1
|
Abstract
Exposure to arsenic in contaminated drinking water is an emerging public health problem that impacts more than 200 million people worldwide. Accumulating lines of evidence from epidemiological studies revealed that chronic exposure to arsenic can result in various human diseases including cancer, type 2 diabetes, and neurodegenerative disorders. Arsenic is also classified as a Group I human carcinogen. In this review, we survey extensively different modes of action for arsenic-induced carcinogenesis, with focus being placed on arsenic-mediated impairment of DNA repair pathways. Inorganic arsenic can be bioactivated by methylation, and the ensuing products are highly genotoxic. Bioactivation of arsenicals also elicits the production of reactive oxygen and nitrogen species (ROS and RNS), which can directly damage DNA and modify cysteine residues in proteins. Results from recent studies suggest zinc finger proteins as crucial molecular targets for direct binding to As3+ or for modifications by arsenic-induced ROS/RNS, which may constitute a common mechanism underlying arsenic-induced perturbations of DNA repair.
Collapse
|
2
|
Arsenic-induced apoptosis in the p53-proficient and p53-deficient cells through differential modulation of NFkB pathway. Food Chem Toxicol 2018; 118:849-860. [PMID: 29944914 DOI: 10.1016/j.fct.2018.06.053] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/21/2018] [Accepted: 06/22/2018] [Indexed: 12/31/2022]
Abstract
Arsenic is a well-known environmental carcinogen and an effective chemotherapeutic agent. The underlying mechanism of this dual-effect, however, is not fully understood. In this study, we applied mouse p53+/+ and p53-/- cells to examine the NFκB pathway and proinflammatory cytokines after arsenic treatment. Arsenic reduced cell viability and increased more apoptosis in the p53-/- cells as compared to p53+/+ cells, which was correlated with activation of SAPK/JNK, p38 MAPK, and AKT pathways. A transcriptional regulatory network analysis revealed that arsenic activated transcription regulatory elements E2F, Egr1, Trp53, Stat6, Bcl6, Creb2 and ATF4 in the p53+/+ cells, while in the p53-/- cells, arsenic treatment altered transcription factors NFκB, Pparg, Creb2, ATF4, and Egr1. We observed dynamic changes in phosphorylated NFκB p65 (p-NFκB p65) and phosphorylated IKKαβ (p-IKKαβ) in both genotypes from 4 h to 24 h after treatment, significant decreases of p-NFκB p65 and p-IKKαβ in the p53-/- cells, whereas increases of p-NFκB p65 and p-IKKαβ were observed in the p53+/+ cells. Our study confirmed the differential modulation of NFκB pathway by arsenic in the p53+/+ or p53-/- cells and this observation of the differential mechanism of cell death between the p53+/+ and p53-/- cells might be linked to the unique ability of arsenic to act as both a carcinogen and a chemotherapeutic agent.
Collapse
|
3
|
Di Paolo C, Müller Y, Thalmann B, Hollert H, Seiler TB. p53 induction and cell viability modulation by genotoxic individual chemicals and mixtures. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:4012-4022. [PMID: 28303539 DOI: 10.1007/s11356-017-8790-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 03/08/2017] [Indexed: 06/06/2023]
Abstract
The binding of the p53 tumor suppression protein to DNA response elements after genotoxic stress can be quantified by cell-based reporter gene assays as a DNA damage endpoint. Currently, bioassay evaluation of environmental samples requires further knowledge on p53 induction by chemical mixtures and on cytotoxicity interference with p53 induction analysis for proper interpretation of results. We investigated the effects of genotoxic pharmaceuticals (actinomycin D, cyclophosphamide) and nitroaromatic compounds (4-nitroquinoline 1-oxide, 3-nitrobenzanthrone) on p53 induction and cell viability using a reporter gene and a colorimetric assay, respectively. Individual exposures were conducted in the absence or presence of metabolic activation system, while binary and tertiary mixtures were tested in its absence only. Cell viability reduction tended to present direct correlation with p53 induction, and induction peaks occurred mainly at chemical concentrations causing cell viability below 80%. Mixtures presented in general good agreement between predicted and measured p53 induction factors at lower concentrations, while higher chemical concentrations gave lower values than expected. Cytotoxicity evaluation supported the selection of concentration ranges for the p53 assay and the interpretation of its results. The often used 80% viability threshold as a basis to select the maximum test concentration for cell-based assays was not adequate for p53 induction assessment. Instead, concentrations causing up to 50% cell viability reduction should be evaluated in order to identify the lowest observed effect concentration and peak values following meaningful p53 induction.
Collapse
Affiliation(s)
- Carolina Di Paolo
- Department of Ecosystem Analysis, Institute for Environmental Research, Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany.
| | - Yvonne Müller
- Department of Ecosystem Analysis, Institute for Environmental Research, Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany
| | - Beat Thalmann
- Department of Ecosystem Analysis, Institute for Environmental Research, Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany
| | - Henner Hollert
- Department of Ecosystem Analysis, Institute for Environmental Research, Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany
- College of Resources and Environmental Science, Chongqing University, 1 Tiansheng Road, Beibei, Chongqing, 400715, China
- College of Environmental Science and Engineering and State Key Laboratory of Pollution Control and Resource Reuse, Tongji University, 1239 Siping Road, Shanghai, China
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, China
| | - Thomas-Benjamin Seiler
- Department of Ecosystem Analysis, Institute for Environmental Research, Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany
| |
Collapse
|
4
|
Prim-O-glucosylcimifugin induces cell cycle arrest and apoptosis in acute lymphoblastic leukemia cells. Anticancer Drugs 2017; 28:943-951. [DOI: 10.1097/cad.0000000000000543] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
5
|
|
6
|
Kim S, Lee SH, Lee S, Park JD, Ryu DY. Arsenite-induced changes in hepatic protein abundance in cynomolgus monkeys (Macaca fascicularis). Proteomics 2014; 14:1833-43. [PMID: 24866292 DOI: 10.1002/pmic.201300509] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 04/01/2014] [Accepted: 05/20/2014] [Indexed: 11/07/2022]
Abstract
Arsenic is an environmental pollutant, and its liver toxicity has long been recognized. The effect of arsenic on liver protein expression was analyzed using a proteomic approach in monkeys. Monkeys were orally administered sodium arsenite (SA) for 28 days. As shown by 2D-PAGE in combination with MS, the expression levels of 16 proteins were quantitatively changed in SA-treated monkey livers compared to control-treated monkey livers. Specifically, the levels of two proteins, mortalin and tubulin beta chain, were increased, and 14 were decreased, including plastin-3, cystathionine-beta-synthase, selenium-binding protein 1, annexin A6, alpha-enolase, phosphoenolpyruvate carboxykinase-M, erlin-2, and arginase-1. In view of their functional roles, differential expression of these proteins may contribute to arsenic-induced liver toxicity, including cell death and carcinogenesis. Among the 16 identified proteins, four were selected for validation by Western blot and immunohistochemistry. Additional Western blot analyses indicated arsenic-induced dysregulation of oxidative stress related, genotoxicity-related, and glucose metabolism related proteins in livers from SA-treated animals. Many changes in the abundance of toxicity-related proteins were also demonstrated in SA-treated human hepatoma cells. These data on the arsenic-induced regulation of proteins with critical roles may help elucidate the specific mechanisms underlying arsenic-induced liver toxicity.
Collapse
Affiliation(s)
- Soohee Kim
- BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | | | | | | | | |
Collapse
|
7
|
Qi Y, Li H, Zhang M, Zhang T, Frank J, Chen G. Autophagy in arsenic carcinogenesis. ACTA ACUST UNITED AC 2014; 66:163-8. [DOI: 10.1016/j.etp.2014.01.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 01/10/2014] [Accepted: 01/20/2014] [Indexed: 12/31/2022]
|
8
|
Olarte I, Martinez A, Ramos-Peñafiel C, Castellanos-Sinco H, Zamora J, Collazo-Jaloma J, Gutiérrez M, Gutiérrez-Kobeh L, Chavez-Olmos P, Manzanilla H, Garrido-Guerrero E, Ordoñez-Razo RM, Miranda EI. MAGE-A3 expression is an adverse prognostic factor in diffuse large B-cell lymphoma. ACTA ACUST UNITED AC 2012; 16:368-72. [PMID: 22183072 DOI: 10.1179/102453311x13085644680384] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
This study evaluates the prognostic value of MAGE-A3 expression in 28 diffuse large B-cell lymphoma (DLBCL) patients. A significant association was observed between MAGE-A3 expressions, assessed by quantitative real-time RT-polymerase chain reaction (PCR), with advanced stages of disease (P < 0.05). Elevated serum lactate dehydrogenase (LDH) levels and International Prognostic Index (IPI) score were significantly higher in MAGE-A3-positive patients (P = 0.025 and P = 0.004, respectively). Expression of MAGE-A3 was associated with poor response to treatment and a significantly shorter overall survival (P < 0.001). Our data address new information in the association of MAGE-A3 expression and poor prognosis in DLBCL patients.
Collapse
Affiliation(s)
- Irma Olarte
- Laboratorio de Biología Molecular, Servicio de Hematología, Hospital General de México, México D.F., México
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Wang ZF, Guo X. Arsenite-induced apoptosis is prevented by selenite in A375 cell line. Biol Trace Elem Res 2011; 140:7-17. [PMID: 20390378 DOI: 10.1007/s12011-010-8674-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Accepted: 03/04/2010] [Indexed: 10/19/2022]
Abstract
Arsenic trioxide induces apoptosis and clinical remission in patients diagnosed with acute promyelocytic leukemia. The human malignant melanoma A375 cells were treated with NaAsO2 (0.1–130 μM) and also treated with combined 10 μM NaAsO2 and 10 μM Na2SeO3. NaAsO2 arrested cell growth in the G1 phase and induced apoptosis in a concentration- and time-dependent manner. In contrast, administration of Na2SeO3 antagonized the cell growth inhibition and apoptosis induced by NaAsO2. The NaAsO2 treatment resulted in a marked increase in p53 protein as early as 4 h and in Bcl-2 protein level by 12 h. In addition, p53 downregulation accompanied the combined treatment of NaAsO2 and Na2SeO3. Thus, our results indicate upregulation of p53 and Bcl-2 play acrucial role in the NaAsO2-induced G1 arrest and apoptosis of A375 cells and that downregulation p53 appears to contribute to the inhibition by Na2SeO3 of the effects induced by NaAsO2.
Collapse
Affiliation(s)
- Zhi-Fang Wang
- Faculty of Public Health, College of Medicine, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | | |
Collapse
|
10
|
Habib GM. Arsenite causes down-regulation of Akt and c-Fos, cell cycle dysfunction and apoptosis in glutathione-deficient cells. J Cell Biochem 2010; 110:363-71. [PMID: 20336670 PMCID: PMC2862122 DOI: 10.1002/jcb.22548] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Arsenic is a well-known environmental toxicant but the mechanism by which it causes cytotoxicity is poorly understood. Arsenite induces apoptosis in glutathione (GSH)-deficient GCS-2 cells by causing cell cycle dysfunction and down-regulating critical signaling pathways. This study was designed to examine the effect of arsenite on redox-sensitive phosphatidylinositol 3-kinase (PI3K)/Akt, a signaling pathway involved in cell survival and growth, and transcription factor, activating protein-1 (AP-1). Arsenite significantly diminished Akt and c-Fos levels and caused accelerated degradation of these proteins by ubiquitnation. Arsenite also induced cell cycle arrest and apoptosis. The cell cycle arrest involved the down-regulation of cyclin A2, cyclin D1, cyclin E, cyclin dependent kinases (CDK) 2, CDK4, and CDK6. Apoptosis involved down-regulation of anti-apoptotic proteins Bcl-2, Bcl-xL, survivin, and inhibitor of apoptosis protein (IAP) and up-regulation of pro-apoptotic protein Bax. Taken together, our results suggest that a possible mechanism of arsenite-induced toxicity under low/no GSH conditions, is to negatively regulate GCS-2 cell proliferation by attenuating Akt and AP-1 by ubiquitination and causing cell cycle dysfunction and apoptosis.
Collapse
Affiliation(s)
- Geetha M Habib
- Department of Pathology, Baylor College of Medicine, Houston, Texas 77030, USA.
| |
Collapse
|
11
|
Sumi D, Shinkai Y, Kumagai Y. Signal transduction pathways and transcription factors triggered by arsenic trioxide in leukemia cells. Toxicol Appl Pharmacol 2010; 244:385-92. [PMID: 20193703 DOI: 10.1016/j.taap.2010.02.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 02/09/2010] [Accepted: 02/21/2010] [Indexed: 12/23/2022]
Abstract
Arsenic trioxide (As(2)O(3)) is widely used to treat acute promyelocytic leukemia (APL). Several lines of evidence have indicated that As(2)O(3) affects signal transduction and transactivation of transcription factors, resulting in the stimulation of apoptosis in leukemia cells, because some transcription factors are reported to associate with the redox condition of the cells, and arsenicals cause oxidative stress. Thus, the disturbance and activation of the cellular signaling pathway and transcription factors due to reactive oxygen species (ROS) generation during arsenic exposure may explain the ability of As(2)O(3) to induce a complete remission in relapsed APL patients. In this report, we review recent findings on ROS generation and alterations in signal transduction and in transactivation of transcription factors during As(2)O(3) exposure in leukemia cells.
Collapse
Affiliation(s)
- Daigo Sumi
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180, Yamashiro-cho, Tokushima-city, Tokushima 770-8514, Japan.
| | | | | |
Collapse
|
12
|
Bolt AM, Byrd RM, Klimecki WT. Autophagy is the predominant process induced by arsenite in human lymphoblastoid cell lines. Toxicol Appl Pharmacol 2010; 244:366-73. [PMID: 20153345 DOI: 10.1016/j.taap.2010.01.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 12/21/2009] [Accepted: 01/13/2010] [Indexed: 10/19/2022]
Abstract
Arsenic is a widespread environmental toxicant with a diverse array of molecular targets and associated diseases, making the identification of the critical mechanisms and pathways of arsenic-induced cytotoxicity a challenge. In a variety of experimental models, over a range of arsenic exposure levels, apoptosis is a commonly identified arsenic-induced cytotoxic pathway. Human lymphoblastoid cell lines (LCL) have been used as a model system in arsenic toxicology for many years, but the exact mechanism of arsenic-induced cytotoxicity in LCL is still unknown. We investigated the cytotoxicity of sodium arsenite in LCL 18564 using a set of complementary markers for cell death pathways. Markers indicative of apoptosis (phosphatidylserine externalization, PARP cleavage, and sensitivity to caspase inhibition) were uniformly negative in arsenite exposed cells. Interestingly, electron microscopy, acidic vesicle fluorescence, and expression of LC3 in LCL 18564 identified autophagy as an arsenite-induced process that was associated with cytotoxicity. Autophagy, a cellular programmed response that is associated with both cellular stress adaptation as well as cell death appears to be the predominant process in LCL cytotoxicity induced by arsenite. It is unclear, however, whether LCL autophagy is an effector mechanism of arsenite cytotoxicity or alternatively a cellular compensatory mechanism. The ability of arsenite to induce autophagy in lymphoblastoid cell lines introduces a potentially novel mechanistic explanation of the well-characterized in vitro and in vivo toxicity of arsenic to lymphoid cells.
Collapse
Affiliation(s)
- Alicia M Bolt
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85724, USA
| | | | | |
Collapse
|
13
|
Gentry PR, McDonald TB, Sullivan DE, Shipp AM, Yager JW, Clewell HJ. Analysis of genomic dose-response information on arsenic to inform key events in a mode of action for carcinogenicity. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:1-14. [PMID: 19551812 DOI: 10.1002/em.20505] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
A comprehensive literature search was conducted to identify information on gene expression changes following exposures to inorganic arsenic compounds. This information was organized by compound, exposure, dose/concentration, species, tissue, and cell type. A concentration-related hierarchy of responses was observed, beginning with changes in gene/protein expression associated with adaptive responses (e.g., preinflammatory responses, delay of apoptosis). Between 0.1 and 10 microM, additional gene/protein expression changes related to oxidative stress, proteotoxicity, inflammation, and proliferative signaling occur along with those related to DNA repair, cell cycle G2/M checkpoint control, and induction of apoptosis. At higher concentrations (10-100 microM), changes in apoptotic genes dominate. Comparisons of primary cell results with those obtained from immortalized or tumor-derived cell lines were also evaluated to determine the extent to which similar responses are observed across cell lines. Although immortalized cells appear to respond similarly to primary cells, caution must be exercised in using gene expression data from tumor-derived cell lines, where inactivation or overexpression of key genes (e.g., p53, Bcl-2) may lead to altered genomic responses. Data from acute in vivo exposures are of limited value for evaluating the dose-response for gene expression, because of the transient, variable, and uncertain nature of tissue exposure in these studies. The available in vitro gene expression data, together with information on the metabolism and protein binding of arsenic compounds, provide evidence of a mode of action for inorganic arsenic carcinogenicity involving interactions with critical proteins, such as those involved in DNA repair, overlaid against a background of chemical stress, including proteotoxicity and depletion of nonprotein sulfhydryls. The inhibition of DNA repair under conditions of toxicity and proliferative pressure may compromise the ability of cells to maintain the integrity of their DNA.
Collapse
|
14
|
Genome-wide analysis of BEAS-2B cells exposed to trivalent arsenicals and dimethylthioarsinic acid. Toxicology 2010; 268:31-9. [DOI: 10.1016/j.tox.2009.11.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 10/30/2009] [Accepted: 11/20/2009] [Indexed: 01/22/2023]
|
15
|
Duan Q, Komissarova E, Dai W. Arsenic trioxide suppresses paclitaxel-induced mitotic arrest. Cell Prolif 2009; 42:404-11. [PMID: 19397590 DOI: 10.1111/j.1365-2184.2009.00606.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES To understand if there exists a functional interaction between arsenic trioxide and paclitaxel in vitro. MATERIALS AND METHODS HeLa and HCT116 (rho53(+/+) and rho53(-/-)) cells were treated with As2O3 and/or paclitaxel for various times. Treated cells were collected for analyses using a combination of flow cytometry, fluorescence microscopy and Western blotting. RESULTS Because As(2)O(3) is capable of inhibiting tubulin polymerization and inducing mitotic arrest, we examined whether there existed any functional interaction between As(2)O(3) and paclitaxel, a well-known microtubule poison. Flow cytometry and fluorescence microscopy revealed that although As(2)O(3) alone caused a moderate level of mitotic arrest, it greatly attenuated paclitaxel-induced mitotic arrest in cells with p53 deficiency. Western blot analysis showed that As(2)O(3) significantly blocked phosphorylation of BubR1, Cdc20, and Cdc27 in cells treated with paclitaxel, suggesting that arsenic compromised the activation of the spindle checkpoint. Our further studies revealed that the attenuation of paclitaxel-induced mitotic arrest by As(2)O(3) resulted primarily from sluggish cell cycle progression at S phase but not enhanced mitotic exit. CONCLUSION The observations that As(2)O(3) has a negative impact on the cell cycle checkpoint activation by taxol should have significant clinical implications because the efficacy of taxol in the clinics is associated with its ability to induce mitotic arrest and subsequent mitotic catastrophe.
Collapse
Affiliation(s)
- Q Duan
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | | |
Collapse
|
16
|
Yu X, Robinson JF, Gribble E, Hong SW, Sidhu JS, Faustman EM. Gene expression profiling analysis reveals arsenic-induced cell cycle arrest and apoptosis in p53-proficient and p53-deficient cells through differential gene pathways. Toxicol Appl Pharmacol 2008; 233:389-403. [PMID: 18929588 PMCID: PMC2677763 DOI: 10.1016/j.taap.2008.09.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Revised: 09/08/2008] [Accepted: 09/09/2008] [Indexed: 12/30/2022]
Abstract
Arsenic (As) is a well-known environmental toxicant and carcinogen as well as an effective chemotherapeutic agent. The underlying mechanism of this dual capability, however, is not fully understood. Tumor suppressor gene p53, a pivotal cell cycle checkpoint signaling protein, has been hypothesized to play a possible role in mediating As-induced toxicity and therapeutic efficiency. In this study, we found that arsenite (As(3+)) induced apoptosis and cell cycle arrest in a dose-dependent manner in both p53(+/+) and p53(-/-) mouse embryonic fibroblasts (MEFs). There was, however, a distinction between genotypes in the apoptotic response, with a more prominent induction of caspase-3 in the p53(-/-) cells than in the p53(+/+) cells. To examine this difference further, a systems-based genomic analysis was conducted comparing the critical molecular mechanisms between the p53 genotypes in response to As(3+). A significant alteration in the Nrf2-mediated oxidative stress response pathway was found in both genotypes. In p53(+/+) MEFs, As(3+) induced p53-dependent gene expression alterations in DNA damage and cell cycle regulation genes. However, in the p53(-/-) MEFs, As(3+) induced a significant up-regulation of pro-apoptotic genes (Noxa) and down-regulation of genes in immune modulation. Our findings demonstrate that As-induced cell death occurs through a p53-independent pathway in p53 deficient cells while apoptosis induction occurs through p53-dependent pathway in normal tissue. This difference in the mechanism of apoptotic responses between the genotypes provides important information regarding the apparent dichotomy of arsenic's dual mechanisms, and potentially leads to further advancement of its utility as a chemotherapeutic agent.
Collapse
Affiliation(s)
- Xiaozhong Yu
- Institute for Risk Analysis and Risk Communication, Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA.
| | | | | | | | | | | |
Collapse
|
17
|
Salazar AM, Sordo M, Ostrosky-Wegman P. Relationship between micronuclei formation and p53 induction. Mutat Res 2008; 672:124-8. [PMID: 19041731 DOI: 10.1016/j.mrgentox.2008.10.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2008] [Revised: 10/25/2008] [Accepted: 10/31/2008] [Indexed: 11/30/2022]
Abstract
Human exposure to multiple chemicals compromises the integrity of genetic material. Hence, it is essential to determine the extent of DNA damage induced by xenobiotics. In cell lines, the induction of p53 expression in response to treatments with DNA-damaging agents has been proposed as a tool for the detection of genotoxic damage, although a direct correlation between a marker of chromosomal damage and p53 expression has not previously been studied. The micronucleus assay is a widely used genotoxicity test that has been shown to detect structural and numerical chromosomal damage. The present study was designed to characterize the relationship between micronuclei and p53 induction. RKO cells were cultured and treated with non-cytotoxic concentrations of colchicine, vinblastine, bleomycin or arsenic. Mannitol and clofibrate, which are non-genotoxic chemicals, were also included. The frequency of micronuclei was evaluated using the cytokinesis-block assay, and p53 induction was measured by Western blot assay. Our data showed that a significant induction of micronuclei and of p53 protein occurred only with the genotoxic chemicals. No differences in p53 induction were associated with the clastogenic or aneuplodogenic potential of the chemical exposure. The linear regression analysis revealed a direct relationship between p53 levels and the induction of micronuclei (p=0.0001, r(2)=0.9372), indicating that the level of p53 is associated with chromosomal damage.
Collapse
Affiliation(s)
- Ana María Salazar
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, D.F., Mexico.
| | | | | |
Collapse
|
18
|
Wu YC, Yen WY, Yih LH. Requirement of a functional spindle checkpoint for arsenite-induced apoptosis. J Cell Biochem 2008; 105:678-87. [DOI: 10.1002/jcb.21861] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
19
|
The activity of calpains in lymphocytes is glucose-dependent and is decreased in diabetic patients. Blood Cells Mol Dis 2007; 40:414-9. [PMID: 17964829 DOI: 10.1016/j.bcmd.2007.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2007] [Accepted: 08/22/2007] [Indexed: 11/20/2022]
Abstract
Calpains are nonlysosomal calcium-dependent cysteine proteases that participate in insulin secretion and action. Polymorphisms in the calpain-10 gene have been shown to increase the risk for type 2 diabetes. Since white blood cells have been used to study glucose homeostasis, the present study was carried to find out if calpains have different activity and/or expression in accessible cells such as lymphocytes of individuals with or without type 2 diabetes. Fasting blood glucose concentration was significantly higher in diabetic subjects, whereas the difference in the activity of calpains evaluated in basal and stimulating extracellular glucose concentration was significantly higher in the lymphocytes from the control group. The mRNA expression of calpain-10 was similar in the lymphocytes of both patients and controls. The protein blots showed four bands that ranged between 75 and 50 kDa; however, no statistical differences were observed in the expression of the calpain-10 isoforms between controls and patients. Data obtained showed that human lymphocytes express calpain-10 mRNA and protein, showing a similar expression between diabetic and control subjects, nevertheless in the diabetic group calpain activity was less glucose-sensitive.
Collapse
|
20
|
Tapia-Vieyra JV, Ostrosky-Wegman P, Mas-Oliva J. Proapoptotic role of novel gene-expression factors. Clin Transl Oncol 2007; 9:355-63. [PMID: 17594949 DOI: 10.1007/s12094-007-0067-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The mechanisms that control cellular proliferation, as well as those related with programmed cell death or apoptosis, require precise regulation systems to prevent diseases such as cancer. Events related to cellular proliferation as well as those associated with apoptosis involve the regulation of gene expression carried out by three basic genetic expression regulation mechanisms: transcription, splicing of the primary transcript for mature mRNA formation, and RNA translation, a ribosomal machinery-dependent process for protein synthesis. While development of each one of these processes requires energy for recognition and assembly of a number of molecular complexes, it has been reported that an increased expression of several members of these protein complexes promotes apoptosis in distinct cell types. The question of how these factors interact with other proteins in order to incorporate themselves into the different transduction cascades and stimulate the development of programmed cell death, although nowadays actively studied, is still waiting for a clear-cut answer. This review focuses on the interactions established between different families of transcription, elongation, translation and splicing factors associated to the progression of apoptosis.
Collapse
Affiliation(s)
- J V Tapia-Vieyra
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México DF, México
| | | | | |
Collapse
|
21
|
Wang YH, Wu MM, Hong CT, Lien LM, Hsieh YC, Tseng HP, Chang SF, Su CL, Chiou HY, Chen CJ. Effects of arsenic exposure and genetic polymorphisms of p53, glutathione S-transferase M1, T1, and P1 on the risk of carotid atherosclerosis in Taiwan. Atherosclerosis 2007; 192:305-312. [PMID: 16973168 DOI: 10.1016/j.atherosclerosis.2006.07.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2005] [Revised: 05/25/2006] [Accepted: 07/25/2006] [Indexed: 11/18/2022]
Abstract
To evaluate the joint effects between genetic polymorphisms of glutathione S-transferase M1, T1, P1, and p53, and arsenic exposure through drinking well water on the risk of carotid atherosclerosis, 605 residents including 289 men and 316 women were recruited from a northeastern area of Taiwan. Carotid atherosclerosis was diagnosed by either a carotid artery intima-media thickness (IMT) of >1.0 mm, a plaque score of > or =1, or stenosis of >50%. A significant age- and gender-adjusted odds ratio of 3.3 for the development of carotid atherosclerosis was observed among the high-arsenic exposure group who drank well water containing arsenic at levels >50 microg/L. The high-arsenic exposure group with GSTP1 variant genotypes of Ile/Val and Val/Val, and with the p53 variant genotypes of Arg/Pro and Pro/Pro had 6.0- and 3.1-fold higher risks of carotid atherosclerosis, respectively. In addition, the high-arsenic exposure group with one or two variant genotypes of GSTP1 and p53 had 2.8- and 6.1-fold higher risks of carotid atherosclerosis, respectively, and showed a dose-dependent relationship. A multivariate-adjusted odds ratio of 3.4 for the risk of carotid atherosclerosis among study subjects with the two variant genotypes of GSTP1 and p53 was also found. Our study showed the joint effects on the risk of carotid atherosclerosis between the genetic polymorphisms of GSTP1 and p53, and arsenic exposure.
Collapse
Affiliation(s)
- Yuan-Hung Wang
- School of Public Health, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Tang F, Liu G, He Z, Ma WY, Bode AM, Dong Z. Arsenite inhibits p53 phosphorylation, DNA binding activity, and p53 target gene p21 expression in mouse epidermal JB6 cells. Mol Carcinog 2006; 45:861-70. [PMID: 16739126 DOI: 10.1002/mc.20245] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Epidemiologic investigations demonstrated that arsenite exposure increases the risk of various human cancers, including skin, lung, bladder, and kidney cancers. However, oral administration of arsenite alone has failed to induce tumors in animal models, suggesting that arsenic may act to enhance mutagenicity induced by other carcinogens. Arsenite may function as a co-carcinogen, acting by inhibiting repair of carcinogen-induced DNA damage mediated by p53 and p21, a p53 target gene. To elucidate the interaction between arsenite and p53 tumor suppressor protein, we studied the effect of arsenite on ultraviolet B (UVB)-induced p53 phosphorylation, p53 DNA binding activity, and p53-induced target gene transactivation in the JB6 Cl41 mouse epidermal skin cell model. Our results indicated that arsenite suppressed UVB-induced p53 phosphorylation and p53 DNA binding activity. Arsenite also inhibited casein kinase 2 (CK2) activity and decreased p53-regulated p21 protein expression. These data suggest that the direct inhibition of p53 functional activation is one of the mechanisms through which arsenite interferes with p53 function, and thus may be a significant mechanism for the co-carcinogenic effects of arsenite.
Collapse
Affiliation(s)
- Faqing Tang
- Hormel Institute, University of Minnesota, Austin, Minnesota 55912, USA
| | | | | | | | | | | |
Collapse
|
23
|
Taylor BF, McNeely SC, Miller HL, Lehmann GM, McCabe MJ, States JC. p53 suppression of arsenite-induced mitotic catastrophe is mediated by p21CIP1/WAF1. J Pharmacol Exp Ther 2006; 318:142-51. [PMID: 16614167 DOI: 10.1124/jpet.106.103077] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Arsenic trioxide, an acute promyelocytic leukemia chemotherapeutic, may be an efficacious treatment for other cancers. Understanding the mechanism as well as genetic and molecular characteristics associated with sensitivity to arsenite-induced cell death is key to providing effective chemotherapeutic usage of arsenite. Arsenite sensitivity correlates with deficient p53 pathways in multiple cell lines. The role of p53 in preventing arsenite-induced mitotic arrest-associated apoptosis (MAAA), a form of mitotic catastrophe, was examined in TR9-7 cells, a model cell line with p53 exogenously regulated in a tetracycline-off expression system. Arsenite activated G1 and G2 cell cycle checkpoints independently of p53, but mitotic catastrophe occurred preferentially in p53- cells. Cyclin B/CDC2(CDK1) stabilization and caspase-3 activation persisted in arsenite-treated p53- cells consistent with MAAA/mitotic catastrophe. N-Benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone, a pan-caspase inhibitor, completely abolished arsenite-induced MAAA/mitotic catastrophe and greatly increased the mitotic index. WEE1 and p21CIP1/WAF1 inhibit cyclin B/CDC2 by CDC2 tyrosine-15 phosphorylation and direct binding, respectively. CDC2-Y15-P was transiently elevated in arsenite-treated p53+ cells but persisted in p53- cells. Arsenite induced p53-S15-P and p21CIP1/WAF1 only in p53+ cells. P21CIP1/WAF1-siRNA-treated p53+ cells were similar to p53- cells in mitotic index and cell cycle protein levels. p53-inducible proteins GADD45alpha and 14-3-3sigma are capable of inhibiting cyclin B/CDC2 but did not play a p53-dependent role in mitotic escape in TR9-7 cells. The data indicate that p53 mediates cyclin B/CDC2 inactivation and mitotic release directly via p21CIP1/WAF1 induction.
Collapse
Affiliation(s)
- B Frazier Taylor
- Department of Pharmacology and Toxicology, University of Louisville, 570 South Preston Street, Suite 221, Louisville, KY 40202, USA
| | | | | | | | | | | |
Collapse
|
24
|
Tapio S, Grosche B. Arsenic in the aetiology of cancer. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2006; 612:215-246. [PMID: 16574468 DOI: 10.1016/j.mrrev.2006.02.001] [Citation(s) in RCA: 195] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2005] [Revised: 01/25/2006] [Accepted: 02/07/2006] [Indexed: 12/28/2022]
Abstract
Arsenic, one of the most significant hazards in the environment affecting millions of people around the world, is associated with several diseases including cancers of skin, lung, urinary bladder, kidney and liver. Groundwater contamination by arsenic is the main route of exposure. Inhalation of airborne arsenic or arsenic-contaminated dust is a common health problem in many ore mines. This review deals with the questions raised in the epidemiological studies such as the dose-response relationship, putative confounders and synergistic effects, and methods evaluating arsenic exposure. Furthermore, it describes the metabolic pathways of arsenic, and its biological modes of action. The role of arsenic in the development of cancer is elucidated in the context of combined epidemiological and biological studies. However, further analyses by means of molecular epidemiology are needed to improve the understanding of cancer aetiology induced by arsenic.
Collapse
Affiliation(s)
- Soile Tapio
- Federal Office for Radiation Protection, Department of Radiation Protection and Health, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany.
| | - Bernd Grosche
- Federal Office for Radiation Protection, Department of Radiation Protection and Health, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| |
Collapse
|
25
|
Wong HK, Fricker M, Wyttenbach A, Villunger A, Michalak EM, Strasser A, Tolkovsky AM. Mutually exclusive subsets of BH3-only proteins are activated by the p53 and c-Jun N-terminal kinase/c-Jun signaling pathways during cortical neuron apoptosis induced by arsenite. Mol Cell Biol 2005; 25:8732-47. [PMID: 16166651 PMCID: PMC1265744 DOI: 10.1128/mcb.25.19.8732-8747.2005] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The c-Jun N-terminal protein kinase (JNK)/c-Jun and p53 pathways form distinct death-signaling modules in neurons that culminate in Bax-dependent apoptosis. To investigate whether this signaling autonomy is due to recruitment of particular BH3-only proteins, we searched for a toxic signal that would activate both pathways in the same set of neurons. We show that arsenite activates both the JNK/c-Jun and p53 pathways in cortical neurons, which together account for >95% of apoptosis, as determined by using the mixed-lineage kinase (JNK/c-Jun) pathway inhibitor CEP11004 and p53-null mice. Despite the coexistence of both pathways in at least 30% of the population, Bim mRNA and protein expression was increased only by the JNK/c-Jun signaling pathway, whereas Noxa and Puma mRNA and Puma protein expression was entirely JNK/c-Jun independent. About 50% of Puma/Noxa expression was p53 dependent, with the remaining signal being independent of both pathways and possibly facilitated by arsenite-induced reduction in P-Akt. However, functionally, Puma was predominant in mediating Bax-dependent apoptosis, as evidenced by the fact that more than 90% of apoptosis was prevented in Puma-null neurons, although Bim was still upregulated, while Bim- and Noxa-null neurons died similarly to wild-type neurons. Thus, the p53 and JNK/c-Jun pathways can activate mutually exclusive subclasses of BH3-only proteins in the same set of neurons. However, other factors besides expression may determine which BH3-only proteins mediate apoptosis.
Collapse
Affiliation(s)
- Hon Kit Wong
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1QW, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
26
|
Sidhu JS, Ponce RA, Vredevoogd MA, Yu X, Gribble E, Hong SW, Schneider E, Faustman EM. Cell cycle inhibition by sodium arsenite in primary embryonic rat midbrain neuroepithelial cells. Toxicol Sci 2005; 89:475-84. [PMID: 16251481 DOI: 10.1093/toxsci/kfj032] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Arsenite (As3+) exposure during development has been associated with neural tube defects and other structural malformations, and with behavioral alterations including altered locomotor activity and operant learning. The molecular mechanisms underlying these effects are uncertain. Because arsenic can cross the placenta and accumulate in the developing neuroepithelium, we examined cell cycling effects of sodium arsenite (As3+ 0, 0.5, 1, 2, and 4 microM) on embryonic primary rat midbrain (gestational day [GD] 12) neuroepithelial cells over 48 h. There was a concentration- and time-dependent As3+-induced reduction in cell viability assessed by neutral red dye uptake assay but minimal apoptosis at concentrations below 4 microM. Morphologically, apoptosis was not apparent until 4 microM at 24 h, which was demonstrated by a marginal but statistically significant increase in cleaved caspase-3/7 activity. Cell cycling effects over several rounds of replication were determined by continuous 5-bromo-2'-deoxyuridine (BrdU) labeling and bivariate flow cytometric Hoechst-Propidium Iodide analysis. We observed a time- and concentration-dependent inhibition of cell cycle progression as early as 12 h after exposure (> or =0.5 microM). In addition, data demonstrated a concentration-dependent increase in cytostasis within all cell cycle phases, a decreased proportion of cells able to reach the second cell cycle, and a reduced cell cycle entry from gap 1 phase (G1). The proportion of affected cells and the severity of the cell cycle perturbation, which ranged from a decreased transition probability to complete cytostasis in all cell cycle phases, were also found to be concentration-dependent. Together, these data support a role for perturbed cell cycle progression in As3+ mediated neurodevelopmental toxicity.
Collapse
Affiliation(s)
- Jaspreet S Sidhu
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Yildirim M, Ikbal M, Tos T, Seven B, Pirim I, Varoglu E. Genotoxicity of thallium-201 in patients with angina pectoris undergoing myocardial perfusion study. TOHOKU J EXP MED 2005; 206:299-304. [PMID: 15997200 DOI: 10.1620/tjem.206.299] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Thallium-201 (201Tl) has been widely used as a nuclear reagent for myocardial blood flow imaging. The purpose of this study was to investigate genotoxic effects of 201Tl in patients with angina pectoris (n = 21), who had undergone myocardial perfusion imaging. Lymphocytes were isolated from each patient before, and 3, 30 and 90 days after 201Tl administration (111 MBq, 3 mCi) and were analyzed for chromosomal aberrations, sister chromatid exchanges, mitotic index and replicative index. There were significant increases in chromosomal aberrations and sister chromatid exchanges 3 days after 201Tl administration (p < 0.001), although no difference was noted in these values after 30 and 90 days (p > 0.05). Moreover, decreased mitotic index and replicative index were noted after 3 days of 201Tl administration (p < 0.001). These results suggest that the administration of 201Tl for myocardial blood flow imaging may induce genetic damage.
Collapse
Affiliation(s)
- Mustafa Yildirim
- Department of Nuclear Medicine, Ataturk University, Medical Faculty, Erzurum, Turkey.
| | | | | | | | | | | |
Collapse
|
28
|
Hernández-Zavala A, Córdova E, Del Razo LM, Cebrián ME, Garrido E. Effects of arsenite on cell cycle progression in a human bladder cancer cell line. Toxicology 2005; 207:49-57. [PMID: 15590121 DOI: 10.1016/j.tox.2004.08.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2004] [Revised: 08/18/2004] [Accepted: 08/20/2004] [Indexed: 11/16/2022]
Abstract
Bladder cancer is one of the most important diseases associated with arsenic (As) exposure in view of its high prevalence and mortality rate. Experimental studies have shown that As exposure induces cell proliferation in the bladder of sodium arsenite (iAsIII) subchronically treated mice. However, there is little available information on its effects on the cell cycle of bladder cells. Thus, our purpose was to evaluate the effects of iAsIII on cell cycle progression and the response of p53 and p21 on the human-derived epithelial bladder cell line HT1197. iAsIII treatment (1-10 microM) for 24 h induced a dose-dependent increase in the proportion of cells in S-phase, which reached 65% at the highest dose. A progressive reduction in cell proliferation was also observed. BrdU was incorporated to cellular DNA in an interrupted form, suggesting an incomplete DNA synthesis. The time-course of iAsIII effects (10 microM) showed an increase in p53 protein content and a transient increase in p21 protein levels accompanying the changes in S-phase. These effects were correlated with iAs concentrations inside the cells, which were not able to metabolize inorganic arsenic. Our findings suggest that p21 was not able to block CDK2-cyclin E complex activity and was therefore unable to arrest cells in G1 allowing their progression into the S-phase. Further studies are needed to ascertain the mechanisms underlying the effects of iAsIII on the G1 to S phase transition in bladder cells.
Collapse
Affiliation(s)
- A Hernández-Zavala
- Sección de Toxicología, CINVESTAV-IPN, Av. Instituto Politécnico Nacional, No. 2508, Col. San Pedro Zacatenco, C.P. 07360, México
| | | | | | | | | |
Collapse
|
29
|
Hsu JL, Huang SY, Chow NH, Chen SH. Stable-isotope dimethyl labeling for quantitative proteomics. Anal Chem 2004; 75:6843-52. [PMID: 14670044 DOI: 10.1021/ac0348625] [Citation(s) in RCA: 554] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In this paper, we report a novel, stable-isotope labeling strategy for quantitative proteomics that uses a simple reagent, formaldehyde, to globally label the N-terminus and epsilon-amino group of Lys through reductive amination. This labeling strategy produces peaks differing by 28 mass units for each derivatized site relative to its nonderivatized counterpart and 4 mass units for each derivatized isotopic pair. This labeling reaction is fast (less than 5 min) and complete without any detectable byproducts based on the analysis of MALDI and LC/ESI-MS/MS spectra of both derivatized and nonderivatized peptide standards and tryptic peptides of hemoglobin molecules. The intensity of the a(1) and y(n-1) ions produced, which were not detectable from most of the nonderivatized fragments, was substantially enhanced upon labeling. We further tested the method based on the analysis of an isotopic pair of peptide standards and a pair of defined protein mixtures with known H/D ratios. Using LC/MS for quantification and LC/MS/MS for peptide sequencing, the results show a negligible isotopic effect, a good mass resolution between the isotopic pair, and a good correlation between the experimental and theoretical data (errors 0-4%). The relative standard deviation of H/D values calculated from peptides deduced from the same protein are less than 13%. The applicability of the method for quantitative protein profiling was also explored by analyzing changes in nuclear protein abundance in an immortalized E7 cell with and without arsenic treatment.
Collapse
Affiliation(s)
- Jue-Liang Hsu
- Department of Chemistry, National Cheng Kung University, No.1 Ta-Hsueh Road, Tainan, 701, Taiwan
| | | | | | | |
Collapse
|
30
|
Gao N, Shen L, Zhang Z, Leonard SS, He H, Zhang XG, Shi X, Jiang BH. Arsenite induces HIF-1alpha and VEGF through PI3K, Akt and reactive oxygen species in DU145 human prostate carcinoma cells. Mol Cell Biochem 2004; 255:33-45. [PMID: 14971644 DOI: 10.1023/b:mcbi.0000007259.65742.16] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Arsenite is widely distributed environmental toxicant in water, food and air. It is a known human carcinogen, which is strongly associated with human cancers originated from liver, nasal cavity, lung, skin, bladder, kidney, and prostate. In this study, we investigated whether arsenite induces expression of hypoxia-inducible factor 1 (HIF-1). HIF-1 is a heterodimeric basic helix-loop-helix transcription factor, composed of HIF-1alpha and HIF-1beta/ARNT subunits; and is involved in tumor growth and angiogenesis. Here we demonstrate that arsenite induces the expression of HIF-1alpha but not HIF-1beta subunit in DU145 human prostate carcinoma cells. Arsenite also increases the expression of VEGF through the induction of HIF-1. We also found that arsenite activates PI3K and Akt that are required for arsenite-induced expression of HIF-1alpha and VEGF. The induction of HIF-1 and VEGF by arsenite can not be inhibited by MAP kinase inhibitors. Arsenite causes production of reactive oxygen species (ROS). The major species of ROS required for the induction of HIF-1 and VEGF is H2O2. These data indicate that the arsenite-induced activation of PI3K/Akt signaling and the expression of HIF-1 and VEGF through the generation of ROS could be an important mechanism in the arsenite-induced carcinogenesis.
Collapse
Affiliation(s)
- Ning Gao
- Mary Babb Randolph Cancer Center, Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV 26506, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Arsenic is a known toxin and carcinogen that is present in industrial settings and in the environment. The mechanisms of disease initiation and progression are not fully understood. In the last a few years, there has been increasing evidence of the correlation between the generation of reactive oxygen species (ROS), DNA damage, tumor promotion, and arsenic exposure. This article summarizes the current literature on the arsenic mediated generation of ROS and reactive nitrogen species (RNS) in various biological systems. This article also discusses the role of ROS and RNS in arsenic-induced DNA damage and activation of oxidative sensitive gene expression.
Collapse
Affiliation(s)
- Honglian Shi
- College of Pharmacy, University of New Mexico, Albuquerque, NM 87131, USA
| | | | | |
Collapse
|
32
|
Abstract
Arsenic is a metalloid compound that is widely distributed in the environment. Human exposure of this compound has been associated with increased cancer incidence. Although the exact mechanisms remain to be investigated, numerous carcinogenic pathways have been proposed. Potential carcinogenic actions for arsenic include oxidative stress, genotoxic damage, DNA repair inhibition, epigenetic events, and activation of certain signal transduction pathways leading to abberrant gene expression. In this article, we summarize current knowledge on the molecular mechanisms of arsenic carcinogenesis with an emphasis on ROS and signal transduction pathways.
Collapse
Affiliation(s)
- Chuanshu Huang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA.
| | | | | | | |
Collapse
|
33
|
Tchounwou PB, Centeno JA, Patlolla AK. Arsenic toxicity, mutagenesis, and carcinogenesis--a health risk assessment and management approach. Mol Cell Biochem 2004; 255:47-55. [PMID: 14971645 DOI: 10.1023/b:mcbi.0000007260.32981.b9] [Citation(s) in RCA: 199] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
A comprehensive analysis of published data indicates that arsenic exposure induces cardiovascular diseases, developmental abnormalities, neurologic and neurobehavioral disorders, diabetes, hearing loss, hematologic disorders, and various types of cancer. Although exposure may occur via the dermal, and parenteral routes, the main pathways of exposure include ingestion, and inhalation. The severity of adverse health effects is related to the chemical form of arsenic, and is also time- and dose-dependent. Recent reports have pointed out that arsenic poisoning appears to be one of the major public health problems of pandemic nature. Acute and chronic exposure to arsenic has been reported in several countries of the world where a large proportion of drinking water (groundwater) is contaminated with high concentrations of arsenic. Research has also pointed significantly higher standardized mortality rates for cancers of the bladder, kidney, skin, liver, and colon in many areas of arsenic pollution. There is therefore a great need for developing a comprehensive health risk assessment (RA) concept that should be used by public health officials and environmental managers for an effective management of the health effects associated with arsenic exposure. With a special emphasis on arsenic toxicity, mutagenesis, and carcinogenesis, this paper is aimed at using the National Academy of Science's RA framework as a guide, for developing a RA paradigm for arsenic based on a comprehensive analysis of the currently available scientific information on its physical and chemical properties, production and use, fate and transport, toxicokinetics, systemic and carcinogenic health effects, regulatory and health guidelines, analytical guidelines and treatment technologies.
Collapse
Affiliation(s)
- Paul B Tchounwou
- Molecular Toxicology Research Laboratory, NIH-Center for Environmental Health, School of Science and Technology, Jackson State University, Jackson, MS 39217, USA.
| | | | | |
Collapse
|
34
|
Shi H, Hudson LG, Liu KJ. Oxidative stress and apoptosis in metal ion-induced carcinogenesis. Free Radic Biol Med 2004; 37:582-93. [PMID: 15288116 DOI: 10.1016/j.freeradbiomed.2004.03.012] [Citation(s) in RCA: 172] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2003] [Accepted: 03/19/2004] [Indexed: 12/25/2022]
Abstract
Epidemiological evidence suggests that exposure to certain metals causes carcinogenesis. The mechanisms of metal-induced carcinogenesis have been pursued in chemical, biochemical, cellular, and animal models. Significant evidence has accumulated that oxidative stress may be a common pathway in cellular responses to exposure to different metals. For example, in the last few years evidence in support of a correlation between the generation of reactive oxygen species, DNA damage, tumor promotion, and arsenic exposure has strengthened. This article summarizes the current literature on metal-mediated oxidative stress, apoptosis, and their relation to metal-mediated carcinogenesis, concentrating on arsenic and chromium.
Collapse
Affiliation(s)
- Honglian Shi
- College of Pharmacy, University of New Mexico, Albuquerque, NM 87131, USA
| | | | | |
Collapse
|
35
|
Tchounwou PB, Patlolla AK, Centeno JA. Carcinogenic and systemic health effects associated with arsenic exposure--a critical review. Toxicol Pathol 2004; 31:575-88. [PMID: 14585726 DOI: 10.1080/01926230390242007] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Arsenic and arsenic containing compounds are human carcinogens. Exposure to arsenic occurs occupationally in several industries, including mining, pesticide, pharmaceutical, glass and microelectronics, as well as environmentally from both industrial and natural sources. Inhalation is the principal route of arsenic exposure in occupational settings, while ingestion of contaminated drinking water is the predominant source of significant environmental exposure globally. Drinking water contamination by arsenic remains a major public health problem. Acute and chronic arsenic exposure via drinking water has been reported in many countries of the world, where a large proportion of drinking water is contaminated with high concentrations of arsenic. General health effects that are associated with arsenic exposure include cardiovascular and peripheral vascular disease, developmental anomalies, neurologic and neurobehavioural disorders, diabetes, hearing loss, portal fibrosis, hematologic disorders (anemia, leukopenia and eosinophilia) and multiple cancers: significantly higher standardized mortality rates and cumulative mortality rates for cancers of the skin, lung, liver, urinary bladder, kidney, and colon in many areas of arsenic pollution. Although several epidemiological studies have documented the sources of exposure and the global impact of arsenic contamination, the mechanisms by which arsenic induces health effects, including cancer, are not well characterized. Further research is needed to provide a better understanding of the pathobiology of arsenic-induced diseases and to better define the toxicologic pathology of arsenic in various organ systems. In this review, we provide and discuss the underlying pathology and nature of arsenic-induced lesions. Such information is critical for understanding the magnitude of health effects associated with arsenic exposure throughout the world.
Collapse
Affiliation(s)
- Paul B Tchounwou
- Molecular Toxicology Research Laboratory, NIH-Center for Environmental Health, School of Science and Technology, Jackson State University, Jackson, Mississippi 39217, USA.
| | | | | |
Collapse
|
36
|
Galicia G, Leyva R, Tenorio EP, Ostrosky-Wegman P, Saavedra R. Sodium arsenite retards proliferation of PHA-activated T cells by delaying the production and secretion of IL-2. Int Immunopharmacol 2003; 3:671-82. [PMID: 12757736 DOI: 10.1016/s1567-5769(03)00049-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Arsenic is a metalloid that commonly contaminates drinking water, and is a known human carcinogen. It has been shown that peripheral blood mononuclear cells (PBMCs) from healthy donors treated in vitro with NaAsO(2) and stimulated with phytohemagglutinin (PHA) show a lower proliferation than nontreated cells. We reported previously a reduction in the secretion of IL-2 in NaAsO(2)-treated PBMCs stimulated with PHA, an observation that might explain, in part, the reduction in proliferation. Since arsenic induces cytoskeleton alterations, which in turn may affect protein transport of the cell, we assumed that NaAsO(2) induced an accumulation of IL-2 inside the cells, and thus a reduction in the secretion of IL-2. In order to demonstrate this hypothesis, we assessed the intracellular IL-2 at the single cell level by flow cytometry, and unexpectedly found a reduction in the percentage of IL-2 producing T cells in the presence of NaAsO(2). We tracked the proliferation of T cells by using the 5,6-carboxyfluorescein diacetate succinimidyl ester (CFSE) dye and found that NaAsO(2) slows down the entrance to cell division and delays the proliferation of cells that have already entered the cell cycle. Nevertheless, the expression of the activation molecules, CD25 and CD69, was unaltered. Assessment of the intracellular and secreted IL-2 in kinetic experiments showed that in fact, NaAsO(2) delays the production of IL-2, given that a recovery of both intracellular and secreted IL-2 was detected at 72 h. Evaluation of the cell cycle showed a higher proportion of cells in G(0)/G(1) and a lower proportion in G(2)/M in the presence of NaAsO(2). We thus conclude that NaAsO(2) reduces proliferation of T cells by delaying the production and secretion of IL-2, thus blocking T cells in G(1); as a consequence, the entry to cell cycle and the rounds of cell division are retarded, and a lower proliferation of T cells is hence observed.
Collapse
Affiliation(s)
- Georgina Galicia
- Departamento de Inmunología y Enfermedades Infecciosas, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Apartado Postal 70228, CU, Mexico City CP 04510, Mexico
| | | | | | | | | |
Collapse
|
37
|
He Z, Ma WY, Liu G, Zhang Y, Bode AM, Dong Z. Arsenite-induced phosphorylation of histone H3 at serine 10 is mediated by Akt1, extracellular signal-regulated kinase 2, and p90 ribosomal S6 kinase 2 but not mitogen- and stress-activated protein kinase 1. J Biol Chem 2003; 278:10588-93. [PMID: 12529330 DOI: 10.1074/jbc.m208581200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Arsenite is known to be an environmental human carcinogen. However, the mechanism of action of this compound in skin carcinogenesis is not completely clear. Here, we provide evidence that arsenite can induce phosphorylation of histone H3 at serine 10 in a time- and dose-dependent manner in JB6 Cl 41 cells. Arsenite induces phosphorylation of Akt1 at serine 473 and increases Akt1 activity. A dominant-negative mutant of Akt1 inhibits the arsenite-induced phosphorylation of histone H3 at serine 10. Additionally, active Akt1 kinase strongly phosphorylates histone H3 at serine 10 in vitro. The arsenite-induced phosphorylation of histone H3 at serine 10 was almost completely blocked by a dominant-negative mutant of extracellular signal-regulated kinase 2 and the mitogen-activated protein kinase/extracellular signal-regulated kinase inhibitor PD98059. N- or C-terminal mutant mitogen- and stress-activated protein kinase 1 or its inhibitor H89 had no effect on arsenite-induced phosphorylation of histone H3 at serine 10 in JB6 Cl 41 cells. However, cells deficient in p90 ribosomal S6 kinase 2 (Rsk2(-/-)) totally block this phosphorylation in a dose- and time-dependent manner. Taken together, these results suggested that arsenite-induced phosphorylation of histone H3 at serine 10 is mediated by Akt1, extracellular signal-regulated kinase 2 and p90 ribosomal S6 kinase 2 but not mitogen- and stress-activated protein kinase 1.
Collapse
Affiliation(s)
- Zhiwei He
- Hormel Institute, University of Minnesota, Austin 55912, USA
| | | | | | | | | | | |
Collapse
|
38
|
Filippova M, Duerksen-Hughes PJ. Inorganic and dimethylated arsenic species induce cellular p53. Chem Res Toxicol 2003; 16:423-31. [PMID: 12641444 DOI: 10.1021/tx025606a] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Arsenic compounds are known for their ability both to cause and to treat human cancers, although the molecular mechanisms underlying these actions are incompletely understood. The simplest explanation is that arsenic causes DNA damage that leads to mutations. However, the majority of scientific evidence indicates that arsenic is not a genotoxin or DNA-damaging agent. DNA damage typically leads to cellular responses designed to minimize the replication of damaged DNA, such as the induction of p53, and p53 induction has therefore been used as an indicator of DNA damage. Because this approach can be applied to human cells and does not rely on a specific, heritable mutation occurring at a particular site, it seemed possible that this method could detect DNA damage that was undetectable using other techniques. To examine the genotoxic potential of arsenic compounds, therefore, seven of these compounds (sodium arsenite, sodium arsenate, methyloxoarsine, iododimethylarsine, disodium methyl arsonate, dimethylarsinic acid, and arsenic trioxide) were tested for their ability to increase the cellular level of p53 as measured by ELISA. Of this group, arsenic trioxide was the strongest inducer of cellular p53, while dimethylarsinic acid, iododimethylarsine, and sodium arsenite also caused p53 induction in a dose- and time-dependent manner. Sodium arsenate, as well as the two monomethyl compounds tested, methyloxoarsine and disodium methyl arsonate, did not cause detectable increases in cellular p53. Our results indicate, therefore, that cells respond to several of these arsenic compounds as they do to chemicals that damage DNA, suggesting that exposure of cells to these compounds does in fact cause DNA damage. Such damage could then result in mutations and the observed development of cancer.
Collapse
Affiliation(s)
- Maria Filippova
- Department of Biochemistry and Microbiology, Center for Molecular Biology and Gene Therapy, Loma Linda University School of Medicine, Loma Linda, California 92354, USA
| | | |
Collapse
|
39
|
Drobná Z, Jaspers I, Thomas DJ, Stýblo M. Differential activation of AP-1 in human bladder epithelial cells by inorganic and methylated arsenicals. FASEB J 2003; 17:67-9. [PMID: 12475910 DOI: 10.1096/fj.02-0287fje] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Chronic exposures to inorganic arsenic (iAs) have been linked to increased incidences of various cancers, including cancer of the urinary bladder. Mechanisms by which iAs promotes cancer may include stimulation of activator protein-1 (AP-1) DNA binding through increased expression and/or phosphorylation of the AP-1 constituents. However, the role of methylated metabolites of iAs in AP-1 activation has not been thoroughly examined. In this study, we show that short-time exposures to 0.1-5 microM arsenite (iAsIII) or the methylated trivalent arsenicals methylarsine oxide (MAsIIIO), or iododimethylarsine (DMAsIIII) induce phosphorylation of c-Jun and increase AP-1 DNA binding activity in human bladder epithelial cells. DMAsIIII and especially MAsIIIO are considerably more potent than iAsIII as inducers of c-Jun phosphorylation and AP-1 activation. Phosphorylated c-Jun, JunB, JunD, and Fra-1, but not c-Fos, FosB, or ATF-2, are detected in the AP-1-DNA binding complex in cells exposed to trivalent arsenicals. In cells transiently transfected with an AP-1-dependent promoter-reporter construct, MAsIIIO was more potent than iAsIII in inducing the AP-1-dependent gene transcription. Exposures to trivalent arsenicals induce phosphorylation of extracellular signal-regulated kinase (ERK), but not c-Jun N-terminal kinases or p38 kinases. These results indicate that an ERK-dependent signal transduction pathway is at least partially responsible for c-Jun phosphorylation and AP-1 activation in UROtsa cells exposed to inorganic or methylated trivalent arsenicals.
Collapse
Affiliation(s)
- Zuzana Drobná
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina 27599-7220, USA
| | | | | | | |
Collapse
|
40
|
Correa I, Cerbón MA, Salazar AM, Solano JD, García-Carrancá A, Quintero A. Differential p53 protein expression level in human cancer-derived cell lines after estradiol treatment. Arch Med Res 2002; 33:455-9. [PMID: 12459315 DOI: 10.1016/s0188-4409(02)00386-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND p53 has a remarkable number of biological activities, including a central role in cell cycle checkpoints, apoptosis, senescence, and maintenance of genomic integrity. Its expression is modified by estradiol in some epithelial cancer-derived cell lines from the reproductive tract. The aim of this study was to evaluate the effect of low and high doses of estradiol in p53 gene expression in epithelial cancer-derived cell lines from the reproductive tract. METHODS p53 gene expression was assessed by Northern and Western blot methods in three human epithelial cancer-derived cell lines after estradiol treatment. RESULTS These indicated that no changes in p53 mRNA content occurred after estradiol treatment at both low (10 nM) and high (1 micro M) doses of estradiol in HeLa, CaLo, and C-33 cell lines. p53 protein content was nearly constant in HeLa and C-33 cell lines at administration of 10 nM of estradiol. However, when estradiol was administered at a higher dose (1 micro M), an increase in p53 protein was observed over time in HeLa and CaLo cell lines. In contrast, estradiol was without variations in C-33. CONCLUSIONS Overall results indicate that estradiol induces variations of p53 protein levels in epithelial cancer-derived cell lines from the reproductive tract in vitro and that this effect may be related with status p53 and/or presence of E6/E7 from human papillomavirus.
Collapse
Affiliation(s)
- Irene Correa
- Unidad de Investigación Médica en Enfermedades Oncológicas, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | | | | | | | | | | |
Collapse
|
41
|
Menéndez D, Bendesky A, Rojas E, Salamanca F, Ostrosky-Wegman P. Role of P53 functionality in the genotoxicity of metronidazole and its hydroxy metabolite. Mutat Res 2002; 501:57-67. [PMID: 11934438 DOI: 10.1016/s0027-5107(02)00012-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
P53 mediates several biological processes for preservation of genetic stability such as the induction of cell cycle arrest, DNA repair or apoptosis in response to DNA damage. The antiparasitic drug, 1-(2-hydroxyethyl)-2-methyl-5-nitroimidazole (metronidazole, MTZ) is able to increase lymphocyte proliferation inducing at the same time chromosomal aberrations. Trying to understand this unexpected event we used cell lines with different P53 functionality, determining the proliferation capacity and the induction of micronuclei (MN) after the treatment with MTZ or its hydroxy metabolite. Our results show that MTZ increased proliferation in a dose response manner in all P53 functional cell lines without inducing changes on the levels of P53 nor MN. However, MTZ hydroxy metabolite induced a dose response increase of P53 and MN, while cell proliferation was not increased. Several studies have shown that the hydroxy metabolite is more potent than MTZ itself. Only in cell lines that do not have a functional P53, MTZ and its metabolite increased both cell proliferation and MN. MTZ use is increasing and its carcinogenicity has not been discarded. Our data indicate that MTZ hydroxy metabolite is potentially a carcinogen and needs to be further studied.
Collapse
Affiliation(s)
- D Menéndez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | | | | | | |
Collapse
|
42
|
States JC, Reiners JJ, Pounds JG, Kaplan DJ, Beauerle BD, McNeely SC, Mathieu P, McCabe MJ. Arsenite disrupts mitosis and induces apoptosis in SV40-transformed human skin fibroblasts. Toxicol Appl Pharmacol 2002; 180:83-91. [PMID: 11969375 DOI: 10.1006/taap.2002.9376] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic ingestion of arsenite-contaminated drinking water causes skin, bladder, and liver cancer. The mechanism of arsenite-induced carcinogenesis is unknown. Arsenite is known to disrupt mitosis and to delay transit through M phase in normal diploid fibroblasts. SV40-transformed human fibroblasts were observed to be hypersensitive to the cytotoxic and cytostatic effects of NaAsO(2) compared with normal diploid fibroblasts in concentration-response experiments. Five to 20 microM NaAsO(2) induced cytostasis in cycling normal diploid fibroblasts but not overt lethality in quiescent normal diploid fibroblasts. High concentrations of arsenite were overtly lethal in both cycling and quiescent cells. The IC50 for cycling SV40-transformed fibroblasts was 3.8 and 4.8 microM for the SV40-transformed lines GM4429 and GM0637, respectively, whereas, in cycling normal diploid fibroblasts (GM0024), the IC50 was 24.7 microM. Microscopic examination of NaAsO(2)-treated SV40-transformed fibroblasts suggested a concentration-dependent accumulation of cells in mitosis undergoing apoptosis. Treatment of SV40-transformed fibroblasts with 0-10 microM NaAsO(2) caused a concentration-dependent inhibition of cell proliferation, accumulation of cells having G2/M DNA contents, and increases in the mitotic index. Phase microscopy, annexin V binding, and electron microscopy demonstrated that arrested mitotic cells underwent apoptosis. These results indicate that SV40-transformation sensitizes cells to arsenite-induced mitotic arrest and induction of apoptosis in the mitotic cells.
Collapse
Affiliation(s)
- J Christopher States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Bode AM, Dong Z. The paradox of arsenic: molecular mechanisms of cell transformation and chemotherapeutic effects. Crit Rev Oncol Hematol 2002; 42:5-24. [PMID: 11923065 DOI: 10.1016/s1040-8428(01)00215-3] [Citation(s) in RCA: 186] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Arsenic is a well-documented carcinogen that also appears to be a valuable therapeutic tool in cancer treatment. This creates a paradox for which no unified hypothesis has been reached regarding the molecular mechanisms that determine whether arsenic will act as a carcinogen or as an effectual chemotherapeutic agent. Much of our knowledge with respect to the actions of arsenic has been drawn from epidemiological or clinical studies. The actions of arsenic are likely to be related to cell type, arsenic species, and length and dose of exposure. Arsenic unquestionably induces apoptosis and may specifically target certain tumor cells. Research data strongly suggest that arsenic influences distinct signaling pathways involved in mediating proliferation or apoptosis, including mitogen-activated protein kinases, p53, activator protein-1 or nuclear factor kappa B. The primary purpose of this review is to examine recent findings, from this laboratory and others, that focus on the molecular mechanisms of arsenic's actions in cell transformation and as a therapeutic agent.
Collapse
Affiliation(s)
- Ann M Bode
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
| | | |
Collapse
|
44
|
Ho IC, Lee TC. Arsenite pretreatment attenuates benzo[a]pyrene cytotoxicity in a human lung adenocarcinoma cell line by decreasing cyclooxygenase-2 levels. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2002; 65:245-263. [PMID: 11911489 DOI: 10.1080/15287390252800846] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Both simultaneous and sequential exposure to arsenite and benzo[a]pyrene (BaP) potentially occur in human populations drinking arsenic-contaminated water or burning arsenic-contaminated coal. Although arsenite and BaP are both well-documented hazardous substances and human carcinogens, interactions between these two agents have not been well defined. In this study, we demonstrated that posttreatment with arsenite synergistically enhanced the cytotoxicity of BaP for a human lung adenocarcinoma cell line, CL3. In contrast, pretreatment of CL3 cells with arsenite attenuated BaP cytotoxicity. Involvement of heat-shock protein 70 and heme oxygenase-1 in this arsenite-mediated attenuation of BaP cytotoxicity was ruled out. Our data also indicated that arsenite pretreatment did not affect the BaP-mediated induction of CYP1A1, the initial enzyme involved in its metabolic activation, but did result in a significant decrease in mRNA and protein levels of cyclooxygenase-2 (COX-2), which is required to convert the BaP metabolite BaP 7,8-dihydrodiol to the ultimate epoxide. In contrast to the high susceptibility of CL3 cells to BaP, the human lung carcinoma cells, H460, and CL3R15 cells (arsenic-resistant CL3 cells) showed normal CYP1A1 inducibility by BaP, had negligible amounts of COX-2, and were highly resistant to BaP. The involvement of COX-2 in BaP activation was confirmed by transfection of H460 cells with a recombinant adenovirus, Ad-pgk-Cox2, coding for COX-2, which resulted in a significant increase in the levels of the COX-2 product prostaglandin E2 in the medium and in the susceptibility of H460 cells to BaP. The present study confirms the importance of COX-2 in BaP activation and demonstrates that the arsenite-mediated attenuation of BaP cytotoxicity is mediated by a reduction in COX-2 levels.
Collapse
Affiliation(s)
- I-Ching Ho
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Republic of China
| | | |
Collapse
|
45
|
Chou RH, Huang H. Sodium arsenite suppresses human papillomavirus-16 E6 gene and enhances apoptosis in E6-transfected human lymphoblastoid cells. J Cell Biochem 2002. [DOI: 10.1002/jcb.10055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
46
|
Rizki M, Kossatz E, Xamena N, Creus A, Marcos R. Influence of sodium arsenite on the genotoxicity of potassium dichromate and ethyl methanesulfonate: studies with the wing spot test in Drosophila. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2002; 39:49-54. [PMID: 11813296 DOI: 10.1002/em.10042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
The wing spot test in Drosophila melanogaster was used to investigate the genotoxicity of arsenic and its effects on the action of two clearly genotoxic agents: potassium dichromate (PDC) and ethyl methanesulfonate (EMS). This assay is based on the principle that the loss of heterozygosity of the suitable recessive markers multiple wing hairs (mwh) and flare-3 (flr(3)) can lead to the formation of mutant clones of larval cells, which are then expressed as spots on the wings of adult flies. These spots can be attributed to different genotoxic events: either mitotic recombination or mutation (deletion, point mutation, and specific types of translocation). Pretreatments and chronic cotreatments were comparatively used for combined treatments. From the results obtained it is evident that sodium arsenite (SA) does not increase the frequency of any of the three categories of spots recorded (small, large, and twin spots) at the concentrations tested. The effects of SA in combination with PDC, in both cotreatments and pretreatments, indicate that SA almost suppressed the clones induced by PDC. Nevertheless, no effects of arsenic were observed with respect to the pre- and cotreatments with EMS. Thus, SA does not modify the frequencies of mutant clones induced by EMS.
Collapse
Affiliation(s)
- Mostapha Rizki
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | | | | | | | | |
Collapse
|
47
|
Vogt BL, Rossman TG. Effects of arsenite on p53, p21 and cyclin D expression in normal human fibroblasts -- a possible mechanism for arsenite's comutagenicity. Mutat Res 2001; 478:159-68. [PMID: 11406180 DOI: 10.1016/s0027-5107(01)00137-3] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Arsenite, the most likely environmental carcinogenic form of arsenic, is not significantly mutagenic at non-toxic concentrations, but is able to enhance the mutagenicity of other agents. Evidence suggests that this comutagenic effect of arsenite is due to inhibition of DNA repair, but no specific repair enzyme has been found to be sensitive to low (<1 microM) concentrations of arsenite. To determine whether arsenite affects signaling which might alter DNA repair, this study assesses the effect of arsenite on p53-related signal transduction pathways after ionizing radiation. Long-term (14 day) low dose (0.1 microM) arsenite caused a modest increase in p53 expression in WI38 normal human fibroblasts, while only toxic (50 microM) concentrations increased p53 levels after short-term (18 h) exposure. When cells were irradiated (6 Gy), p53 and p21 protein concentrations were increased after 4h, as expected. Both long-term, low dose and short-term, high dose exposure to arsenite greatly suppressed the radiation-induced increase in p21 abundance. In addition, long-term, low dose (but not short-term, high dose) exposure to arsenite resulted in increased expression of cyclin D1. These results show that in cells treated with arsenite, p53-dependent increase in p21 expression, normally a block to cell cycle progression after DNA damage, is deficient. At the same time, low (non-toxic) exposure to arsenite enhances positive growth signaling. We suggest that the absence of normal p53 functioning, along with increased positive growth signaling in the presence of DNA damage may result in defective DNA repair and account for the comutagenic effects of arsenite.
Collapse
Affiliation(s)
- B L Vogt
- Nelson Institute of Environmental Medicine and Kaplan Cancer Center, New York University School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987, USA
| | | |
Collapse
|
48
|
Abdullaev FI, Rivera-Luna R, García-Carrancá A, Ayala-Fierro F, Espinosa-Aguirre JJ. Cytotoxic effect of three arsenic compounds in HeLa human tumor and bacterial cells. Mutat Res 2001; 493:31-8. [PMID: 11516713 DOI: 10.1016/s1383-5718(01)00161-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Numerous epidemiological studies suggest that arsenic (As) compounds are carcinogens, however, recent data have renewed the interest in their anticarcinogenic properties. The cytotoxic effects of three arsenic compounds were assessed: sodium arsenite, sodium arsenate and sodium cacodylate, representing the trivalent and pentavalent species of arsenic, along with a dimethylated pentavalent arsenic species. HeLa cells and Salmonella typhimurium (strains TA98 and TA100) were exposed to As compounds and the cytotoxic effects were evaluated. Alterations on RNA and DNA synthesis in HeLa cells were also examined. All arsenic compounds produced a dose-dependent inhibition on colony formation and DNA synthesis in HeLa cells, yet any of them significantly influenced RNA synthesis in these cells. No evidence of arsenic-induced mutagenicity or antimutagenicity was observed using the Ames assay. In bacterial cells, only sodium arsenite caused a dose-dependent inhibition of colony formation.Collectively, these results indicate that in both, HeLa and S. typhimurium cell systems, only trivalent sodium arsenite can act as an effective inhibitor of cell growth. The possible mechanism(s) of the cytotoxic effect of arsenite in these two different cell systems might be due to its reactivity with intracellular sulfhydryl groups.
Collapse
Affiliation(s)
- F I Abdullaev
- Laboratorio Oncologia Experimental, Instituto Nacional de Pediatría, Insurgentes Sur 3700-C, Torre de Investigation, 6 piso, 04530 México D.F., Mexico.
| | | | | | | | | |
Collapse
|
49
|
Kitchin KT. Recent advances in arsenic carcinogenesis: modes of action, animal model systems, and methylated arsenic metabolites. Toxicol Appl Pharmacol 2001; 172:249-61. [PMID: 11312654 DOI: 10.1006/taap.2001.9157] [Citation(s) in RCA: 489] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent advances in our knowledge of arsenic carcinogenesis include the development of rat or mouse models for all human organs in which inorganic arsenic is known to cause cancer-skin, lung, urinary bladder, liver, and kidney. Tumors can be produced from either promotion of carcinogenesis protocols (mouse skin and lungs, rat bladder, kidney, liver, and thyroid) or from complete carcinogenesis protocols (rat bladder and mouse lung). Experiments with p53(+/-) and K6/ODC transgenic mice administered dimethylarsinic acid or arsenite have shown some degree of carcinogenic, cocarcinogenic, or promotional activity in skin or bladder. At present, with the possible exception of skin, the arsenic carcinogenesis models in wild-type animals are more highly developed than in transgenic mice. Recent advances in arsenic metabolism have suggested that methylation of inorganic arsenic may be a toxification, rather than a detoxification, pathway and that trivalent methylated arsenic metabolites, particularly monomethylarsonous acid and dimethylarsinous acid, have a great deal of biological activity. Accumulating evidence indicates that these trivalent, methylated, and relatively less ionizable arsenic metabolites may be unusually capable of interacting with cellular targets such as proteins and even DNA. In risk assessment of environmental arsenic, it is important to know and to utilize both the mode of carcinogenic action and the shape of the dose-response curve at low environmental arsenic concentrations. Although much progress has been recently made in the area of arsenic's possible mode(s) of carcinogenic action, a scientific concensus has not yet been reached. In this review, nine different possible modes of action of arsenic carcinogenesis are presented and discussed-induced chromosomal abnormalities, oxidative stress, altered DNA repair, altered DNA methylation patterns, altered growth factors, enhanced cell proliferation, promotion/progression, gene amplification, and suppression of p53.
Collapse
Affiliation(s)
- K T Kitchin
- Environmental Carcinogenesis Division, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, 27711, USA
| |
Collapse
|
50
|
Bode A, Dong Z. Apoptosis induction by arsenic: mechanisms of action and possible clinical applications for treating therapy-resistant cancers. Drug Resist Updat 2000; 3:21-29. [PMID: 11498362 DOI: 10.1054/drup.2000.0114] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Arsenic, a known carcinogen, may be useful in cancer treatment. Arsenic may be effective in counteracting drug resistance because it appears to induce apoptosis in tumor cells independently of p53 activation, thereby allowing it to be directed against p53-defective cancers. The role of MAP kinases in arsenic-induced apoptosis in tumor cells is important and may be influenced by reactive oxygen species or glutathione. This review focuses on recent findings from this and other laboratories regarding the mechanism(s) of arsenic-induced apoptosis in tumor cells and considers their relevance in the clinical treatment of therapy-resistant cancers. Copyright 2000 Harcourt Publishers Ltd.
Collapse
Affiliation(s)
- Ann Bode
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | | |
Collapse
|