T6G 2G3
| | - Xing-Fang Li
- Department
of Laboratory Medicine
and Pathology, 10-102 Clinical Sciences Building, University
of Alberta, Edmonton, Alberta, Canada, T6G 2G3
| | - William R. Cullen
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver,
British Columbia, Canada, V6T 1Z1
| | - Michael Weinfeld
- Department of Oncology, Cross
Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, Alberta, Canada, T6G 1Z2
| | - X. Chris Le
- Department
of Laboratory Medicine
and Pathology, 10-102 Clinical Sciences Building, University
of Alberta, Edmonton, Alberta, Canada, T6G 2G3
| |
Collapse
|
26
|
De Coster S, van Leeuwen DM, Jennen DGJ, Koppen G, Den Hond E, Nelen V, Schoeters G, Baeyens W, van Delft JHM, Kleinjans JCS, van Larebeke N. Gender-specific transcriptomic response to environmental exposure in Flemish adults. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2013; 54:574-588. [PMID: 23653218 DOI: 10.1002/em.21774] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 02/01/2013] [Accepted: 02/21/2013] [Indexed: 06/02/2023]
Abstract
Flanders, Belgium, is one of the most densely populated areas in Europe. The Flemish Environment and Health Survey (2002-2006) aimed at determining exposure to pollutants of neonates, adolescents, and older adults and to assess associated biological and health effects. This study investigated genome wide gene expression changes associated with a range of environmental pollutants, including cadmium, lead, PCBs, dioxin, hexachlorobenzene, p,p'-DDE, benzene, and PAHs. Gene expression levels were measured in peripheral blood cells of 20 adults with relatively high and 20 adults with relatively low combined internal exposure levels, all non-smokers aged 50-65. Pearson correlation was used to analyze associations between pollutants and gene expression levels, separately for both genders. Pollutant- and gender-specific correlation analysis results were obtained. For organochlorine pollutants, analysis within genders revealed that genes were predominantly regulated in opposite directions in males and females. Significantly modulated pathways were found to be associated with each of the exposure biomarkers measured. Pathways and/or genes related to estrogen and STAT5 signaling were correlated to organochlorine exposures in both genders. Our work demonstrates that gene expression in peripheral blood is influenced by environmental pollutants. In particular, gender-specific changes are associated with organochlorine pollutants, including gender-specific modulation of endocrine related pathways and genes. These pathways and genes have previously been linked to endocrine disruption related disorders, which in turn have been associated with organochlorine exposure. Based on our results, we recommend that males and females be considered separately when analyzing gene expression changes associated with exposures that may include chemicals with endocrine disrupting properties.
Collapse
Affiliation(s)
- Sam De Coster
- Study Centre for Carcinogenesis and Primary Prevention of Cancer, Ghent University, Ghent, 9000, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
The carcinogenicity of cadmium, arsenic, and chromium(VI) compounds has been recognized for some decades. However, the underlying molecular mechanisms seem to be complex and are not completely understood at present. Although, with the exception of chromium(VI), direct DNA damage seems to be of minor importance, interactions with DNA repair processes, tumor suppressor functions, and signal transduction pathways have been described in diverse biological systems. In addition to the induction of damage to cellular macromolecules by reactive oxygen species, the interference with cellular redox regulation by reaction with redox-sensitive protein domains or amino acids may provide one plausible mechanism involved in metal carcinogenicity. Consequences are the distortion of zinc-binding structures and the activation or inactivation of redox-regulated signal transduction pathways, provoking metal-induced genomic instability. Nevertheless, the relevance of the respective mechanisms depends on the actual metal or metal species under consideration and more research is needed to further strengthen this hypothesis.
Collapse
Affiliation(s)
- Andrea Hartwig
- Institute of Applied Biosciences, Department of Food Chemistry and Toxicology, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany.
| |
Collapse
|
28
|
Huang S, Guo S, Guo F, Yang Q, Xiao X, Murata M, Ohnishi S, Kawanishi S, Ma N. CD44v6 expression in human skin keratinocytes as a possible mechanism for carcinogenesis associated with chronic arsenic exposure. Eur J Histochem 2013; 57:e1. [PMID: 23549458 PMCID: PMC3683606 DOI: 10.4081/ejh.2013.e1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 09/30/2012] [Accepted: 10/01/2012] [Indexed: 01/06/2023] Open
Abstract
Inorganic arsenic is a well-known human skin carcinogen. Chronic arsenic exposure results in various types of human skin lesions, including squamous cell carcinoma (SCC). To investigate whether mutant stem cells participate in arsenic-associated carcinogenesis, we repeatedly exposed the human spontaneously immortalized skin keratinocytes (HaCaT) cell line to an environmentally relevant level of arsenic (0.05 ppm) in vitrofor 18 weeks. Following sodium arsenite administration, cell cycle, colony-forming efficiency (CFE), cell tumorigenicity, and expression of CD44v6, NF-κB and p53, were analyzed at different time points (0, 5, 10, 15, 20, 25 and 30 passages). We found that a chronic exposure of HaCaT cells to a low level of arsenic induced a cancer stem-like phenotype. Furthermore, arsenictreated HaCaT cells also became tumorigenic in nude mice, their growth cycle was predominantly in G2/M and S phases. Relative to nontreated cells, they exhibited a higher growth rate and a significant increase in CFE. Western blot analysis found that arsenic was capable of increasing cell proliferation and sprouting of cancer stem-like phenotype. Additionally, immunohistochemical analysis demonstrated that CD44v6 expression was upregulated in HaCaT cells exposed to a low level of arsenic during early stages of induction. The expression of CD44v6 in arsenic-treated cells was positively correlated with their cloning efficiency in soft agar (r=0.949, P=0.01). Likewise, the expressions of activating transcription factor NF-κB and p53 genes in the arsenic-treated HaCaT cells were significantly higher than that in non-treated cells. Higher expressions of CD44v6, NF-κB and p53 were also observed in tumor tissues isolated from Balb/c nude mice. The present results suggest that CD44v6 may be a biomarker of arsenicinduced neoplastic transformation in human skin cells, and that arsenic promotes malignant transformation in human skin lesions through a NF-κB signaling pathway-stimulated expression of CD44v6.
Collapse
Affiliation(s)
- S Huang
- Suzuka University of Medical Science, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Chitta KR, Landero Figueroa JA, Caruso JA, Merino EJ. Selenium mediated arsenic toxicity modifies cytotoxicity, reactive oxygen species and phosphorylated proteins. Metallomics 2013; 5:673-85. [DOI: 10.1039/c3mt20213e] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
30
|
Xu Y, Li Y, Li H, Pang Y, Zhao Y, Jiang R, Shen L, Zhou J, Wang X, Liu Q. The accumulations of HIF-1α and HIF-2α by JNK and ERK are involved in biphasic effects induced by different levels of arsenite in human bronchial epithelial cells. Toxicol Appl Pharmacol 2013. [DOI: 10.1016/j.taap.2012.11.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
31
|
Li Y, Jiang R, Zhao Y, Xu Y, Ling M, Pang Y, Shen L, Zhou Y, Zhang J, Zhou J, Wang X, Liu Q. Opposed arsenite-mediated regulation of p53-survivin is involved in neoplastic transformation, DNA damage, or apoptosis in human keratinocytes. Toxicology 2012; 300:121-31. [DOI: 10.1016/j.tox.2012.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 05/17/2012] [Accepted: 06/08/2012] [Indexed: 10/28/2022]
|
32
|
|
33
|
Xu Y, Li Y, Pang Y, Ling M, Shen L, Yang X, Zhang J, Zhou J, Wang X, Liu Q. EMT and stem cell-like properties associated with HIF-2α are involved in arsenite-induced transformation of human bronchial epithelial cells. PLoS One 2012; 7:e37765. [PMID: 22662215 PMCID: PMC3360629 DOI: 10.1371/journal.pone.0037765] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 04/24/2012] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Arsenic is well-established as a human carcinogen, but the molecular mechanisms leading to arsenic-induced carcinogenesis are complex and elusive. It is not been determined if the epithelial-mesenchymal transition (EMT) and stem cell-like properties contribute in causing to carcinogen-induced malignant transformation and subsequent tumor formation. METHODS To investigate the molecular mechanisms underlying EMT and the emergence of cancer stem cell-like properties during neoplastic transformation of human bronchial epithelial (HBE) cells induced by chronic exposure to arsenite. HBE cells were continuously exposed to arsenite. Spheroid formation assays and analyses of side populations (SPs) were performed to confirm that arsenite induces the acquired EMT and cancer stem cell-like phenotype. Treated HBE cells were molecularly characterized by RT-PCR, Western blots, immunofluorescence, Southwestern assays, reporter assays, and chromatin immunoprecipitation. RESULTS With chronic exposure to arsenite, HBE cells undergo an EMT and then acquire a malignant cancer stem cell-like phenotype. Twist1 and Bmi1 are involved in arsenite-induced EMT. The process is directly regulated by HIF-2α. The self-renewal genes, Oct4, Bmi1, and ALDH1, are necessary for arsenite-mediated maintenance of stem cells. CONCLUSIONS EMT, regulated by HIF-2α, and the development of a cancer stem cell-like phenotype are associated with arsenite-induced transformation of HBE cells.
Collapse
Affiliation(s)
- Yuan Xu
- Institute of Toxicology, Nanjing Medical University, Nanjing, People's Republic of China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Yuan Li
- Institute of Toxicology, Nanjing Medical University, Nanjing, People's Republic of China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Ying Pang
- Institute of Toxicology, Nanjing Medical University, Nanjing, People's Republic of China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Min Ling
- Institute of Toxicology, Nanjing Medical University, Nanjing, People's Republic of China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Lu Shen
- Institute of Toxicology, Nanjing Medical University, Nanjing, People's Republic of China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Xiaojun Yang
- Department of General Surgery, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Jianping Zhang
- Department of General Surgery, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Jianwei Zhou
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Xinru Wang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Qizhan Liu
- Institute of Toxicology, Nanjing Medical University, Nanjing, People's Republic of China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
34
|
DNA-PKcs-mediated stabilization of p53 by JNK2 is involved in arsenite-induced DNA damage and apoptosis in human embryo lung fibroblast cells. Toxicol Lett 2012; 210:302-10. [DOI: 10.1016/j.toxlet.2012.02.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Accepted: 02/07/2012] [Indexed: 11/23/2022]
|
35
|
Blockade of p53 by HIF-2α, but not HIF-1α, is involved in arsenite-induced malignant transformation of human bronchial epithelial cells. Arch Toxicol 2012; 86:947-59. [PMID: 22447124 DOI: 10.1007/s00204-012-0810-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 01/31/2012] [Indexed: 12/15/2022]
Abstract
Hypoxia-inducible factors (HIFs), which consist of α and β subunits, are transcription factors involved in regulation of a variety of cellular functions. By blocking the function of the tumor suppressor p53, over-expressions of HIFs are linked to carcinogenesis and tumor progression. Inorganic arsenic, a ubiquitous environmental contaminant, is associated with an increased risk of cancer. Although there are several hypotheses regarding arsenic-induced carcinogenesis, the mechanism of action remains obscure. We have shown that long-term exposure of human bronchial epithelial (HBE) cells to a low level of arsenite increases their proliferation rate and anchorage-independent growth. When introduced into nude mice, the transformed cells are tumorigenic. The present report demonstrates that, with increased time of exposure to arsenite, there is more increased expression of HIF-2α, but not HIF-1α. These factors are known to have different functions, and, in some cases, opposite effects. Arsenite induces accumulation of HIF-2α by inhibiting its degradation through the ubiquitin-mediated proteasome pathway. HIF-2α knockdown, but not HIF-1α knockdown, increases the activation of p53. Finally, inhibition of HIF-2α blocks arsenite-induced proliferation and malignant transformation. Thus, our studies show that blockade of p53 function by inhibiting the ubiquitin-mediated proteasome degradation of HIF-2α, but not that of HIF-1α, is involved in arsenite-induced proliferation and neoplastic transformation of HBE cells.
Collapse
|
36
|
Naranmandura H, Carew MW, Xu S, Lee J, Leslie EM, Weinfeld M, Le XC. Comparative Toxicity of Arsenic Metabolites in Human Bladder Cancer EJ-1 Cells. Chem Res Toxicol 2011; 24:1586-96. [DOI: 10.1021/tx200291p] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Hua Naranmandura
- Department of Pharmacology, Toxicology, and Biochemical Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| | - Michael W. Carew
- Department of Physiology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Shi Xu
- Department of Pharmacology, Toxicology, and Biochemical Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jane Lee
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada
| | - Elaine M. Leslie
- Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
- Department of Physiology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Michael Weinfeld
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada
| | - X. Chris Le
- Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| |
Collapse
|
37
|
Li Y, Shen L, Xu H, Pang Y, Xu Y, Ling M, Zhou J, Wang X, Liu Q. Up-regulation of cyclin D1 by JNK1/c-Jun is involved in tumorigenesis of human embryo lung fibroblast cells induced by a low concentration of arsenite. Toxicol Lett 2011; 206:113-20. [PMID: 21726611 DOI: 10.1016/j.toxlet.2011.06.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 06/15/2011] [Accepted: 06/18/2011] [Indexed: 12/12/2022]
Abstract
Inorganic arsenic, a ubiquitous environmental contaminant, is associated with an increased risk of cancer. There are several hypotheses regarding arsenic-induced carcinogenesis. The mechanism of action remains obscure, although hyper-proliferation of cells is involved. In the present study, the molecular mechanisms underlying the proliferation and malignant transformation of human embryo lung fibroblast (HELF) cells induced by a low concentration of arsenite were investigated. The results reveal that a low concentration of arsenite induces cell proliferation and promotes cell cycle transition from the G(1) to the S phase. Moreover, arsenite activates the JNK1/c-Jun signal pathway, but not JNK2, which up-regulates the expression of cyclin D1/CDK4 and phosphorylates the retinoblastoma (Rb) protein. Blocking of the JNK1/c-Jun signal pathway suppresses the increases of cyclin D1 expression and Rb phosphorylation, which attenuates cell proliferation, reduces the transition from the G1 to the S phase, and thereby inhibits the neoplastic transformation of HELF cells induced by a low concentration of arsenite. Thus, activation of the JNK1/c-Jun pathway up-regulates the expression of cyclin D1, which is involved in the tumorigenesis caused by a low concentration of arsenite.
Collapse
Affiliation(s)
- Yuan Li
- Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Heintze E, Aguilera C, Davis M, Fricker A, Li Q, Martinez J, Gage MJ. Toxicity of depleted uranium complexes is independent of p53 activity. J Inorg Biochem 2011; 105:142-8. [PMID: 21194611 PMCID: PMC3018829 DOI: 10.1016/j.jinorgbio.2010.10.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Revised: 10/19/2010] [Accepted: 10/20/2010] [Indexed: 11/22/2022]
Abstract
The p53 tumor suppressor protein is one of the key checkpoints in cellular response to a variety of stress mechanisms, including exposure to various toxic metal complexes. Previous studies have demonstrated that arsenic and chromium complexes are able to activate p53, but there is a dearth of data investigating whether uranium complexes exhibit similar effects. The use of depleted uranium (DU) has increased in recent years, raising concern about DU's potential carcinogenic effects. Previous studies have shown that uranyl acetate and uranyl nitrate are capable of inducing DNA strand breaks and potentially of inducing oxidative stress through free radical generation, two potential mechanisms for activation of p53. Based on these studies, we hypothesized that either uranyl acetate or uranyl nitrate could act as an activator of p53. We tested this hypothesis using a combination of cytotoxicity assays, p53 activity assays, western blotting and flow cytometry. All of our results demonstrate that there is not a p53-mediated response to either uranyl acetate or uranyl nitrate, demonstrating that any cellular response to uranium exposure likely occurs in a p53-independent fashion under the conditions studied.
Collapse
Affiliation(s)
- Ellie Heintze
- Department of Chemistry and Biochemistry, Northern Arizona University, Flagstaff, AZ 86011
| | - Camille Aguilera
- Department of Chemistry and Biochemistry, Northern Arizona University, Flagstaff, AZ 86011
| | - Malia Davis
- Department of Chemistry and Biochemistry, Northern Arizona University, Flagstaff, AZ 86011
| | - Avery Fricker
- Department of Chemistry and Biochemistry, Northern Arizona University, Flagstaff, AZ 86011
| | - Qiang Li
- Department of Cell Biology and Anatomy, Arizona Cancer Center, Tucson, AZ, 85724
| | - Jesse Martinez
- Department of Cell Biology and Anatomy, Arizona Cancer Center, Tucson, AZ, 85724
| | - Matthew J. Gage
- Department of Chemistry and Biochemistry, Northern Arizona University, Flagstaff, AZ 86011
| |
Collapse
|
39
|
Singh KP, Kumari R, Treas J, DuMond JW. Chronic Exposure to Arsenic Causes Increased Cell Survival, DNA Damage, and Increased Expression of Mitochondrial Transcription Factor A (mtTFA) in Human Prostate Epithelial Cells. Chem Res Toxicol 2011; 24:340-9. [DOI: 10.1021/tx1003112] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Kamaleshwar P. Singh
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH), Texas Tech University, Lubbock, Texas 79409, United States
| | - Ragini Kumari
- Department of Environmental Science and Technology, Texas Southern University, Houston, Texas 77004, United States
| | - Justin Treas
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH), Texas Tech University, Lubbock, Texas 79409, United States
| | - James W. DuMond
- Department of Environmental Science and Technology, Texas Southern University, Houston, Texas 77004, United States
| |
Collapse
|
40
|
Rossman TG, Klein CB. Genetic and epigenetic effects of environmental arsenicals. Metallomics 2011; 3:1135-41. [DOI: 10.1039/c1mt00074h] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
41
|
Li Y, Xu Y, Ling M, Yang Y, Wang S, Li Z, Zhou J, Wang X, Liu Q. mot-2-Mediated cross talk between nuclear factor-B and p53 is involved in arsenite-induced tumorigenesis of human embryo lung fibroblast cells. ENVIRONMENTAL HEALTH PERSPECTIVES 2010; 118:936-942. [PMID: 20199942 PMCID: PMC2920912 DOI: 10.1289/ehp.0901677] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 03/03/2010] [Indexed: 05/28/2023]
Abstract
BACKGROUND Inactivation of p53 is involved in arsenite-induced tumorigenesis; however, the molecular mechanisms remain poorly understood. OBJECTIVE We investigated the molecular mechanisms underlying the inactivation of p53 and neoplastic transformation induced by arsenite in human embryo lung fibroblast (HELF) cells. METHODS Anchorage-independent growth assays were performed, and tumorigenicity in intact animals was assessed to confirm arsenite-induced neoplastic transformation. We determined the levels and functions of p53, nuclear factor-kappa B (NF-B; a key transcriptional regulator), and mot-2 (a p53 inhibitor) and their relationships in arsenite-induced transformed HELF cells by two-dimensional electrophoresis, reverse-transcriptase polymerase chain reaction, Western blot, immunofluorescence, and co-immunoprecipitation assays. RESULTS Exposure of HELF cells to low levels of arsenite increased their proliferation rate and anchorage-independent growth and disrupted normal contact inhibition. When introduced into nude mice, transformed cells were tumorigenic. We used proteomic analysis to identify proteins with altered expression between untreated and arsenite-exposed cells. We found decreased expression of NF-B repressing factor (NKRF; an inhibitor of NF-B-mediated gene transcription), increased expression of mot-2, and increased activation of NF-B. Changes in cells exposed to 1.0 microM arsenite were more marked than changes in cells exposed to 0.5 or 2.0 microM arsenite. Inactivation of NF-B prevented malignant transformation induced by 1.0 microM arsenite. Moreover, we also identified a mechanism whereby NF-B regulated p53. Specifically, activation of NF-B up-regulated mot-2 expression, which prevented nuclear translocation of p53 and switched the binding preference of the p53 and NF-B coactivator CBP [cyclic AMP-responsive element binding protein (CREB) binding protein] from p53 to NF-B. CONCLUSIONS mot-2-mediated cross talk between NF-B and p53 appears to be involved in arsenite-induced tumorigenesis of HELF cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Qizhan Liu
- Address correspondence to Q.Z. Liu, Department of Molecular Cell Biology and Toxicology, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P.R. China. Telephone: 86-25-8686-2834. Fax: 86-25-8652-7613. E-mail:
| |
Collapse
|
42
|
Liu SY, Wen CY, Lee YJ, Lee TC. XPC silencing sensitizes glioma cells to arsenic trioxide via increased oxidative damage. Toxicol Sci 2010; 116:183-93. [PMID: 20403967 DOI: 10.1093/toxsci/kfq113] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Arsenic exerts its cytotoxicity via the generation of reactive oxygen species and inhibition of DNA repair. How arsenic disturbs oxidative DNA damage repair is, however, unclear. We found that arsenic trioxide (ATO), like ultraviolet (UV) irradiation, induced the expression of xeroderma pigmentosum group C (XPC) but not of xeroderma pigmentosum A in a human glioma cell line, U87. To explore the role of XPC in the toxic effects of ATO, small interfering RNA was used to silence XPC (siXPC) in U87 cells. siXPC cells were more susceptible to UV irradiation and ATO-induced cell death than control cells. Increased siXPC cell death induced by ATO was accompanied by increased senescence and autophagy. Because increased DNA strand breaks in siXPC cells were observed only when cells were concomitantly treated with ATO and DNA repair inhibitors, XPC silencing apparently did not interfere with repair of ATO-induced DNA damage. Although intracellular ROS levels were not significantly enhanced in siXPC cells, ATO treatment did result in increased 8-hydroxy-2'-deoxyguanosine and hyperoxidized peroxiredoxin. Enhanced superoxide production and autophagy by ATO in siXPC cells were suppressed by co-incubation with N-acetylcysteine (NAC). Furthermore, XPC silencing caused decreased glutathione levels and increased catalase and Mn-superoxide dismutase activities. Increased catalase activity in siXPC cells was suppressed by ATO treatment. XPC silencing also enhanced reporter activity of activator protein-1, whereas enhanced activity was suppressed by NAC. Taken together, our results indicate that XPC silencing causes increased ATO susceptibility by disturbing redox homeostasis rather than reducing DNA repair.
Collapse
Affiliation(s)
- Shin-Yi Liu
- Department of Biomedical Image and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | | | | | | |
Collapse
|
43
|
Li Y, Ling M, Xu Y, Wang S, Li Z, Zhou J, Wang X, Liu Q. The Repressive Effect of NF-κB on p53 by Mot-2 Is Involved in Human Keratinocyte Transformation Induced by Low Levels of Arsenite. Toxicol Sci 2010; 116:174-82. [DOI: 10.1093/toxsci/kfq109] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
44
|
Hirner AV, Rettenmeier AW. Methylated Metal(loid) Species in Humans. ORGANOMETALLICS IN ENVIRONMENT AND TOXICOLOGY 2010. [DOI: 10.1039/9781849730822-00465] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
While the metal(loid)s arsenic, bismuth, and selenium (probably also tellurium) have been shown to be enzymatically methylated in the human body, this has not yet been demonstrated for antimony, cadmium, germanium, indium, lead, mercury, thallium, and tin, although the latter elements can be biomethylated in the environment. Methylated metal(loid)s exhibit increased mobility, thus leading to a more efficient metal(loid) transport within the body and, in particular, opening chances for passing membrane barriers (blood-brain barrier, placental barrier). As a consequence human health may be affected. In this review, relevant data from the literature are compiled, and are discussed with respect to the evaluation of assumed and proven health effects caused by alkylated metal(loid) species.
Collapse
Affiliation(s)
- Alfred V. Hirner
- Institute of Analytical Chemistry, University of Duisburg-Essen D-45117 Essen Germany
| | - Albert W. Rettenmeier
- Institute of Hygiene and Occupational Medicine, University of Duisburg-Essen D-45122 Essen Germany
| |
Collapse
|
45
|
Saunders JR, Knopper LD, Koch I, Reimer KJ. Arsenic transformations and biomarkers in meadow voles (Microtus pennsylvanicus) living on an abandoned gold mine site in Montague, Nova Scotia, Canada. THE SCIENCE OF THE TOTAL ENVIRONMENT 2010; 408:829-835. [PMID: 19945142 DOI: 10.1016/j.scitotenv.2009.11.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Revised: 11/03/2009] [Accepted: 11/04/2009] [Indexed: 05/28/2023]
Abstract
Arsenic is one of the most widely encountered environmental contaminants because of a number of anthropogenic sources; in Canada the main anthropogenic release of arsenic is from mine tailings ponds. The present study is part of a series of studies to measure chemical and biological effects of exposure for meadow voles (Microtus pennsylvanicus) living on arsenic contaminated sites. Two additional objectives were addressed in the present study: the effect of higher arsenic concentrations compared with previous studies, and the comparison of chemical speciation and biological effects. To obtain the higher environmental concentrations, specimens were collected from a former gold mining site in Montague, NS that contains highly elevated concentrations of arsenic in soils and plants. Meadow voles were collected and their tissues were analyzed for total arsenic to measure uptake, and arsenic speciation to examine the chemical effects of the high arsenic exposure. In addition to the arsenic analysis, a biomonitoring study was undertaken to examine the sub-cellular effects in meadow voles resulting from the elevated arsenic exposure. Meadow voles living on the contaminated site had substantially higher concentrations of total arsenic than animals from the background (reference) location. The extractable arsenic in internal tissues was present mainly as monomethylarsonic acid (up to 14% of total arsenic). A statistically significant relationship was observed between the reduction of glutathione in vole livers and the increase in liver arsenic concentrations, and micronucleated monochromatic red blood cells were also significantly elevated in voles from the arsenic contaminated site. This is one of the few field studies where sub-cellular effects were observed, and the first to show a co-existence of such effects with relatively high proportions of monomethylarsonic acid in voles living near mine tailings.
Collapse
Affiliation(s)
- Jared R Saunders
- Environmental Sciences Group, Royal Military College of Canada, Kingston, Ontario, Canada K7K 7B4
| | | | | | | |
Collapse
|
46
|
Escudero-Lourdes C, Medeiros MK, Cárdenas-González MC, Wnek SM, Gandolfi JA. Low level exposure to monomethyl arsonous acid-induced the over-production of inflammation-related cytokines and the activation of cell signals associated with tumor progression in a urothelial cell model. Toxicol Appl Pharmacol 2010; 244:162-73. [PMID: 20045430 DOI: 10.1016/j.taap.2009.12.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Revised: 12/15/2009] [Accepted: 12/18/2009] [Indexed: 02/07/2023]
Abstract
Human bladder cancer has been associated with chronic exposure to arsenic. Chronic exposure of an immortalized non-tumorigenic urothelial cell line (UROtsa cells) to arsenicals has transformed these cells to a malignant phenotype, but the involved mechanisms are not fully understood. Chronic inflammation has been linked with cancer development mainly because many pro-inflammatory cytokines, growth factors as well as angiogenic chemokines have been found in tumors. In this study the chronology of inflammatory cytokines production was profiled in UROtsa cells chronically exposed to the toxic arsenic metabolite, monomethylarsonous acid [50 nM MMA(III)] to know the role of inflammation in cell transformation. Acute 50 nM MMA(III) exposure induced over-production of many pro-inflammatory cytokines as soon as 12 h after acute exposure. The same cytokines remain over-regulated after chronic exposure to 50 nM MMA(III), especially after 3 mo exposure. At 3 mo exposure the sustained production of cytokines like IL-1, IL-6, IL-8 and TNF is coincident with the appearance of characteristics associated with cell transformation seen in other arsenic-UROtsa studies. The sustained and increased activation of NFkappaB and c-Jun is also present along the transformation process and the phosphorylated proteins p38 MAPK and ERK 1/2 are increased also through the time line. Taken together these results support the notion that chronic inflammation is associated within MMA(III)-induced cell transformation and may act as a promoting factor in UROtsa cell transformation.
Collapse
Affiliation(s)
- C Escudero-Lourdes
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, San Luis Potosí, SLP 78240, Mexico.
| | | | | | | | | |
Collapse
|
47
|
Kligerman AD, Malik SI, Campbell JA. Cytogenetic insights into DNA damage and repair of lesions induced by a monomethylated trivalent arsenical. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2010; 695:2-8. [DOI: 10.1016/j.mrgentox.2009.09.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 09/22/2009] [Accepted: 09/24/2009] [Indexed: 01/23/2023]
|
48
|
|
49
|
Arsenite enhances the benzo[a]pyrene diol epoxide (BPDE)-induced mutagenesis with no marked effect on repair of BPDE-DNA adducts in human lung cells. Toxicol In Vitro 2009; 23:897-905. [DOI: 10.1016/j.tiv.2009.05.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Revised: 05/01/2009] [Accepted: 05/18/2009] [Indexed: 12/16/2022]
|
50
|
Nollen M, Ebert F, Moser J, Mullenders LHF, Hartwig A, Schwerdtle T. Impact of arsenic on nucleotide excision repair: XPC function, protein level, and gene expression. Mol Nutr Food Res 2009; 53:572-82. [PMID: 19382146 DOI: 10.1002/mnfr.200800480] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The ubiquitous occurrence of the human carcinogen arsenic results in multiple exposure possibilities to humans. The human diet, especially drinking water, is the primary source of inorganic arsenic intake in the general population. The ingested arsenic is metabolized to methylated derivatives; some of these metabolites are today considered to be more toxic than the inorganic species. Various modes of action have been proposed to contribute to arsenic carcinogenicity; inhibition of nucleotide excision repair (NER), removing DNA helix distorting DNA adducts induced by environmental mutagens, is likely to be of primary importance. Here, we report that arsenite and its metabolite monomethylarsonous acid (MMA(III)) strongly decreased expression and protein level of Xeroderma pigmentosum complementation group C (XPC), which is believed to be the principle initiator of global genome NER. This led to diminished association of XPC to sites of local UVC damage, resulting in decreased recruitment of further NER proteins. Additionally Xeroderma pigmentosum complementation group E protein (XPE) expression was reduced, which encodes for another important NER protein and similarly to XPC is regulated by the activity of the transcription factor p53. In summary, our data demonstrate that in human skin fibroblasts arsenite and even more pronounced MMA(III) interact with XPC expression, resulting in decreased XPC protein level and diminished assembly of the NER machinery.
Collapse
Affiliation(s)
- Maike Nollen
- Institut für Lebensmittelchemie, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | | | | | | | | | | |
Collapse
|