Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Prostate ; 82(13): 1273-1283, 2022 09.
Article in English | MEDLINE | ID: mdl-35747940

ABSTRACT

BACKGROUND: Previously we reported that arsenic and estrogen cause synergistic effects in the neoplastic transformation of human prostate epithelial cells. In addition to receptor-mediated pathways, DNA-reactive estrogen metabolites have also been shown to play a critical role in mutagenicity and carcinogenicity. Both estrogen and arsenic are known prostate carcinogens, however, the effect of coexposure to these two chemicals on genes involved in estrogen metabolism is not known. Therefore, the objective of this study was to evaluate the role of arsenic and estrogen coexposure on the expression of estrogen receptors and estrogen metabolism-associated genes. Earlier, we also reported the synergistic effect of arsenic and estrogen on decreased expression of MBD4 genes that play an important role in DNA repair through its DNA glycosylase activity. To further understand the mechanism, the promoter methylation of this gene was also analyzed. METHODS: Total RNA and protein were isolated from RWPE-1 human prostate epithelial cells that were coexposed to arsenic and estrogen for a chronic duration (6 months). The expression of estrogen receptors, estrogen metabolism associated phase I genes (CYP 1A1, 1A2, 3A4, and 1B1) and phase II gene catechol-O-methyltransferase (COMT), as well as antioxidant MnSOD, were analyzed either at the RNA level by quantitative reverse transcriptase-polymerase chain reaction or at the protein level by western blot. Promoter methylation of MBD4 was analyzed by pyrosequencing. RESULTS: Expression of MnSOD and phase I genes that convert E2 into genotoxic metabolites 2-OH-E2 and 4-OH-E2 were significantly increased, whereas the expression of phase II gene COMT that detoxifies estrogen metabolites was significantly decreased in arsenic and estrogen coexposed cells. MBD4 promoter was hypermethylated in arsenic and estrogen coexposed cells. Coexposure to arsenic and estrogen has synergistic effects on the expression of these genes as well as in MBD4 promoter hypermethylation. CONCLUSIONS: These novel findings suggest that coexposure to arsenic and estrogen acts synergistically in the activation of not only the estrogen receptors but also the genes associated with genotoxic estrogen metabolism and epigenetic inactivation of DNA glycosylase MBD4. Together, these genetic and epigenetic aberrations provide the molecular basis for the potentiation of carcinogenicity of arsenic and estrogen coexposure in prostate epithelial cells.


Subject(s)
Arsenic , DNA Damage , DNA Glycosylases , Estrogens , Prostate , Arsenic/metabolism , Arsenic/toxicity , Catechol O-Methyltransferase/genetics , Catechol O-Methyltransferase/metabolism , Catechol O-Methyltransferase/pharmacology , DNA Damage/drug effects , DNA Damage/genetics , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA Glycosylases/pharmacology , DNA Methylation , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/pharmacology , Environmental Exposure , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Estrogens/adverse effects , Estrogens/pharmacology , Humans , Male , Metabolic Networks and Pathways , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , RNA , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism
2.
Breast Cancer Res Treat ; 149(3): 655-68, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25663548

ABSTRACT

Epigenetic therapy by DNA demethylating agent 5-aza-2'-deoxycytidine (5-aza 2'dC) is clinically effective in acute myeloid leukemia; however, it has shown limited results in treatment of breast cancer and has significant toxicity to normal cells. Green tea polyphenol (-)-epigallocatechin-3-gallate (EGCG) has anti-cancer and DNA demethylating properties with no significant toxicity toward normal cells. Therefore, the objective of this study was to evaluate the therapeutic efficacy of a combination of non-toxic, low dose of 5-aza 2' dC with EGCG, on growth inhibition of breast cancer cells. Human breast cancer cell lines (MCF-7, MDA-MB 231) and non-tumorigenic MCF-10A breast epithelial cells were treated with either 5-aza 2' dC, EGCG, or their combination for 7 days. Cell growth inhibition was determined by cell count, cell viability, cell cycle, and soft agar assay, whereas genes expression changes were determined by quantitative real-time PCR and/or Western blot analysis. Histone modifications and global DNA methylation changes were determined by Western blot and RAPD-PCR, respectively. The results revealed significantly greater inhibition of growth of breast cancer cells by co-treatment with 5-aza 2' dC and EGCG compared to individual treatments, whereas it has no significant toxicity to MCF-10A cells. This was further confirmed by gene expression analysis. Changes in DNA methylation and histone modifications were also greater in cells with combination treatment. Findings of this study suggest that potentiation of growth inhibition of breast cancer cells by 5-aza 2' dC and EGCG combination treatment, at least in part, is mediated by epigenetic mechanism.


Subject(s)
Breast Neoplasms/drug therapy , Catechin/analogs & derivatives , Cell Proliferation/drug effects , Epigenesis, Genetic/drug effects , Plant Extracts/administration & dosage , Azacitidine/administration & dosage , Azacitidine/analogs & derivatives , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Catechin/administration & dosage , Catechin/chemistry , Cell Survival/drug effects , DNA Methylation/drug effects , Decitabine , Female , Gene Expression Regulation, Neoplastic/drug effects , Histones/genetics , Humans , MCF-7 Cells , Plant Extracts/chemistry , Random Amplified Polymorphic DNA Technique , Tea/chemistry
3.
Ecotoxicology ; 22(10): 1461-6, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24085605

ABSTRACT

The role that endocrine disruption could play in sexual selection remains relatively untested, and although estrogens occur in insects, little information exists about their biological role in insect reproduction. Atrazine is a commonly applied herbicide that mimics estrogen in vertebrates. Tenebrio molitor were raised from egg to adult under a gradation of environmentally relevant atrazine exposures and a non-treated control. Atrazine was delivered in the drinking water ad libitum. Female T. molitor were provided with a choice between unrelated males raised under three levels of atrazine exposures. Female preference for males demonstrated a non-monotonic inverted U-shaped response to atrazine exposure. There was no significant difference between the control and the high exposure to atrazine. Excluding the control, female preference increased as exposure concentration increased. These results have important repercussions for nonlethal effects of endocrine disruption on populations, their capacity to interfere with sexual selection, and the role of estrogen in pheromone communication among insects.


Subject(s)
Atrazine/toxicity , Endocrine Disruptors/toxicity , Environmental Exposure , Herbicides/toxicity , Mating Preference, Animal/drug effects , Tenebrio/drug effects , Water Pollutants, Chemical/toxicity , Animals , Body Weight/drug effects , Female , Fertility/drug effects , Larva/drug effects , Larva/growth & development , Longevity/drug effects , Male , Tenebrio/growth & development
4.
Prostate ; 73(15): 1660-72, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23804311

ABSTRACT

BACKGROUND: Chronic exposure to arsenic and estrogen is associated with risk of prostate cancer, but their mechanism is not fully understood. Additionally, the carcinogenic effects of their co-exposure are not known. Therefore, the objective of this study was to evaluate the effects of chronic exposure to arsenic, estrogen, and their combination, on cell growth and transformation, and identify the mechanism behind these effects. METHODS: RWPE-1 human prostate epithelial cells were chronically exposed to arsenic and estrogen alone and in combination. Cell growth was measured by cell count and cell cycle, whereas cell transformation was evaluated by colony formation assay. Gene expression was measured by quantitative real-time PCR and confirmed at protein level by Western blot analysis. MLH1 promoter methylation was determined by pyrosequencing method. RESULTS: Exposure to arsenic, estrogen, and their combinations increases cell growth and transformation in RWPE-1 cells. Increased expression of Cyclin D1 and Bcl2, whereas decreased expression of mismatch repair genes MSH4, MSH6, and MLH1 was also observed. Hypermethylation of MLH1 promoter further suggested the epigenetic inactivation of MLH1 expression in arsenic and estrogen treated cells. Arsenic and estrogen combination caused greater changes than their individual treatments. CONCLUSIONS: Findings of this study for the first time suggest that arsenic and estrogen exposures cause increased cell growth and survival potentially through epigenetic inactivation of MLH1 resulting in decreased MLH1-mediated apoptotic response, and consequently increased cellular transformation.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Arsenic/administration & dosage , Cell Enlargement/drug effects , Cell Transformation, Neoplastic/drug effects , Epithelial Cells/drug effects , Estrogens/administration & dosage , Nuclear Proteins/genetics , Prostate/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle/drug effects , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression/drug effects , Gene Silencing , Humans , Male , Methylation , MutL Protein Homolog 1 , Nuclear Proteins/metabolism , Prostate/metabolism
5.
PLoS One ; 7(8): e43880, 2012.
Article in English | MEDLINE | ID: mdl-22952798

ABSTRACT

Chronic exposures to arsenic and estrogen are known risk factors for prostate cancer. Though the evidence suggests that exposure to arsenic or estrogens can disrupt normal DNA methylation patterns and histone modifications, the mechanisms by which these chemicals induce epigenetic changes are not fully understood. Moreover, the epigenetic effects of co-exposure to these two chemicals are not known. Therefore, the objective of this study was to evaluate the effects of chronic exposure to arsenic and estrogen, both alone and in combination, on the expression of epigenetic regulatory genes, their consequences on DNA methylation, and histone modifications. Human prostate epithelial cells, RWPE-1, chronically exposed to arsenic and estrogen alone and in combination were used for analysis of epigenetic regulatory genes expression, global DNA methylation changes, and histone modifications at protein level. The result of this study revealed that exposure to arsenic, estrogen, and their combination alters the expression of epigenetic regulatory genes and changes global DNA methylation and histone modification patterns in RWPE-1 cells. These changes were significantly greater in arsenic and estrogen combination treated group than individually treated group. The findings of this study will help explain the epigenetic mechanism of arsenic- and/or estrogen-induced prostate carcinogenesis.


Subject(s)
Arsenic/pharmacology , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Estrogens/pharmacology , Prostate/cytology , Cell Line , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/drug effects , DNA Methylation/genetics , Drug Interactions , Estradiol/pharmacology , Histones/metabolism , Humans , Male , Time Factors
6.
Cancer Lett ; 316(1): 62-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22082530

ABSTRACT

Prolonged exposure to elevated levels of estrogen is a risk factor for breast cancer. Though increased cell growth and loss of DNA repair capacity is one of the proposed mechanisms for estrogen-induced cancers, the mechanism through which estrogen induces cell growth and decreases DNA repair capacity is not clear. DNA hypermethylation is known to inactivate DNA repair genes and apoptotic response in cancer cells. Therefore, the objective of this study was to determine the role of DNA hypermethylation in estrogen-induced cell growth and regulation of DNA repair genes expression in breast cancer cells. To achieve this objective, the estrogen-responsive MCF-7 cells either pretreated with 5-aza-2-deoxycytidine (5-aza-dC) or untreated (as control) were exposed to 17 beta-estradiol (E2), and its effect on cell growth and expression of DNA repair genes were measured. The result revealed that 5-aza-dC abrogates the E2-induced growth in MCF-7 cells. An increased expression of OGG1, MSH4, and MLH1 by 5-aza-dC treatment alone, suggest the DNA hypermethylation as a potential cause for decreased expression of these genes in MCF-7 cells. The decreased expression of ERCC1, XPC, OGG1, and MLH1 by E2 alone and its restoration by co-treatment with 5-aza-dC further suggest that E2 reduces the expression of these DNA repair genes potentially through promoter hypermethylation. Reactivation of mismatch repair (MMR) gene MLH1 and abrogation of E2-induced cell growth by 5-aza-dC treatment suggest that estrogen causes increased growth in breast cancer cells potentially through the inhibition of MMR-mediated apoptotic response. In summary, this study suggests that estrogen increases cell growth and decreases the DNA repair capacity in breast cancer cells, at least in part, through epigenetic mechanism.


Subject(s)
Azacitidine/analogs & derivatives , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , DNA Methylation/drug effects , DNA Repair , Estradiol/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Azacitidine/pharmacology , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Growth Processes/drug effects , Cell Growth Processes/genetics , DNA Mismatch Repair , Decitabine , Drug Interactions , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Promoter Regions, Genetic
7.
Chem Res Toxicol ; 24(3): 340-9, 2011 Mar 21.
Article in English | MEDLINE | ID: mdl-21235219

ABSTRACT

Arsenic is a known carcinogen, and its exposure is associated with cancers in multiple target organs including the prostate. Whether arsenic causes cancer by increased cell proliferation or cell survival is not clear. Additionally, mitochondria have been shown to play important roles in arsenic-induced DNA damage and carcinogenesis. However, the mechanism of mitochondrial involvement in arsenic-induced cancer is not clear. Therefore, the objectives of this study were to investigate the effect of arsenic on cell proliferation/survival and genotoxicity, and to determine the effect of arsenic on the expression of mitochondrial transcription factor A (mtTFA) in human prostate epithelial cells, RWPE-1. Results of this study revealed that chronic exposure to arsenic causes increased cell survival. Arsenic also induced nuclear DNA damage and mutations in mitochondrial DNA. Expressions of DNA repair genes ERCC6, XPC, OGG1, and reactive oxygen species (ROS) scavenger MnSOD was also altered in arsenic-exposed cells. Arsenic concentration-dependent increased expression of mtTFA and its regulator NRF-1 was observed in arsenic-exposed cells, suggesting that arsenic regulates mitochondrial activity through an NRF-1-dependent pathway. In summary, this study suggests that chronic exposure to arsenic causes DNA damage and increased cell survival that may ultimately result in neoplastic transformation of human prostate epithelial cells. Additionally, this study also provides evidence that arsenic controls mitochondrial function by regulating mtTFA expression.


Subject(s)
Arsenic/toxicity , DNA Damage , DNA-Binding Proteins/metabolism , Epithelial Cells/drug effects , Mitochondrial Proteins/metabolism , Prostate/drug effects , Transcription Factors/metabolism , Cell Line , Cell Proliferation , Cell Survival , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Repair , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , Epithelial Cells/metabolism , Humans , Male , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mutation , NF-E2-Related Factor 1/genetics , NF-E2-Related Factor 1/metabolism , Poly-ADP-Ribose Binding Proteins , Prostate/cytology , Reactive Oxygen Species/metabolism , Sequence Analysis, DNA , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...