Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Genes (Basel) ; 15(3)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38540381

ABSTRACT

The use of E-cigarettes, often considered a safer alternative to traditional smoking, has been associated with high rates of cellular toxicity, genetic alterations, and inflammation. Neuroinflammatory impacts of cigarette smoking during pregnancy have been associated with increased risks of adverse childhood health outcomes; however, it is still relatively unknown if the same propensity is conferred on offspring by maternal vaping during gestation. Results from our previous mouse inhalation studies suggest such a connection. In this earlier study, pregnant C57BL/6 mice were exposed daily to inhaled E-cig aerosols (i.e., propylene glycol and vegetable glycerin, [PG/VG]), with or without nicotine (16 mg/mL) by whole-body inhalation throughout gestation (3 h/d; 5 d/week; total ~3-week) and continuing postnatally from post-natal day (PND) 4-21. As neuroinflammation is involved in the dysregulation of glucose homeostasis and weight gain, this study aimed to explore genes associated with these pathways in 1-mo.-old offspring (equivalent in humans to 12-18 years of age). Results in the offspring demonstrated a significant increase in glucose metabolism protein levels in both treatment groups compared to filtered air controls. Gene expression analysis in the hypothalamus of 1 mo. old offspring exposed perinatally to E-cig aerosols, with and without nicotine, revealed significantly increased gene expression changes in multiple genes associated with neuroinflammation. In a second proof-of-principal parallel study employing the same experimental design, we shifted our focus to the hippocampus of the postpartum mothers. We targeted the mRNA levels of several neurotrophic factors (NTFs) indicative of neuroinflammation. While there were suggestive changes in mRNA expression in this study, levels failed to reach statistical significance. These studies highlight the need for ongoing research on E-cig-induced alterations in neuroinflammatory pathways.


Subject(s)
Electronic Nicotine Delivery Systems , Nicotine , Humans , Pregnancy , Female , Animals , Mice , Child , Nicotine/toxicity , Neuroinflammatory Diseases , Mice, Inbred C57BL , Aerosols/adverse effects , RNA, Messenger
6.
Toxicol Sci ; 162(1): 276-286, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29161446

ABSTRACT

Recent epidemiological data indicate that the popularity of electronic cigarettes (e-cigarettes), and consequently nicotine use, is rising in both adolescent and adult populations. As nicotine is a known developmental neurotoxin, these products present a potential threat for those exposed during early life stages. Despite this, few studies have evaluated the toxicity of e-cigarettes on the developing central nervous system. The goal of this study was to assess neurotoxicity resulting from early-life exposure to electronic cigarette aerosols in an in vivo model. Specifically, studies here focused on neuro-parameters related to neuroinflammation and neurotrophins. To accomplish this, pregnant and neonatal C57BL/6 mice were exposed to aerosols produced from classic tobacco flavor e-cigarette cartridges (with [13 mg/ml] and without nicotine) during gestation (∼3 weeks) and lactation (∼3 weeks) via whole-body inhalation. Exposure to e-cigarette aerosols with and without nicotine caused significant reductions in hippocampal gene expression of Ngfr and Bdnf, as well as in serum levels of cytokines IL-1ß, IL-2, and IL-6. Exposure to e-cigarette aerosols without nicotine enhanced expression of Iba-1, a specific marker of microglia, in the cornus ammonis 1 region of the hippocampus. Overall, our novel results indicate that exposure to e-cigarette aerosols, with and without nicotine, poses a considerable risk to the developing central nervous system. Consequently, e-cigarettes should be considered a potential public health threat, especially early in life, requiring further research and policy considerations.


Subject(s)
Aerosols/toxicity , Electronic Nicotine Delivery Systems , Hippocampus/drug effects , Microglia/drug effects , Nerve Growth Factors/genetics , Prenatal Exposure Delayed Effects/chemically induced , Transcriptome/drug effects , Administration, Inhalation , Animals , Animals, Newborn , Cytokines/blood , Female , Gene Expression Profiling , Hippocampus/growth & development , Hippocampus/metabolism , Mice, Inbred C57BL , Microglia/immunology , Microglia/metabolism , Nicotine/toxicity , Pregnancy , Prenatal Exposure Delayed Effects/metabolism
7.
Int J Environ Res Public Health ; 13(4): 417, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27077873

ABSTRACT

Electronic cigarettes (e-cigarettes), battery-powered devices containing nicotine, glycerin, propylene glycol, flavorings, and other substances, are increasing in popularity. They pose a potential threat to the developing brain, as nicotine is a known neurotoxicant. We hypothesized that exposure to e-cigarettes during early life stages induce changes in central nervous system (CNS) transcriptome associated with adverse neurobiological outcomes and long-term disease states. To test the hypothesis, pregnant C57BL/6 mice were exposed daily (via whole body inhalation) throughout gestation (3 h/day; 5 days/week) to aerosols produced from e-cigarettes either with nicotine (13-16 mg/mL) or without nicotine; following birth, pups and dams were exposed together to e-cigarette aerosols throughout lactation beginning at postnatal day (PND) 4-6 and using the same exposure conditions employed during gestational exposure. Following exposure, frontal cortex recovered from ~one-month-old male and female offspring were excised and analyzed for gene expression by RNA Sequencing (RNA-Seq). Comparisons between the treatment groups revealed that e-cigarette constituents other than nicotine might be partly responsible for the observed biological effects. Transcriptome alterations in both offspring sexes and treatment groups were all significantly associated with downstream adverse neurobiological outcomes. Results from this study demonstrate that e-cigarette exposure during early life alters CNS development potentially leading to chronic neuropathology.


Subject(s)
Electronic Nicotine Delivery Systems/adverse effects , Frontal Lobe/drug effects , Prenatal Exposure Delayed Effects/physiopathology , Transcriptome/drug effects , Administration, Inhalation , Animals , Female , Gene Expression Profiling , Glycerol/adverse effects , Male , Mice , Mice, Inbred C57BL , Nicotine/adverse effects , Pregnancy , Propylene Glycol/adverse effects
8.
Environ Mol Mutagen ; 57(2): 137-50, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26581878

ABSTRACT

The mechanisms by which arsenic-induced genomic instability is initiated and maintained are poorly understood. To investigate potential epigenetic mechanisms, in this study we evaluated global DNA methylation levels in V79 cells and human HaCaT keratinocytes at several time points during expanded growth of cell cultures following removal of arsenite exposures. We have found altered genomic methylation patterns that persisted up to 40 cell generations in HaCaT cells after the treatments were withdrawn. Moreover, mRNA expression levels were evaluated by RT-PCR for DNMT1, DNMT3A, DNMT3B, HMLH1, and HMSH2 genes, demonstrating that the down regulation of DNMT3A and DNMT3B genes, but not DNMT1, occurred in an arsenic dose-dependent manner, and persisted for many cell generations following removal of the arsenite, offering a plausible mechanism of persistently genotoxic arsenic action. Analyses of promoter methylation status of the DNA mismatch repair genes HMLH1 and HMSH2 show that HMSH2, but not HMLH1, was epigenetically regulated by promoter hypermethylation changes following arsenic treatment. The results reported here demonstrate that arsenic exposure promptly induces genome-wide global DNA hypomethylation, and some specific gene promoter methylation changes, that persist for many cell generations following withdrawal of arsenite, supporting the hypothesis that the cells undergo epigenetic reprogramming at both the gene and genome level that is durable over many cell generations in the absence of further arsenic treatment. These DNA methylation changes, in concert with other known epigenome alterations, are likely contributing to long-lasting arsenic-induced genomic instability that manifests in several ways, including aberrant chromosomal effects.


Subject(s)
Arsenic/toxicity , DNA Methylation/drug effects , Genomic Instability/drug effects , 5-Methylcytosine/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Arsenites/toxicity , Cells, Cultured , Cricetulus , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Fibroblasts/drug effects , Keratinocytes/drug effects , Long Interspersed Nucleotide Elements/drug effects , MutL Protein Homolog 1 , MutS Homolog 2 Protein/genetics , Nuclear Proteins/genetics , Promoter Regions, Genetic/drug effects , DNA Methyltransferase 3B
9.
Environ Mol Mutagen ; 54(3): 153-7, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23444128

ABSTRACT

In late 2012, the members of the Environmental Mutagen Society voted to change its name to the Environmental Mutagenesis and Genomics Society. Here, we describe the thought process that led to adoption of the new name, which both respects the rich history of a Society founded in 1969 and reflects the many advances in our understanding of the nature and breadth of gene-environment interactions during the intervening 43 years.


Subject(s)
Environmental Pollutants/toxicity , Genomics/history , Mutagenesis , Mutagens/toxicity , Societies, Scientific/history , Genomics/organization & administration , Genomics/trends , History, 20th Century , History, 21st Century , Names , Societies, Scientific/organization & administration , Societies, Scientific/trends , Toxicogenetics/history , Toxicogenetics/organization & administration , Toxicogenetics/trends , United States
10.
Mutat Res ; 752(1): 6-9, 2013.
Article in English | MEDLINE | ID: mdl-22935230

ABSTRACT

Next-generation sequencing technologies can now be used to directly measure heritable de novo DNA sequence mutations in humans. However, these techniques have not been used to examine environmental factors that induce such mutations and their associated diseases. To address this issue, a working group on environmentally induced germline mutation analysis (ENIGMA) met in October 2011 to propose the necessary foundational studies, which include sequencing of parent-offspring trios from highly exposed human populations, and controlled dose-response experiments in animals. These studies will establish background levels of variability in germline mutation rates and identify environmental agents that influence these rates and heritable disease. Guidance for the types of exposures to examine come from rodent studies that have identified agents such as cancer chemotherapeutic drugs, ionizing radiation, cigarette smoke, and air pollution as germ-cell mutagens. Research is urgently needed to establish the health consequences of parental exposures on subsequent generations.


Subject(s)
Gene-Environment Interaction , Genetic Diseases, Inborn/genetics , Genomics , Animals , Environmental Pollutants/toxicity , Germ-Line Mutation , Humans , Radiation Effects , Tobacco Products/adverse effects
11.
Biometals ; 25(5): 927-37, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22692362

ABSTRACT

Arsenite (As) causes transformation of human osteogenic sarcoma cells (HOS) when applied continuously at low doses (0.1-0.5 µM) during 8-weeks of exposure. However, the mechanisms by which As transforms human cells are not known. We investigated whether alterations occurred in gene expression and protein levels of antioxidant defense proteins, such as superoxide dismutase 1 (SOD1) and ferritin. In comparison to control HOS cells, 0.1 µM As induced greater cell proliferation and decreased anti-oxidant defenses. The tumor suppressor protein p53 was also decreased at both mRNA and protein levels. Further, pig3 (p53-induced-gene 3), a homolog of NQO1 (NADPH quinone oxidoreductase 1), was also down-regulated after 8 weeks of As challenge. The treatment of HOS cells with dicumarol, a NQO1 inhibitor, caused a dose-dependent decline in p53 protein levels, proving the effect of an antioxidant enzyme on p53 expression and, potentially, down-stream processes. Caffeic acid phenethyl ester, an antioxidant, prevented the As-induced decreases in SOD1, p53, and ferritin mRNA and protein levels. SOD1, p53 and ferritin levels were inversely related to As-induced cell proliferation. Cumulatively, these results strongly suggest that impairment in antioxidant defenses contributes to As-induced human cell transformation and that the p53 pathway is involved in the process.


Subject(s)
Antioxidants/metabolism , Arsenites/toxicity , Cell Transformation, Neoplastic/drug effects , Caffeic Acids/pharmacology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Dicumarol/pharmacology , Environmental Pollutants/toxicity , Ferritins/genetics , Ferritins/metabolism , Gene Expression/drug effects , Genes, p53/drug effects , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Models, Biological , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Oxidative Stress/drug effects , Phenylethyl Alcohol/analogs & derivatives , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
12.
Metallomics ; 3(11): 1135-41, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21976018

ABSTRACT

Environmental arsenic compounds and their methylated metabolites do not form adducts with DNA, but do cause oxidative DNA damage. Chromosome aberrations are seen at toxic concentrations. Genetic effects that occur at non-toxic concentrations include aneuploidy, comutagenesis (resulting from indirect effects on DNA repair), and delayed mutagenesis (probably secondary to aneuploidy and/or epigenetic effects). Effects of trivalent arsenicals on poly(ADP ribose) polymerase and P53 activation may mediate effects on DNA repair and aneuploidy. A growing literature points to the epigenetic effects of arsenic compounds in cells and in vivo. A review of the current literature on DNA methylation, histone modifications and microRNA effects is presented.


Subject(s)
Arsenicals/pharmacology , Chromosome Aberrations/chemically induced , DNA Damage/drug effects , DNA Repair/drug effects , DNA/drug effects , Environmental Pollutants/pharmacology , Epigenomics , Arsenicals/chemistry , Arsenicals/metabolism , DNA/metabolism , DNA Methylation/drug effects , Dose-Response Relationship, Drug , Histones/metabolism , Humans , Mutagenesis , Poly(ADP-ribose) Polymerases/metabolism
13.
Cancer Lett ; 308(1): 43-53, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21570765

ABSTRACT

Breast cancer (BC) patients use alternative and natural remedies more than patients with other malignancies. Specifically, 63-83% use at least one type of alternative medicine and 25-63% use herbals and vitamins. Propolis is a naturopathic honeybee product, and CAPE (caffeic acid phenethyl ester), is a major medicinal component of propolis. CAPE, in a concentration dependent fashion, inhibits MCF-7 (hormone receptor positive, HR+) and MDA-231 (a model of triple negative BC (TNBC) tumor growth, both in vitro and in vivo without much effect on normal mammary cells and strongly influences gene and protein expression. It induces cell cycle arrest, apoptosis and reduces expression of growth and transcription factors, including NF-κB. Notably, CAPE down-regulates mdr-1 gene, considered responsible for the resistance of cancer cells to chemotherapeutic agents. Further, CAPE dose-dependently suppresses VEGF formation by MDA-231 cells and formation of capillary-like tubes by endothelial cells, implicating inhibitory effects on angiogenesis. In conclusion, our results strongly suggest that CAPE inhibits MDA-231 and MCF-7 human breast cancer growth via its apoptotic effects, and modulation of NF-κB, the cell cycle, and angiogenesis.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Caffeic Acids/pharmacology , NF-kappa B/antagonists & inhibitors , Phenylethyl Alcohol/analogs & derivatives , Propolis/chemistry , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Bees , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Caffeic Acids/chemistry , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Female , Humans , Mice , Mice, SCID , NF-kappa B/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Phenylethyl Alcohol/chemistry , Phenylethyl Alcohol/pharmacology , Xenograft Model Antitumor Assays
14.
Cancer Epidemiol ; 34(6): 717-23, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20627767

ABSTRACT

BACKGROUND: Promoter methylation of tumor suppressor genes is a frequent and early event in breast carcinogenesis. Paired tumor tissue and serum samples from women with breast cancer show that promoter methylation is detectable in both sample types, with good concordance. This suggests the potential for these serum markers to be used for breast cancer detection. METHODS: The current study was a case-control study nested within the prospective New York University Women's Health Study cohort aimed to assess the ability of promoter methylation in serum to detect pre-clinical disease. Cases were women with blood samples collected within the 6 months preceding breast cancer diagnosis (n=50). Each case was matched to 2 healthy cancer-free controls and 1 cancer-free control with a history of benign breast disease (BBD). RESULTS: Promoter methylation analysis of four cancer-related genes: -RASSF1A, GSTP1, APC and RARß2, - was conducted using quantitative methylation-specific PCR. Results showed that the frequency of methylation was lower than expected among cases and higher than expected among controls. Methylation was detected in the promoter region of: RASSF1A in 22.0%, 22.9% and 17.2% of cases, BBD controls and healthy controls respectively; GSTP1 in 4%, 10.4% and 7.1% respectively; APC in 2.0%, 4.4% and 4.2% respectively and RARß2 in 6.7%, 2.3% and 1.1% respectively. CONCLUSION: Methylation status of the four genes included in this study was unable to distinguish between cases and either control group. This study highlights some methodological issues to be addressed in planning prospective studies to evaluate methylation markers as diagnostic biomarkers.


Subject(s)
Biomarkers, Tumor/blood , Breast Neoplasms/genetics , DNA Methylation , Adult , Aged , Breast Neoplasms/blood , Breast Neoplasms/diagnosis , Case-Control Studies , Cohort Studies , Early Detection of Cancer/methods , Female , Follow-Up Studies , Humans , Middle Aged , Polymerase Chain Reaction , Promoter Regions, Genetic , Prospective Studies
15.
BMC Genomics ; 10: 524, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19917080

ABSTRACT

BACKGROUND: The understanding of the biological function, regulation, and cellular interactions of the yeast genome and proteome, along with the high conservation in gene function found between yeast genes and their human homologues, has allowed for Saccharomyces cerevisiae to be used as a model organism to deduce biological processes in human cells. Here, we have completed a systematic screen of the entire set of 4,733 haploid S. cerevisiae gene deletion strains (the entire set of nonessential genes for this organism) to identify gene products that modulate cellular toxicity to nickel sulfate (NiSO(4)). RESULTS: We have identified 149 genes whose gene deletion causes sensitivity to NiSO(4) and 119 genes whose gene deletion confers resistance. Pathways analysis with proteins whose absence renders cells sensitive and resistant to nickel identified a wide range of cellular processes engaged in the toxicity of S. cerevisiae to NiSO(4). Functional categories overrepresented with proteins whose absence renders cells sensitive to NiSO(4) include homeostasis of protons, cation transport, transport ATPases, endocytosis, siderophore-iron transport, homeostasis of metal ions, and the diphthamide biosynthesis pathway. Functional categories overrepresented with proteins whose absence renders cells resistant to nickel include functioning and transport of the vacuole and lysosome, protein targeting, sorting, and translocation, intra-Golgi transport, regulation of C-compound and carbohydrate metabolism, transcriptional repression, and chromosome segregation/division. Interactome analysis mapped seven nickel toxicity modulating and ten nickel-resistance networks. Additionally, we studied the degree of sensitivity or resistance of the 111 nickel-sensitive and 72 -resistant strains whose gene deletion product has a similar protein in human cells. CONCLUSION: We have undertaken a whole genome approach in order to further understand the mechanism(s) regulating the cell's toxicity to nickel compounds. We have used computational methods to integrate the data and generate global models of the yeast's cellular response to NiSO(4). The results of our study shed light on molecular pathways associated with the cellular response of eukaryotic cells to nickel compounds and provide potential implications for further understanding the toxic effects of nickel compounds to human cells.


Subject(s)
Genome, Fungal/genetics , Nickel/toxicity , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Sequence Deletion/genetics , Signal Transduction/drug effects , Adenosine Triphosphatases/metabolism , Amino Acids/metabolism , Carbohydrate Metabolism/drug effects , Chromosome Segregation/drug effects , Endocytosis/drug effects , Fungal Proteins/genetics , Fungal Proteins/metabolism , Genomics , Homeostasis/drug effects , Humans , Iron/metabolism , Models, Biological , Phenotype , Protein Transport/drug effects , Protons , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Siderophores/metabolism , Transcription, Genetic/drug effects
16.
Genomics ; 94(5): 294-307, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19631266

ABSTRACT

We have used Saccharomyces cerevisiae to identify toxicologically important proteins and pathways involved in arsenic-induced toxicity and carcinogenicity in humans. We performed a systemic screen of the complete set of 4733 haploid S. cerevisiae single-gene-deletion mutants to identify those that have decreased or increased growth, relative to wild type, after exposure to sodium arsenite (NaAsO(2)). IC(50) values for all mutants were determined to further validate our results. Ultimately we identified 248 mutants sensitive to arsenite and 5 mutants resistant to arsenite exposure. We analyzed the proteins corresponding to arsenite-sensitive mutants and determined that they belonged to functional categories that include protein binding, phosphate metabolism, vacuolar/lysosomal transport, protein targeting, sorting, and translocation, cell growth/morphogenesis, cell polarity and filament formation. Furthermore, these data were mapped onto a protein interactome to identify arsenite-toxicity-modulating networks. These networks are associated with the cytoskeleton, ubiquitination, histone acetylation and the MAPK signaling pathway. Our studies have potential implications for understanding toxicity and carcinogenesis in arsenic-induced human conditions, such as cancer and aging.


Subject(s)
Arsenites/toxicity , Gene Expression Regulation, Fungal/drug effects , Gene Regulatory Networks/drug effects , Genome, Fungal , Saccharomyces cerevisiae Proteins/drug effects , Saccharomyces cerevisiae/drug effects , Sodium Compounds/toxicity , Arsenites/pharmacology , Drug Resistance, Fungal , Gene Deletion , Humans , Inhibitory Concentration 50 , Microbial Sensitivity Tests/methods , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sodium Compounds/pharmacology
17.
Environ Mol Mutagen ; 49(1): 36-45, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18181168

ABSTRACT

Dietary agents with chemopreventive potential, including soy-derived genistein and tomato-derived lycopene, have been shown to alter gene expression in ways that can either promote or potentially inhibit the carcinogenic processes. To begin to explore the mechanisms by which these agents may be acting we have examined the DNA methylation modulating capacity of genistein or lycopene for several genes relevant to breast cancer in the breast cancer cell lines MCF-7 and MDA-MB-468, as well as in immortalized but noncancer fibrocystic MCF10A breast cells. We find using methylation specific PCR (MSP) that a low, nontoxic concentration of genistein (3.125 microM, resupplemented every 48 hr for 1 week) or a single dose of lycopene (2 microM) partially demethylates the promoter of the GSTP1 tumor suppressor gene in MDA-MB-468 cells. RT-PCR studies confirm a lack of GSTP1 expression in untreated MDA-MB-468, with restoration of GSTP1 expression after genistein or lycopene treatment. The RARbeta2 gene however, was not demethylated by genistein or lycopene in either of these breast cancer cell lines. But, lycopene (2 microM, once per week for 2 weeks) did induce demethylation of RARbeta2 and the HIN-1 genes in the noncancer MCF10A fibrocystic breast cells. These data show for the first time that the tomato carotenoid lycopene has direct DNA demethylating activity. In summary, both genistein and lycopene, at very low, dietarily relevant concentrations can potentially mitigate tumorigenic processes via promoter methylation modulation of gene expression.


Subject(s)
Breast Neoplasms/genetics , Carotenoids/pharmacology , DNA Methylation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genistein/pharmacology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation , Dose-Response Relationship, Drug , Female , Glutathione S-Transferase pi/drug effects , Glutathione S-Transferase pi/genetics , Humans , Lycopene , Reverse Transcriptase Polymerase Chain Reaction
18.
Toxicol Appl Pharmacol ; 224(1): 1-11, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17688899

ABSTRACT

The potential health benefits of soy-derived phytoestrogens include their reported utility as anticarcinogens, cardioprotectants and as hormone replacement alternatives in menopause. Although there is increasing popularity of dietary phytoestrogen supplementation and of vegetarian and vegan diets among adolescents and adults, concerns about potential detrimental or other genotoxic effects persist. While a variety of genotoxic effects of phytoestrogens have been reported in vitro, the concentrations at which such effects occurred were often much higher than the physiologically relevant doses achievable by dietary or pharmacologic intake of soy foods or supplements. This review focuses on in vitro studies of the most abundant soy phytoestrogen, genistein, critically examining dose as a crucial determinant of cellular effects. In consideration of levels of dietary genistein uptake and bioavailability we have defined in vitro concentrations of genistein >5 microM as non-physiological, and thus "high" doses, in contrast to much of the previous literature. In doing so, many of the often-cited genotoxic effects of genistein, including apoptosis, cell growth inhibition, topoisomerase inhibition and others become less obvious. Recent cellular, epigenetic and microarray studies are beginning to decipher genistein effects that occur at dietarily relevant low concentrations. In toxicology, the well accepted principle of "the dose defines the poison" applies to many toxicants and can be invoked, as herein, to distinguish genotoxic versus potentially beneficial in vitro effects of natural dietary products such as genistein.


Subject(s)
Genistein/toxicity , Mutagens , Animals , Anticarcinogenic Agents , Antimutagenic Agents/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Survival/drug effects , DNA Damage , Diet , Gene Expression/drug effects , Genistein/metabolism , Genistein/pharmacology , Humans , Phytoestrogens/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Glycine max
19.
Toxicol Appl Pharmacol ; 222(3): 289-97, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17316729

ABSTRACT

Arsenic in drinking water, a mixture of arsenite and arsenate, is associated with increased skin and other cancers in Asia and Latin America, but not the United States. Arsenite alone in drinking water does not cause skin cancers in experimental animals; therefore, it is not a complete carcinogen in skin. We recently showed that low concentrations of arsenite enhanced the tumorigenicity of solar UV irradiation in hairless mice, suggesting arsenic cocarcinogenesis with sunlight in skin cancer and perhaps with different carcinogenic partners for lung and bladder tumors. Cocarcinogenic mechanisms could include blocking DNA repair, stimulating angiogenesis, altering DNA methylation patterns, dysregulating cell cycle control, induction of aneuploidy and blocking apoptosis. Arsenicals are documented clastogens but not strong mutagens, with weak mutagenic activity reported at highly toxic concentrations of inorganic arsenic. Previously, we showed that arsenite, but not monomethylarsonous acid (MMA[III]), induced delayed mutagenesis in HOS cells. Here, we report new data on the mutagenicity of the trivalent methylated arsenic metabolites MMA(III) and dimethylarsinous acid [DMA(III)] at the gpt locus in Chinese hamster G12 cells. Both methylated arsenicals seemed mutagenic with apparent sublinear dose responses. However, significant mutagenesis occurred only at highly toxic concentrations of MMA(III). Most mutants induced by MMA(III) and DMA(III) exhibited transgene deletions. Some non-deletion mutants exhibited altered DNA methylation. A critical discussion of cell survival leads us to conclude that clastogenesis occurs primarily at highly cytotoxic arsenic concentrations, casting further doubt as to whether a genotoxic mode of action (MOA) for arsenicals is supportable.


Subject(s)
Cacodylic Acid/analogs & derivatives , Carcinogens , Mutagens , Neoplasms/chemically induced , Neoplasms/genetics , Organometallic Compounds/toxicity , Animals , CHO Cells , Cacodylic Acid/toxicity , Cell Line, Tumor , Cell Survival , Clone Cells , Cricetinae , Cricetulus , Humans , Mice , Mice, Hairless , Reverse Transcriptase Polymerase Chain Reaction
20.
Mutat Res ; 616(1-2): 60-9, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17178418

ABSTRACT

Vanillin (VAN) and cinnamaldehyde (CIN) are dietary flavorings that exhibit antimutagenic activity against mutagen-induced and spontaneous mutations in bacteria. Although these compounds were antimutagenic against chromosomal mutations in mammalian cells, they have not been studied for antimutagenesis against spontaneous gene mutations in mammalian cells. Thus, we initiated studies with VAN and CIN in human mismatch repair-deficient (hMLH1(-)) HCT116 colon cancer cells, which exhibit high spontaneous mutation rates (mutations/cell/generation) at the HPRT locus, permitting analysis of antimutagenic effects of agents against spontaneous mutation. Long-term (1-3 weeks) treatment of HCT116 cells with VAN at minimally toxic concentrations (0.5-2.5mM) reduced the spontaneous HPRT mutant fraction (MF, mutants/10(6) survivors) in a concentration-related manner by 19-73%. A similar treatment with CIN at 2.5-7.5microM yielded a 13-56% reduction of the spontaneous MF. Short-term (4-h) treatments also reduced the spontaneous MF by 64% (VAN) and 31% (CIN). To investigate the mechanisms of antimutagenesis, we evaluated the ability of VAN and CIN to induce DNA damage (comet assay) and to alter global gene expression (Affymetrix GeneChip) after 4-h treatments. Both VAN and CIN induced DNA damage in both mismatch repair-proficient (HCT116+chr3) and deficient (HCT116) cells at concentrations that were antimutagenic in HCT116 cells. There were 64 genes whose expression was changed similarly by both VAN and CIN; these included genes related to DNA damage, stress responses, oxidative damage, apoptosis, and cell growth. RT-PCR results paralleled the Affymetrix results for four selected genes (HMOX1, DDIT4, GCLM, and CLK4). Our results show for the first time that VAN and CIN are antimutagenic against spontaneous mutations in mammalian (human) cells. These and other data lead us to propose that VAN and CIN may induce DNA damage that elicits recombinational DNA repair, which reduces spontaneous mutations.


Subject(s)
Acrolein/analogs & derivatives , Antimutagenic Agents/pharmacology , Benzaldehydes/pharmacology , DNA Damage , DNA Repair , Gene Expression Regulation , Acrolein/pharmacology , Adaptor Proteins, Signal Transducing , Carrier Proteins/genetics , Cell Survival , Comet Assay , Dose-Response Relationship, Drug , Flavoring Agents/pharmacology , HCT116 Cells , Humans , MutL Protein Homolog 1 , Mutation , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...