Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 170
Filter
1.
Toxicol Appl Pharmacol ; 397: 115017, 2020 Apr 25.
Article in English | MEDLINE | ID: mdl-32344290

ABSTRACT

CAsE-PE cells are an arsenic-transformed, human prostate epithelial line containing oncogenic mutations in KRAS compared to immortalized, normal KRAS parent cells, RWPE-1. We previously reported increased copy number of mutated KRAS in CAsE-PE cells, suggesting gene amplification. Here, KRAS flanking genomic and transcriptomic regions were sequenced in CAsE-PE cells for insight into KRAS amplification. Comparison of DNA-Seq and RNA-Seq showed increased reads from background aligning to all KRAS exons in CAsE-PE cells, while a uniform DNA-Seq read distribution occurred in RWPE-1 cells with normal transcript expression. We searched for KRAS fusions in DNA and RNA sequencing data finding a portion of reads aligning to KRAS and viral sequence. After generation of cDNA from total RNA, short and long KRAS probes were generated to hybridize cDNA and KRAS enriched fragments were PacBio sequenced. More KRAS reads were captured from CAsE-PE cDNA versus RWPE-1 by each probe set. Only CAsE-PE cDNA showed KRAS viral fusion transcripts, primarily mapping to LTR and endogenous retrovirus sequences on either 5'- or 3'-ends of KRAS. Most KRAS viral fusion transcripts contained 4 to 6 exons but some PacBio sequences were in unusual orientations, suggesting viral insertions within the gene body. Additionally, conditioned media was extracted for potential retroviral particles. RNA-Seq of culture media isolates identified KRAS retroviral fusion transcripts in CAsE-PE media only. Truncated KRAS transcripts suggested multiple retroviral integration sites occurred within the KRAS gene producing KRAS retroviral fusions of various lengths. Findings suggest activation of endogenous retroviruses in arsenic carcinogenesis should be explored.

2.
Toxicol Appl Pharmacol ; 312: 11-18, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-26721309

ABSTRACT

Inorganic arsenic, an environmental contaminant and a human carcinogen is associated with prostate cancer. Emerging evidence suggests that cancer stem cells (CSCs) are the driving force of carcinogenesis. Chronic arsenic exposure malignantly transforms the human normal prostate stem/progenitor cell (SC) line, WPE-stem to arsenic-cancer SCs (As-CSCs), through unknown mechanisms. MicroRNAs (miRNAs) are small, non-coding RNAs that negatively regulate gene expression at the posttranscriptional level. In prior work, miR-143 was markedly downregulated in As-CSCs, suggesting a role in arsenic-induced malignant transformation. In the present study, we investigated whether loss of miR-143 expression is important in arsenic-induced transformation of prostate SCs. Restoration of miR-143 in As-CSCs was achieved by lentivirus-mediated miR-143 overexpression. Cells were assessed bi-weekly for up to 30weeks to examine mitigation of cancer phenotype. Secreted matrix metalloproteinase (MMP) activity was increased by arsenic-induced malignant transformation, but miR-143 restoration decreased secreted MMP-2 and MMP-9 enzyme activities compared with scramble controls. Increased cell proliferation and apoptotic resistance, two hallmarks of cancer, were decreased upon miR-143 restoration. Increased apoptosis was associated with decreased BCL2 and BCL-XL expression. miR-143 restoration dysregulated the expression of SC/CSC self-renewal genes including NOTCH-1, BMI-1, OCT4 and ABCG2. The anticancer effects of miR-143 overexpression appeared to be mediated by targeting and inhibiting LIMK1 protein, and the phosphorylation of cofilin, a LIMK1 substrate. These findings clearly show that miR-143 restoration mitigated multiple cancer characteristics in the As-CSCs, suggesting a potential role in arsenic-induced transformation of prostate SCs. Thus, miR-143 is a potential biomarker and therapeutic target for arsenic-induced prostate cancer.


Subject(s)
Arsenic/toxicity , Carcinogens/toxicity , MicroRNAs/genetics , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/chemically induced , Cell Line, Tumor , Humans , Lim Kinases/metabolism , Male , Matrix Metalloproteinases/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
3.
Chem Res Toxicol ; 29(9): 1458-67, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27510461

ABSTRACT

Cadmium (Cd) is a potential human prostate carcinogen. Chronic Cd exposure malignantly transforms RWPE-1 human prostate epithelial cells into CTPE cells by an unclear mechanism. Previous studies show that RWPE-1 can also be malignantly transformed by arsenic, and KRAS activation is key to causation and maintenance of this phenotype. Although Cd and arsenic can both transform prostate epithelial cells, it is uncertain whether their mechanisms are similar. Thus, here we determined whether KRAS activation is critical in causing and maintaining Cd-induced malignant transformation in CTPE cells. Expression of KRAS, miRNAs, and other genes of interest was analyzed by Western blot and RT-PCR. Following stable KRAS knockdown (KD) by RNA interference using shRNAmir, the malignant phenotype was assessed by various physical and genetic parameters. CTPE cells greatly overexpressed KRAS by 20-fold, indicating a likely role in Cd transformation. Thus, we attempted to reverse the malignant phenotype via KRAS KD. Two weeks after shRNAmir transduction, KRAS protein was undetectable in CTPE KD cells, confirming stable KD. KRAS KD reduced stimulated RAS/ERK and PI3K/AKT signaling pathways and markedly mitigated multiple physical and molecular malignant cell characteristics including: hypersecretion of MMP-2, colony formation, cell survival, and expression of cancer-relevant genes (reduced proliferation and cell cycle-related genes; activated tumor suppressor PTEN). However, KRAS KD did not reverse miRNA expression originally down-regulated by Cd transformation. These data strongly suggest KRAS is a key gene in development and maintenance of the Cd-induced malignant phenotype, at least in the prostate. It is not, however, the only genetic factor sustaining this phenotype.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epithelial Cells/physiology , Gene Silencing , Prostate/physiopathology , Prostatic Neoplasms/physiopathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Cadmium/chemistry , Cadmium/toxicity , Cell Line , Cell Proliferation/genetics , Cell Transformation, Neoplastic/drug effects , Epithelial Cells/pathology , Gene Expression/genetics , Gene Knockdown Techniques , Humans , Male , Signal Transduction/genetics
4.
Toxicol Appl Pharmacol ; 282(3): 267-74, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25485709

ABSTRACT

We studied how protein metallothionein (MT) impacts arsenic-induced oxidative DNA damage (ODD) using cells that poorly express MT (MT-I/II double knockout embryonic cells; called MT-null cells) and wild-type (WT) MT competent cells. Arsenic (as NaAsO2) was less cytolethal over 24h in WT cells (LC50=11.0±1.3µM; mean±SEM) than in MT-null cells (LC50=5.6±1.2µM). ODD was measured by the immuno-spin trapping method. Arsenic (1 or 5µM; 24h) induced much less ODD in WT cells (121% and 141% of control, respectively) than in MT-null cells (202% and 260%). In WT cells arsenic caused concentration-dependent increases in MT expression (transcript and protein), and in the metal-responsive transcription factor-1 (MTF-1), which is required to induce the MT gene. In contrast, basal MT levels were not detectable in MT-null cells and unaltered by arsenic exposure. Transfection of MT-I gene into the MT-null cells markedly reduced arsenic-induced ODD levels. The transport genes, Abcc1 and Abcc2 were increased by arsenic in WT cells but either showed no or very limited increases in MT-null cells. Arsenic caused increases in oxidant stress defense genes HO-1 and GSTα2 in both WT and MT-null cells, but to much higher levels in WT cells. WT cells appear more adept at activating metal transport systems and oxidant response genes, although the role of MT in these responses is unclear. Overall, MT protects against arsenic-induced ODD in MT competent cells by potential sequestration of scavenging oxidant radicals and/or arsenic.


Subject(s)
Arsenic/toxicity , Carcinogens/toxicity , DNA Damage , Environmental Pollutants/toxicity , Metallothionein/genetics , Animals , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Damage/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation/drug effects , Glutathione Transferase/genetics , Heme Oxygenase-1/genetics , Isoenzymes/genetics , Membrane Proteins/genetics , Metallothionein/metabolism , Mice , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Oxidative Stress , Transcription Factors/genetics , Transcription Factor MTF-1
5.
Toxicol Appl Pharmacol ; 286(3): 159-67, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25922126

ABSTRACT

Previous work shows altered methylation patterns in inorganic arsenic (iAs)- or cadmium (Cd)-transformed epithelial cells. Here, the methylation status near the transcriptional start site was assessed in the normal human prostate epithelial cell line (RWPE-1) that was malignantly transformed by 10µM Cd for 11weeks (CTPE) or 5µM iAs for 29weeks (CAsE-PE), at which time cells showed multiple markers of acquired cancer phenotype. Next generation sequencing of the transcriptome of CAsE-PE cells identified multiple dysregulated genes. Of the most highly dysregulated genes, five genes that can be relevant to the carcinogenic process (S100P, HYAL1, NTM, NES, ALDH1A1) were chosen for an in-depth analysis of the DNA methylation profile. DNA was isolated, bisulfite converted, and combined bisulfite restriction analysis was used to identify differentially methylated CpG sites, which was confirmed with bisulfite sequencing. Four of the five genes showed differential methylation in transformants relative to control cells that was inversely related to altered gene expression. Increased expression of HYAL1 (>25-fold) and S100P (>40-fold) in transformants was correlated with hypomethylation near the transcriptional start site. Decreased expression of NES (>15-fold) and NTM (>1000-fold) in transformants was correlated with hypermethylation near the transcriptional start site. ALDH1A1 expression was differentially expressed in transformed cells but was not differentially methylated relative to control. In conclusion, altered gene expression observed in Cd and iAs transformed cells may result from altered DNA methylation status.


Subject(s)
Arsenic/toxicity , Cadmium/toxicity , DNA Methylation/drug effects , Epithelial Cells/drug effects , Prostate/drug effects , Prostate/physiology , Transcription, Genetic/drug effects , Cell Line , Cell Line, Transformed , DNA Methylation/physiology , Epithelial Cells/pathology , Epithelial Cells/physiology , Humans , Male , Prostate/pathology , Transcription, Genetic/physiology
6.
Toxicol Appl Pharmacol ; 286(1): 36-43, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25804888

ABSTRACT

Inorganic arsenic is a human lung carcinogen. We studied the ability of chronic inorganic arsenic (2 µM; as sodium arsenite) exposure to induce a cancer phenotype in the immortalized, non-tumorigenic human lung peripheral epithelial cell line, HPL-1D. After 38 weeks of continuous arsenic exposure, secreted matrix metalloproteinase-2 (MMP2) activity increased to over 200% of control, levels linked to arsenic-induced cancer phenotypes in other cell lines. The invasive capacity of these chronic arsenic-treated lung epithelial (CATLE) cells increased to 320% of control and colony formation increased to 280% of control. CATLE cells showed enhanced proliferation in serum-free media indicative of autonomous growth. Compared to control cells, CATLE cells showed reduced protein expression of the tumor suppressor gene PTEN (decreased to 26% of control) and the putative tumor suppressor gene SLC38A3 (14% of control). Morphological evidence of epithelial-to-mesenchymal transition (EMT) occurred in CATLE cells together with appropriate changes in expression of the EMT markers vimentin (VIM; increased to 300% of control) and e-cadherin (CDH1; decreased to 16% of control). EMT is common in carcinogenic transformation of epithelial cells. CATLE cells showed increased KRAS (291%), ERK1/2 (274%), phosphorylated ERK (p-ERK; 152%), and phosphorylated AKT1 (p-AKT1; 170%) protein expression. Increased transcript expression of metallothioneins, MT1A and MT2A and the stress response genes HMOX1 (690%) and HIF1A (247%) occurred in CATLE cells possibly in adaptation to chronic arsenic exposure. Thus, arsenic induced multiple cancer cell characteristics in human peripheral lung epithelial cells. This model may be useful to assess mechanisms of arsenic-induced lung cancer.


Subject(s)
Arsenic/toxicity , Carcinogens/toxicity , Epithelial Cells/drug effects , Adenocarcinoma , Adenocarcinoma of Lung , Cell Line, Tumor , Cells, Cultured , Epithelial Cells/physiology , Epithelial-Mesenchymal Transition/drug effects , Humans , Lung Neoplasms , Metallothionein/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phenotype , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , ras Proteins/metabolism
7.
Arch Biochem Biophys ; 583: 27-35, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26253262

ABSTRACT

Previous studies have linked cadmium exposure to disturbances in carbohydrate and lipid metabolism. In this study we investigate the effects in Wistar rats of an oral cadmium exposure in drinking water on carbohydrates, lipids and insulin release. Also, using mathematical models we studied the effect of cadmium on insulin resistance and sensitivity in liver, muscle, adipose and cardiovascular tissue. Cadmium exposure induced hyperglycemia, increased insulin release after a glucose load, and caused increases in serum triglycerides, cholesterol, LDL-C and VLDL-C, and a decrease of HDL-C. In addition, there was an accumulation of cadmium in pancreas and an increase of insulin. After exposure, HOMA-IR was increased, while the HOMA-S%, QUICKI and Matsuda-DeFronzo indexes showed decreases. A decrease of insulin sensitivity was shown in muscle and liver. Additionally, cadmium increases insulin resistance in the liver, adipose tissue and cardiovascular system. Finally, ß-cell functioning was evaluated by HOMA-B% index and insulin disposition index, which were decreased, while insulin generation index increased. In conclusion, cadmium increases insulin release, induces hyperglycemia and alters lipid metabolism. These changes likely occur as a consequence of reduced sensitivity and increased insulin resistance in multiple insulin-dependent and non-dependent tissues, producing a biochemical phenotype similar to metabolic syndrome and diabetes.


Subject(s)
Cadmium/toxicity , Insulin Resistance , Pancreas/drug effects , Adipose Tissue/physiopathology , Animals , Cardiovascular System/physiopathology , Liver/physiopathology , Male , Muscles/physiopathology , Pancreas/physiopathology , Rats , Rats, Wistar , Toxicity Tests, Chronic
8.
Arch Toxicol ; 88(2): 249-61, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24091636

ABSTRACT

Inorganic arsenic (iAs) and its toxic methylated metabolite, methylarsonous acid (MMA(III)), both have carcinogenic potential. Prior study shows iAs-induced malignant transformation in both arsenic methylation-proficient (liver) and methylation-deficient (prostate) cells, but only methylation-proficient cells show oxidative DNA damage (ODD) during this transformation. To further define whether arsenic methylation is necessary for transformation or ODD induction, here we chronically exposed these same liver or prostate cell lines to MMA(III) (0.25-1.0 µM) and tested for acquired malignant phenotype. Various metrics of oncogenic transformation were periodically assessed along with ODD during chronic MMA(III) exposure. Methylation-deficient and methylation-proficient cells both acquired a cancer phenotype with MMA(III) exposure at about 20 weeks, based on increased matrix metalloproteinase secretion, colony formation, and invasion. In contrast, prior work showed iAs-induced transformation took longer in biomethylation-deficient cells (~30 weeks) than in biomethylation-proficient cells (~18 weeks). In the present study, MMA(III) caused similar peak ODD levels at similar concentrations and at similar exposure times (18-22 weeks) in both cell types. At the approximate peak of ODD production, both cell types showed similar alterations in arsenic and oxidative stress adaptation factors (i.e., ABCC1, ABCC2, GST-π, SOD-1). Thus, MMA(III) causes oncogenic transformation associated with ODD in methylation-deficient cells, indicating that further methylation is not required to induce ODD. Together, these results show that MMA(III) and iAs cause an acquired malignant phenotype in methylation-deficient cells, yet iAs does not induce ODD. This indicates iAs likely has both genotoxic and non-genotoxic mechanisms dictated by the target cell's ability to methylate arsenic.


Subject(s)
Arsenicals/pharmacology , Cell Transformation, Neoplastic/chemically induced , DNA Damage/drug effects , Adaptation, Physiological/drug effects , Adaptation, Physiological/physiology , Animals , Arsenic Poisoning/pathology , Cell Line/drug effects , Humans , Liver/cytology , Liver/drug effects , Male , Methylation , Multidrug Resistance-Associated Protein 2 , Oxidation-Reduction , PTEN Phosphohydrolase/genetics , Prostate/cytology , Prostate/metabolism , Rats , Toxicity Tests, Chronic
9.
Arch Toxicol ; 88(2): 263-74, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24068038

ABSTRACT

Accumulating data suggest arsenic may be an endocrine disruptor and tentatively linked to breast cancer by some studies. Therefore, we tested the effects of chronic inorganic arsenic exposure on the normal estrogen receptor (ER)-negative breast epithelial cell line, MCF-10A. Cells were chronically exposed to a low-level arsenite (500 nM) for up to 24 weeks. Markers of cancer cell phenotype and the expression of critical genes relevant to breast cancer or stem cells (SCs) were examined. After 24 weeks, chronic arsenic-exposed breast epithelial (CABE) cells showed increases in secreted MMP activity, colony formation, invasion, and proliferation rate, indicating an acquired cancer cell phenotype. These CABE cells presented with basal-like breast cancer characteristics, including ER-α, HER-2, and progesterone receptor negativity, and overexpression of K5 and p63. Putative CD44(+)/CD24(-/low) breast SCs were increased to 80 % over control in CABE cells. CABE cells also formed multilayer cell mounds, indicative of loss of contact inhibition. These mounds showed high levels of K5 and p63, indicating the potential presence of cancer stem cells (CSCs). Epithelial-to-mesenchymal transition occurred during arsenic exposure. Overexpression of aromatase, a key rate-limiting enzyme in estrogen synthesis, occurred with arsenic starting early on in exposure. Levels of 17ß-estradiol increased in CABE cells and their conditioned medium. The aromatase inhibitor letrozole abolished arsenic-induced increases in 17ß-estradiol production and reversed cancer cell phenotype. Thus, chronic arsenic exposure drives human breast epithelia into a cancer cell phenotype with an apparent overabundance of putative CSCs. Arsenic appears to transform breast epithelia through overexpression of aromatase, thereby activating oncogenic processes independent of ER.


Subject(s)
Aromatase/metabolism , Arsenic/toxicity , Breast Neoplasms/chemically induced , Mammary Glands, Human/cytology , Receptors, Estrogen/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line/drug effects , Cell Transformation, Neoplastic/chemically induced , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Female , Humans , Mammary Glands, Human/drug effects , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplastic Stem Cells/drug effects , Toxicity Tests, Chronic
10.
Arch Toxicol ; 88(8): 1619-29, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25005685

ABSTRACT

In mice, inorganic arsenic in the drinking water in the parts per million range via the dam during in utero life or with whole-life exposure is a multi-site carcinogen in the offspring. However, human arsenic exposure is typically in the parts per billion (ppb) range. Thus, we studied "whole-life" inorganic arsenic carcinogenesis in mice at levels more relevant to humans. Breeder male and female CD1 mice were exposed to 0, 50, 500 or 5,000 ppb arsenic (as sodium arsenite) in the drinking water for 3 weeks prior to breeding, during pregnancy and lactation, and after weaning (at week 3) groups of male and female offspring (initial n = 40) were exposed for up to 2 years. Tumors were assessed in these offspring. Arsenic exposure had no effect on pregnant dam weights or water consumption, litter size, offspring birthweight or weight at weaning compared to control. In male offspring mice, arsenic exposure increased (p < 0.05) bronchiolo-alveolar tumor (adenoma or carcinoma) incidence at 50-ppb group (51 %) and 500-ppb group (54 %), but not at 5,000-ppb group (28 %) compared to control (22 %). These arsenic-induced bronchiolo-alveolar tumors included increased (p < 0.05) carcinoma at 50-ppb group (27 %) compared to controls (8 %). An increase (p < 0.05) in lung adenoma (25 %) in the 50-ppb group compared to control (11 %) occurred in female offspring. Thus, in CD1 mice whole-life arsenic exposure induced lung tumors at human-relevant doses (i.e., 50 and 500 ppb).


Subject(s)
Aging/drug effects , Arsenites/toxicity , Environmental Pollutants/toxicity , Lung Neoplasms/chemically induced , Prenatal Exposure Delayed Effects/chemically induced , Sodium Compounds/toxicity , Aging/pathology , Animals , Carcinogenicity Tests , Dose-Response Relationship, Drug , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/pathology , Male , Mice , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Species Specificity , Survival Analysis , Time Factors
11.
J Hepatol ; 58(4): 778-84, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23207144

ABSTRACT

BACKGROUND & AIMS: Progression from steatosis to steatohepatitic lesions is hypothesized to require a second hit. These lesions have been associated with increased oxidative stress, often ascribed to high levels of leptin and other proinflammatory mediators. Here we have examined the role of leptin in inducing oxidative stress and Kupffer cell activation in CCl4-mediated steatohepatitic lesions of obese mice. METHODS: Male C57BL/6 mice fed with a high-fat diet (60%kcal) at 16 weeks were administered CCl4 to induce steatohepatitic lesions. Approaches included use of immuno-spin trapping for measuring free radical stress, gene-deficient mice for leptin, p47 phox, iNOS and adoptive transfer of leptin primed macrophages in vivo. RESULTS: Diet-induced obese (DIO) mice, treated with CCl4 increased serum leptin levels. Oxidative stress was significantly elevated in the DIO mouse liver, but not in ob/ob mice, or in DIO mice treated with leptin antibody. In ob/ob mice, leptin supplementation restored markers of free radical generation. Markers of free radical formation were significantly decreased by the peroxynitrite decomposition catalyst FeTPPS, the iNOS inhibitor 1400W, the NADPH oxidase inhibitor apocynin, or in iNOS or p47 phox-deficient mice. These results correlated with the decreased expression of TNF-alpha and MCP-1. Kupffer cell depletion eliminated oxidative stress and inflammation, whereas in macrophage-depleted mice, the adoptive transfer of leptin-primed macrophages significantly restored inflammation. CONCLUSIONS: These results, for the first time, suggest that leptin action in macrophages of the steatotic liver, through induction of iNOS and NADPH oxidase, causes peroxynitrite-mediated oxidative stress thus activating Kupffer cells.


Subject(s)
Fatty Liver/metabolism , Kupffer Cells/metabolism , Leptin/metabolism , Oxidative Stress , Animals , Cytokines/metabolism , Disease Models, Animal , Fatty Liver/etiology , Inflammation Mediators/metabolism , Kupffer Cells/pathology , Male , Mice , Mice, Inbred C57BL , NADPH Oxidases/metabolism , Nitric Oxide Synthase Type II/metabolism , Non-alcoholic Fatty Liver Disease , Obesity/complications , Peroxynitrous Acid/metabolism
12.
Toxicol Appl Pharmacol ; 269(3): 297-306, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23438451

ABSTRACT

Today's developed world faces a major public health challenge in the rise in the obese population and the increased incidence in fatty liver disease. There is a strong association among diet induced obesity, fatty liver disease and development of nonalcoholic steatohepatitis but the environmental link to disease progression remains unclear. Here we demonstrate that in obesity, early steatohepatitic lesions induced by the water disinfection byproduct bromodichloromethane are mediated by increased oxidative stress and leptin which act in synchrony to potentiate disease progression. Low acute exposure to bromodichloromethane (BDCM), in diet-induced obesity produced oxidative stress as shown by increased lipid peroxidation, protein free radical and nitrotyrosine formation and elevated leptin levels. Exposed obese mice showed histopathological signs of early steatohepatitic injury and necrosis. Spontaneous knockout mice for leptin or systemic leptin receptor knockout mice had significantly decreased oxidative stress and TNF-α levels. Co-incubation of leptin and BDCM caused Kupffer cell activation as shown by increased MCP-1 release and NADPH oxidase membrane assembly, a phenomenon that was decreased in Kupffer cells isolated from leptin receptor knockout mice. In obese mice that were BDCM-exposed, livers showed a significant increase in Kupffer cell activation marker CD68 and, increased necrosis as assessed by levels of isocitrate dehydrogenase, events that were decreased in the absence of leptin or its receptor. In conclusion, our results show that exposure to the disinfection byproduct BDCM in diet-induced obesity augments steatohepatitic injury by potentiating the effects of leptin on oxidative stress, Kupffer cell activation and cell death in the liver.


Subject(s)
Adipokines/pharmacology , Fatty Liver/chemically induced , Obesity/complications , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Fatty Liver/prevention & control , In Situ Nick-End Labeling , Kupffer Cells/drug effects , Leptin/analysis , Liver/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Microscopy, Confocal , Real-Time Polymerase Chain Reaction , Trihalomethanes/antagonists & inhibitors , Trihalomethanes/toxicity
13.
Toxicol Appl Pharmacol ; 273(2): 281-8, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-23811327

ABSTRACT

Cadmium is a known human lung carcinogen. Here, we attempt to develop an in vitro model of cadmium-induced human lung carcinogenesis by chronically exposing the peripheral lung epithelia cell line, HPL-1D, to a low level of cadmium. Cells were chronically exposed to 5 µM cadmium, a noncytotoxic level, and monitored for acquired cancer characteristics. By 20 weeks of continuous cadmium exposure, these chronic cadmium treated lung (CCT-LC) cells showed marked increases in secreted MMP-2 activity (3.5-fold), invasion (3.4-fold), and colony formation in soft agar (2-fold). CCT-LC cells were hyperproliferative, grew well in serum-free media, and overexpressed cyclin D1. The CCT-LC cells also showed decreased expression of the tumor suppressor genes p16 and SLC38A3 at the protein levels. Also consistent with an acquired cancer cell phenotype, CCT-LC cells showed increased expression of the oncoproteins K-RAS and N-RAS as well as the epithelial-to-mesenchymal transition marker protein Vimentin. Metallothionein (MT) expression is increased by cadmium, and is typically overexpressed in human lung cancers. The major MT isoforms, MT-1A and MT-2A were elevated in CCT-LC cells. Oxidant adaptive response genes HO-1 and HIF-1A were also activated in CCT-LC cells. Expression of the metal transport genes ZNT-1, ZNT-5, and ZIP-8 increased in CCT-LC cells culminating in reduced cadmium accumulation, suggesting adaptation to the metal. Overall, these data suggest that exposure of human lung epithelial cells to cadmium causes acquisition of cancer cell characteristics. Furthermore, transformation occurs despite the cell's ability to adapt to chronic cadmium exposure.


Subject(s)
Cadmium/toxicity , Cell Transformation, Neoplastic/chemically induced , Lung Neoplasms/chemically induced , Lung/drug effects , Respiratory Mucosa/drug effects , Cadmium/administration & dosage , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , DNA Damage/drug effects , DNA Damage/physiology , Humans , Lung/metabolism , Lung/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Time Factors , Tumor Cells, Cultured
14.
Chem Res Toxicol ; 26(1): 96-105, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23137061

ABSTRACT

Inorganic arsenic in the drinking water is a multisite human carcinogen that potentially targets the kidney. Recent evidence also indicates that developmental arsenic exposure impacts renal carcinogenesis in humans and mice. Emerging theory indicates that cancer may be a disease of stem cells (SCs) and that there are abundant active SCs during early life. Therefore, we hypothesized that inorganic arsenic targets SCs, or partially differentiated progenitor cells (PCs), for oncogenic transformation. Thus, a rat kidney SC/PC cell line, RIMM-18, was chronically exposed to low-level arsenite (500 nM) for up to 28 weeks. Multiple markers of acquired cancer phenotype were assessed biweekly during arsenic exposure, including secreted matrix metalloproteinase (MMP) activity, proliferation rate, colony formation in soft agar, and cellular invasiveness. Arsenic exposure by 10 weeks and after also induced marked and sustained increases in colony formation, indicative of the loss of contact inhibition, and increased invasiveness, both cancer cell characteristics. Compared to the passage-matched control, chronic arsenic exposure caused exposure-duration dependent increases in secreted MMP-2 and MMP-9 activity, Cox-2 expression, and more rapid proliferation (all >2-fold), characteristics typical of cancer cells. Dysregulation of SC maintenance genes and signaling pathways are common during oncogenesis. During arsenite exposure, expression of several genes associated with normal kidney development and SC regulation and differentiation (i.e., Wt-1, Wnt-4, Bmp-7, etc.) were aberrantly altered. Arsenic-exposed renal SCs produced more nonadherent spheroid bodies that grew much more aggressively in Matrigel, typical of cancer SCs (CSCs). The transformed cells also showed gene overexpression typical of renal SCs/CSCs (CD24, Osr1, Ncam) and arsenic adaptation such as overexpression of Mt-1, Mt2, Sod-1, and Abcc2. These data suggest that inorganic arsenic induced an acquired cancer phenotype in vitro in these rat kidney SCs potentially forming CSCs and, consistent with data in vivo, indicate that these multipotent SCs may be targets of arsenic during renal carcinogenesis.


Subject(s)
Arsenic/toxicity , Stem Cells/drug effects , AC133 Antigen , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , CD24 Antigen/genetics , CD24 Antigen/metabolism , Cell Line , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cyclooxygenase 2/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Kidney/cytology , Kidney/metabolism , Kidney Neoplasms/etiology , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Metallothionein/genetics , Metallothionein/metabolism , Mice , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Peptides/genetics , Peptides/metabolism , Rats , Stem Cells/cytology , Stem Cells/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
15.
Arch Toxicol ; 87(2): 311-21, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22914987

ABSTRACT

The role of metallothionein (MT) in mitigation of oxidative DNA damage (ODD) induced by either cadmium (Cd) or the direct oxidant hydrogen peroxide (H(2)O(2)) was systematically examined using MT-I/II double knockout (MT-null) or MT-competent wild-type (WT) cells. Both toxicants were much more lethal to MT-null cells (Cd LC(50) = 6.6 µM; H(2)O(2) LC(50) = 550 µM) than to WT cells (Cd LC(50) = 16.5 µM; H(2)O(2) LC(50) = 930 µM). Cd induced concentration-related MT increases in WT cells, while the basal levels were undetectable and not increased by Cd in MT-null cells. ODD, measured by the immuno-spin trapping method, was minimally induced by sub-toxic Cd levels (1 or 5 µM; 24 h) in WT cells, but markedly increased in MT-null cells (>430 %). Similarly, ODD was induced to higher levels by lower concentrations of H(2)O(2) in MT-null cells than WT cells. Transfection of MT-I into MT-null cells reduced both Cd- and H(2)O(2)-induced cytolethality and ODD. Cd increased the expression of the oxidant defense genes, HO-1, and GSTa2 to a much greater extent in MT-null cells than in WT. Cd or H(2)O(2) exposure increased the expression of key transport genes, Mrp1 and Mrp2, in WT cells but not in MT-null cells. MT protects against Cd- and H(2)O(2)-induced ODD in MT-competent cells possibly by multiple mechanisms, potentially including direct metal ion sequestration and sequestration of oxidant radicals by MT. MT-deficient cells appear to adapt to Cd primarily by turning on oxidant response systems, while MT-competent cells activate MT and transport systems.


Subject(s)
DNA Damage , Embryonic Stem Cells/drug effects , Metallothionein/metabolism , Oxidants/toxicity , Oxidative Stress/drug effects , Animals , Cadmium Chloride/toxicity , Cell Line , Cell Survival/drug effects , Embryonic Stem Cells/metabolism , Gene Expression/drug effects , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Glutathione Transferase/genetics , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hydrogen Peroxide/toxicity , Isoenzymes/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Metallothionein/deficiency , Metallothionein/genetics , Mice , Mice, Knockout , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Transfection
16.
Toxicol Mech Methods ; 23(6): 389-95, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23301828

ABSTRACT

The carcinogen inorganic arsenic (iAs) undergoes biomethylation (BMT) in some cells. The methylated metabolite, monomethylarsonous (MMA(3+)), may cause oxidative DNA damage (ODD). With chronic iAs exposure, BMT-competent cells show ODD while BMT-deficient do not. To further define these events, we studied ODD produced by acute iAs or MMA(3+) in the BMT-deficient human prostate cell line, RWPE-1. ODD, measured by the immuno-spin trapping method, was assessed after exposure to iAs or MMA(3+) alone, with the arsenic BMT inhibitor selenite or after glutathione (GSH) depletion. The expression of oxidative stress-related genes (HO-1, SOD-1, SOD-2, Nrf2 and Keap-1) was also assessed. Exposure to iAs at 24 h (0-20 µM), stimulated ODD only at levels above the LC50 of a 48 h exposure (17 µM). If iAs induced ODD, it also activated oxidative stress-related genes. Selenium did not alter iAs-induced ODD. MMA(3+) at 24 h (0-0.5 µM) caused ODD at levels below the LC50 of a 48 h exposure (1.5 µM), which were greatly increased by GSH depletion but not selenite. MMA(3+) induced ODD at levels not activating oxidant stress response genes. Overall, iAs induced ODD in BMT-deficient cells only at toxic levels. MMA(3+) caused ODD at non-toxic levels, independently of cellular BMT capacity and in a fashion not requiring further BMT.


Subject(s)
Arsenites/toxicity , DNA Damage , Epithelial Cells/drug effects , Organometallic Compounds/toxicity , Oxidative Stress/drug effects , Sodium Compounds/toxicity , Water Pollutants, Chemical/toxicity , Arsenites/metabolism , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Epithelial Cells/metabolism , Gene Expression Profiling , Glutathione/metabolism , Humans , Methylation , Organometallic Compounds/metabolism , Sodium Compounds/metabolism , Transcriptome/drug effects , Transcriptome/genetics , Water Pollutants, Chemical/metabolism
17.
Toxicol Appl Pharmacol ; 264(3): 439-50, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22959928

ABSTRACT

Inorganic arsenic (iAs) is a complete transplacental carcinogen in mice. Previous studies have demonstrated that in utero exposure to iAs promotes cancer in adult mouse offspring, possibly acting through epigenetic mechanisms. Humans and rodents enzymatically convert iAs to its methylated metabolites. This reaction requires S-adenosylmethionine (SAM) as methyl group donor. SAM is also required for DNA methylation. Supplementation with folate, a major dietary source of methyl groups for SAM synthesis, has been shown to modify iAs metabolism and the adverse effects of iAs exposure. However, effects of gestational folate supplementation on iAs metabolism and fetal DNA methylation have never been thoroughly examined. In the present study, pregnant CD1 mice were fed control (i.e. normal folate, or 2.2 mg/kg) or high folate diet (11 mg/kg) from gestational day (GD) 5 to 18 and drank water with 0 or 85 ppm of As (as arsenite) from GD8 to 18. The exposure to iAs significantly decreased body weight of GD18 fetuses and increased both SAM and S-adenosylhomocysteine (SAH) concentrations in fetal livers. High folate intake lowered the burden of total arsenic in maternal livers but did not prevent the effects of iAs exposure on fetal weight or hepatic SAM and SAH concentrations. In fact, combined folate-iAs exposure caused further significant body weight reduction. Notably, iAs exposure alone had little effect on DNA methylation in fetal livers. In contrast, the combined folate-iAs exposure changed the CpG island methylation in 2,931 genes, including genes known to be imprinted. Most of these genes were associated with neurodevelopment, cancer, cell cycle, and signaling networks. The canonical Wnt-signaling pathway, which regulates fetal development, was among the most affected biological pathways. Taken together, our results suggest that a combined in utero exposure to iAs and a high folate intake may adversely influence DNA methylation profiles and weight of fetuses, compromising fetal development and possibly increasing the risk for early-onset of disease in offspring.


Subject(s)
Arsenites/toxicity , Epigenomics , Folic Acid/pharmacology , Gene Expression Regulation, Developmental/drug effects , Sodium Compounds/toxicity , Animals , Arsenites/administration & dosage , Female , Fetal Weight/drug effects , Fetus/drug effects , Folic Acid/administration & dosage , Folic Acid/blood , Liver/drug effects , Liver/embryology , Liver/metabolism , Male , Mice , Pregnancy , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism , Sodium Compounds/administration & dosage
18.
Arch Toxicol ; 86(9): 1475-80, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22466070

ABSTRACT

The toxic effect of cadmium varies with sex in experimental animals. Previous studies have demonstrated that pretreatment of male Fischer 344 (F344) rats with the female sex hormone progesterone markedly enhances the susceptibility to cadmium, suggesting a role for progesterone in the sexual dimorphism of cadmium toxicity. In the present study, we attempted to further elucidate the mechanism for sex differences in cadmium-induced toxicity in F344 rats. A single exposure to cadmium (5.0 mg Cd/kg, sc) was lethal in 10/10 (100 %) female compared with 6/10 (60 %) male rats. Using a lower dose of cadmium (3.0 mg Cd/kg), circulating alanine aminotransferase activity, indicative of hepatotoxicity, was highly elevated in the cadmium treated females but not in males. However, no gender-based differences occurred in the hepatic cadmium accumulation, metallothionein or glutathione levels. When cadmium (5.0 mg Cd/kg) was administered to young rats at 5 weeks of age, the sex-related difference in lethality was minimal. Furthermore, although ovariectomy blocked cadmium-induced lethality, the lethal effects of the metal were restored by pretreatment with progesterone (40 mg/kg, sc, 7 consecutive days) or ß-estradiol (200 µg/kg, sc, 7 consecutive days) to ovariectomized rats. These results provide further evidence that female sex hormones such as progesterone and ß-estradiol are involved in the sexual dimorphism of cadmium toxicity in rats.


Subject(s)
Cadmium Poisoning/physiopathology , Estradiol/pharmacology , Progesterone/pharmacology , Animals , Cadmium/administration & dosage , Cadmium/analysis , Cadmium/pharmacokinetics , Cadmium/toxicity , Cadmium Poisoning/metabolism , Dose-Response Relationship, Drug , Female , Glutathione/metabolism , Hepatic Insufficiency/etiology , Hepatic Insufficiency/prevention & control , Kidney/chemistry , Kidney/drug effects , Kidney/metabolism , Kidney/physiopathology , Liver/chemistry , Liver/drug effects , Liver/metabolism , Liver/physiopathology , Male , Metallothionein/metabolism , Ovariectomy , Rats , Rats, Inbred F344 , Renal Insufficiency/etiology , Renal Insufficiency/prevention & control , Sex Characteristics , Survival Analysis , Tissue Distribution
19.
Arch Toxicol ; 86(6): 975-82, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22398986

ABSTRACT

Developmental exposure to inorganic arsenic is carcinogenic in humans and mice, and adult offspring of mice exposed to inorganic arsenic can develop tumors of the lung, liver, adrenal, uterus, and ovary. It has been suggested that methylarsonous acid (MMA3+), a product of the biological methylation of inorganic arsenic, could be a key carcinogenic species. Thus, pregnant CD1 mice were provided drinking water containing MMA3+ at 0 (control), 12.5, or 25 parts per million (ppm) from gestational days 8 to 18. Tumors were assessed in groups of male or female (initial n = 25) offspring up to 2 years of age. In utero treatment had no effect on survival or body weights. Female offspring exhibited increases in total epithelial uterine tumors (control 0%; 12.5 ppm 26%; 25 ppm 30%), oviduct hyperplasia (control 4%; 12.5 ppm 35%; 25 ppm 43%), adrenal cortical adenoma at 25 ppm (control 0%; 12.5 ppm 9%; 25 ppm 26%), and total epithelial ovarian tumors (control 0%; 12.5 ppm 39%; 25 ppm 26%). Male offspring showed dose-related increases in hepatocellular carcinoma (control 0%; 12.5 ppm 12%; 25 ppm 22%), adrenal adenoma (control 0%; 12.5 ppm 28%; 25 ppm 17%), and lung adenocarcinoma (control 17%; 12.5 ppm 44%). Male offspring had unusual testicular lesions, including two rete testis carcinomas, two adenomas, and three interstitial cell tumors. Overall, maternal consumption of MMA3+ during pregnancy in CD1 mice produced some similar proliferative lesions as gestationally applied inorganic arsenic in the offspring during adulthood.


Subject(s)
Arsenic/toxicity , Arsenicals/adverse effects , Carcinogens/toxicity , Maternal Exposure/adverse effects , Neoplasms/chemically induced , Pregnancy Complications, Neoplastic/etiology , Prenatal Exposure Delayed Effects , Adenocarcinoma/chemically induced , Adenoma/chemically induced , Adrenal Gland Neoplasms/chemically induced , Animals , Arsenic/administration & dosage , Carcinoma, Hepatocellular/chemically induced , Dose-Response Relationship, Drug , Female , Liver Neoplasms/chemically induced , Lung Neoplasms/chemically induced , Male , Mice , Ovarian Neoplasms/chemically induced , Pregnancy , Testicular Neoplasms/chemically induced , Uterine Neoplasms/chemically induced
20.
Toxicol Appl Pharmacol ; 255(3): 242-50, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21820459

ABSTRACT

Inorganic arsenic and UV, both human skin carcinogens, may act together as skin co-carcinogens. We find human skin keratinocytes (HaCaT cells) are malignantly transformed by low-level arsenite (100nM, 30weeks; termed As-TM cells) and with transformation concurrently undergo full adaptation to arsenic toxicity involving reduced apoptosis and oxidative stress response to high arsenite concentrations. Oxidative DNA damage (ODD) is a possible mechanism in arsenic carcinogenesis and a hallmark of UV-induced skin cancer. In the current work, inorganic arsenite exposure (100nM) did not induce ODD during the 30weeks required for malignant transformation. Although acute UV-treatment (UVA, 25J/cm(2)) increased ODD in passage-matched control cells, once transformed by arsenic to As-TM cells, acute UV actually further increased ODD (>50%). Despite enhanced ODD, As-TM cells were resistant to UV-induced apoptosis. The response of apoptotic factors and oxidative stress genes was strongly mitigated in As-TM cells after UV exposure including increased Bcl2/Bax ratio and reduced Caspase-3, Nrf2, and Keap1 expression. Several Nrf2-related genes (HO-1, GCLs, SOD) showed diminished responses in As-TM cells after UV exposure consistent with reduced oxidant stress response. UV-exposed As-TM cells showed increased expression of cyclin D1 (proliferation gene) and decreased p16 (tumor suppressor). UV exposure enhanced the malignant phenotype of As-TM cells. Thus, the co-carcinogenicity between UV and arsenic in skin cancer might involve adaptation to chronic arsenic exposure generally mitigating the oxidative stress response, allowing apoptotic by-pass after UV and enhanced cell survival even in the face of increased UV-induced oxidative stress and increased ODD.


Subject(s)
Arsenites/toxicity , DNA Damage/drug effects , DNA Damage/radiation effects , Keratinocytes/drug effects , Keratinocytes/radiation effects , Oxidants/antagonists & inhibitors , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis/radiation effects , Arsenites/pharmacology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Cell Survival/radiation effects , DNA Damage/physiology , Humans , Keratinocytes/metabolism , Oxidants/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Oxidative Stress/radiation effects , Skin/drug effects , Skin/metabolism , Skin/radiation effects , Ultraviolet Rays/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL