Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(21)2023 Oct 25.
Article in English | MEDLINE | ID: mdl-37958568

ABSTRACT

Copper and zinc organometallics have multiple applications and many are considered "data-poor" because the available toxicological information is insufficient for comprehensive health risk assessments. To gain insight into the chemical prioritization and potential structure activity relationship, the current work compares the in vitro toxicity of nine "data-poor" chemicals to five structurally related chemicals and to positive DNA damage inducers (4-nitroquinoline-oxide, aflatoxin-B1). The HC-04 non-cancer human liver cell line was used to investigate the concentration-response effects (24 h and 72 h exposure) on cell proliferation, DNA damage (γH2AX and DNA unwinding assays), and epigenetic effects (global genome changes in DNA methylation and histone modifications using flow cytometry). The 24 h exposure screening data (DNA abundance and damage) suggest a toxicity hierarchy, starting with copper dimethyldithiocarbamate (CDMDC, CAS#137-29-1) > zinc diethyldithiocarbamate (ZDEDC, CAS#14324-55-1) > benzenediazonium, 4-chloro-2-nitro-, and tetrachlorozincate(2-) (2:1) (BDCN4CZ, CAS#14263-89-9); the other chemicals were less toxic and had alternate ranking positions depending on assays. The potency of CDMDC for inducing DNA damage was close to that of the human hepatocarcinogen aflatoxin-B1. Further investigation using sodium-DMDC (SDMDC, CAS#128-04-1), CDMDC and copper demonstrated the role of the interactions between copper and the DMDC organic moiety in generating a high level of CDMDC toxicity. In contrast, additive interactions were not observed with respect to the DNA methylation flow cytometry data in 72 h exposure experiments. They revealed chemical-specific effects, with hypo and hypermethylation induced by copper chloride (CuCl2, CAS#10125-13-0) and zinc-DMDC (ZDMDC, CAS#137-30-4), respectively, but did not show any significant effect of CDMDC or SDMDC. Histone-3 hypoacetylation was a sensitive flow cytometry marker of 24 h exposure to CDMDC. This study can provide insights regarding the prioritization of chemicals for future study, with the aim being to mitigate chemical hazards.


Subject(s)
Copper , Zinc , Humans , Copper/metabolism , Zinc/metabolism , Liver/metabolism , DNA Damage , Cell Line , Aflatoxin B1/toxicity , Epigenesis, Genetic , DNA/metabolism
2.
Data Brief ; 48: 109097, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37077652

ABSTRACT

The Syrian hamster (SH) is an animal model used in virology, toxicology, and carcinogenesis, where a better understanding of epigenetic mechanisms is required. Finding genetic loci regulated by DNA methylation may assist in the development of DNA methylation-based in vitro assays for the identification of carcinogens. This dataset informs on the regulation of gene expression by DNA methylation. Primary cultures of SH male fetal cells (sex determined by differences in kdm5 loci on the X and Y chromosome) were exposed for 7 days to the carcinogen benzo[a]pyrene (20 µM) from which a morphologically transformed colony was collected and reseeded. The colony bypassed senescence and sustained growth. After 210 days of culture, the cells were collected and divided in 16 aliquots to create 4 experimental groups to test the effects of the DNA methylation inhibitor 5-aza-2'-deoxycytidine (5adC). The experiment was initiated 24 h after cell seeding in 10 cm plates. The groups are naïve cells (N), cells exposed for 48 h to either 0.05% DMSO as vehicle (V), or to 5adC at 1 µM and 5 µM. DNA and RNA libraries were sequenced on an Illumina NextSeq 500. Gene expression was analysed by RNAseq and differentially methylated DNA regions (DMRs: clusters of 200 base pairs (bp), read depth >20, q< 0.05, methylation difference >|25%|) were identified by reduce representation bisulfite sequencing (RRBS). Global genome DNA methylation was similar between the N (mean±SD, 47.3%±0.02) and V groups (47.3%±0.01). Although 5adC reduced methylation, the reduction was larger in the 1 µM (39.2%±0.002) than in the 5 µM group (44.3%±0.01). 5adC induced a total of 612 and 190 DMRs by 1 µM and 5 µM, among which 79 and 23 were in the promoter regions (±3,000 bp from the transcription start site), respectively. 5adC induced a total of 1,170 and 1,797 differentially expressed genes (DEGs) by 1 µM and 5 µM, respectively. The 5 µM treatment induced statistically significant toxicity (% cell viability: group N 97%±8, V 98.8%±1.3, 1 µM 97.3%±0.5, 5 µM 93.8%±1.5), which perhaps reduced cell division and daughter cell numbers with inherited changes in methylation, but increased number of DEGs due to both toxicity and methylation changes. As usually observed in the literature, a small portion of DEGs (4% and 4% at 1 µM and 5 µM, respectively) are associated with DMRs in their promoters. These promoter DMRs by themselves are sufficient among other epigenetic marks to induce DEGs. The dataset provides the genomic coordinates of the DMRs and an opportunity to further examine their roles in distal putative promoters or enhancers (yet to be described in the SH) in contributing to gene expression changes, senescence bypass and sustained proliferation as essential carcinogenic events (see companion paper [1]). Finally, this experiment confirms the possibility in future experiments to use 5adC as a positive control for effects on DNA methylation in cells derived from SH.

3.
Toxicology ; 487: 153451, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36754249

ABSTRACT

Current chemical testing strategies are limited in their ability to detect non-genotoxic carcinogens (NGTxC). Epigenetic anomalies develop during carcinogenesis regardless of whether the molecular initiating event is associated with genotoxic (GTxC) or NGTxC events; therefore, epigenetic markers may be harnessed to develop new approach methodologies that improve the detection of both types of carcinogens. This study used Syrian hamster fetal cells to establish the chronology of carcinogen-induced DNA methylation changes from primary cells until senescence-bypass as an essential carcinogenic step. Cells exposed to solvent control for 7 days were compared to naïve primary cultures, to cells exposed for 7 days to benzo[a]pyrene, and to cells at the subsequent transformation stages: normal colonies, morphologically transformed colonies, senescence, senescence-bypass, and sustained proliferation in vitro. DNA methylation changes identified by reduced representation bisulphite sequencing were minimal at day-7. Profound DNA methylation changes arose during cellular senescence and some of these early differentially methylated regions (DMRs) were preserved through the final sustained proliferation stage. A set of these DMRs (e.g., Pou4f1, Aifm3, B3galnt2, Bhlhe22, Gja8, Klf17, and L1l) were validated by pyrosequencing and their reproducibility was confirmed across multiple clones obtained from a different laboratory. These DNA methylation changes could serve as biomarkers to enhance objectivity and mechanistic understanding of cell transformation and could be used to predict senescence-bypass and chemical carcinogenicity.


Subject(s)
Benzo(a)pyrene , DNA Methylation , Cricetinae , Animals , Mesocricetus , Benzo(a)pyrene/toxicity , Reproducibility of Results , Carcinogens/toxicity , Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics
4.
Int J Mol Sci ; 22(20)2021 Oct 11.
Article in English | MEDLINE | ID: mdl-34681626

ABSTRACT

Epigenetics involves a series of mechanisms that entail histone and DNA covalent modifications and non-coding RNAs, and that collectively contribute to programing cell functions and differentiation. Epigenetic anomalies and DNA mutations are co-drivers of cellular dysfunctions, including carcinogenesis. Alterations of the epigenetic system occur in cancers whether the initial carcinogenic events are from genotoxic (GTxC) or non-genotoxic (NGTxC) carcinogens. NGTxC are not inherently DNA reactive, they do not have a unifying mode of action and as yet there are no regulatory test guidelines addressing mechanisms of NGTxC. To fil this gap, the Test Guideline Programme of the Organisation for Economic Cooperation and Development is developing a framework for an integrated approach for the testing and assessment (IATA) of NGTxC and is considering assays that address key events of cancer hallmarks. Here, with the intent of better understanding the applicability of epigenetic assays in chemical carcinogenicity assessment, we focus on DNA methylation and histone modifications and review: (1) epigenetic mechanisms contributing to carcinogenesis, (2) epigenetic mechanisms altered following exposure to arsenic, nickel, or phenobarbital in order to identify common carcinogen-specific mechanisms, (3) characteristics of a series of epigenetic assay types, and (4) epigenetic assay validation needs in the context of chemical hazard assessment. As a key component of numerous NGTxC mechanisms of action, epigenetic assays included in IATA assay combinations can contribute to improved chemical carcinogen identification for the better protection of public health.


Subject(s)
DNA Methylation , Epigenomics , Histones/metabolism , Animals , Arsenicals/pharmacology , DNA Methylation/drug effects , Hazardous Substances/toxicity , Humans , Methyltransferases/metabolism , MicroRNAs/metabolism , Oxidative Stress/drug effects
5.
Arch Toxicol ; 94(8): 2899-2923, 2020 08.
Article in English | MEDLINE | ID: mdl-32594184

ABSTRACT

While regulatory requirements for carcinogenicity testing of chemicals vary according to product sector and regulatory jurisdiction, the standard approach starts with a battery of genotoxicity tests (which include mutagenicity assays). If any of the in vivo genotoxicity tests are positive, a lifetime rodent cancer bioassay may be requested, but under most chemical regulations (except plant protection, biocides, pharmaceuticals), this is rare. The decision to conduct further testing based on genotoxicity test outcomes creates a regulatory gap for the identification of non-genotoxic carcinogens (NGTxC). With the objective of addressing this gap, in 2016, the Organization of Economic Cooperation and Development (OECD) established an expert group to develop an integrated approach to the testing and assessment (IATA) of NGTxC. Through that work, a definition of NGTxC in a regulatory context was agreed. Using the adverse outcome pathway (AOP) concept, various cancer models were developed, and overarching mechanisms and modes of action were identified. After further refining and structuring with respect to the common hallmarks of cancer and knowing that NGTxC act through a large variety of specific mechanisms, with cell proliferation commonly being a unifying element, it became evident that a panel of tests covering multiple biological traits will be needed to populate the IATA. Consequently, in addition to literature and database investigation, the OECD opened a call for relevant assays in 2018 to receive suggestions. Here, we report on the definition of NGTxC, on the development of the overarching NGTxC IATA, and on the development of ranking parameters to evaluate the assays. Ultimately the intent is to select the best scoring assays for integration in an NGTxC IATA to better identify carcinogens and reduce public health hazards.


Subject(s)
Carcinogenicity Tests/standards , Carcinogens/toxicity , Animals , Consensus , Humans , Reproducibility of Results , Risk Assessment
6.
Int J Mol Sci ; 18(6)2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28587163

ABSTRACT

An emerging vision for toxicity testing in the 21st century foresees in vitro assays assuming the leading role in testing for chemical hazards, including testing for carcinogenicity. Toxicity will be determined by monitoring key steps in functionally validated molecular pathways, using tests designed to reveal chemically-induced perturbations that lead to adverse phenotypic endpoints in cultured human cells. Risk assessments would subsequently be derived from the causal in vitro endpoints and concentration vs. effect data extrapolated to human in vivo concentrations. Much direct experimental evidence now shows that disruption of epigenetic processes by chemicals is a carcinogenic mode of action that leads to altered gene functions playing causal roles in cancer initiation and progression. In assessing chemical safety, it would therefore be advantageous to consider an emerging class of carcinogens, the epigenotoxicants, with the ability to change chromatin and/or DNA marks by direct or indirect effects on the activities of enzymes (writers, erasers/editors, remodelers and readers) that convey the epigenetic information. Evidence is reviewed supporting a strategy for in vitro hazard identification of carcinogens that induce toxicity through disturbance of functional epigenetic pathways in human somatic cells, leading to inactivated tumour suppressor genes and carcinogenesis. In the context of human cell transformation models, these in vitro pathway measurements ensure high biological relevance to the apical endpoint of cancer. Four causal mechanisms participating in pathways to persistent epigenetic gene silencing were considered: covalent histone modification, nucleosome remodeling, non-coding RNA interaction and DNA methylation. Within these four interacting mechanisms, 25 epigenetic toxicity pathway components (SET1, MLL1, KDM5, G9A, SUV39H1, SETDB1, EZH2, JMJD3, CBX7, CBX8, BMI, SUZ12, HP1, MPP8, DNMT1, DNMT3A, DNMT3B, TET1, MeCP2, SETDB2, BAZ2A, UHRF1, CTCF, HOTAIR and ANRIL) were found to have experimental evidence showing that functional perturbations played "driver" roles in human cellular transformation. Measurement of epigenotoxicants presents challenges for short-term carcinogenicity testing, especially in the high-throughput modes emphasized in the Tox21 chemicals testing approach. There is need to develop and validate in vitro tests to detect both, locus-specific, and genome-wide, epigenetic alterations with causal links to oncogenic cellular phenotypes. Some recent examples of cell-based high throughput chemical screening assays are presented that have been applied or have shown potential for application to epigenetic endpoints.


Subject(s)
Carcinogenicity Tests , Epigenesis, Genetic , Epigenomics , Adenosine Triphosphate/metabolism , Animals , CCCTC-Binding Factor/metabolism , Carcinogens/pharmacology , Carcinogens/toxicity , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA Methylation , Epigenomics/methods , Gene Expression Regulation/drug effects , Gene Silencing , Histones/metabolism , Humans , Models, Biological , Nucleosomes/genetics , Nucleosomes/metabolism , Protein Binding , Protein Processing, Post-Translational , RNA, Untranslated/genetics , Risk Assessment , Signal Transduction/drug effects
7.
Biotechniques ; 62(4): 157-164, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28403806

ABSTRACT

Here, we present a DNA restriction enzyme-based, fluorescent cytosine extension assay (CEA) to improve normalization and technical variation among sample-to-sample measurements. The assay includes end-labeling of parallel methylation-sensitive and methylation-insensitive DNA restriction enzyme digests along with co-purification and subsequent co-measurement of incorporated fluorescence. This non-radioactive, two-color fluorescent CEA (TCF-CEA) was shown to be a relatively rapid and accurate, with 3-fold greater precision than the one-color CEA. In addition, TCF-CEA provided an index of global DNA methylation that was sensitive to differences >5%. TCF-CEA results were highly correlated with LUminometric Methylation Assay (LUMA) results using human liver cell lines (HepG2, HepaRG, HC-04) as well as a human liver primary cell culture. Hypomethylation was observed in cells treated with the de-methylating agent 5-aza-2'-deoxycytidine. These results demonstrate that TCF-CEA provides a simple method for measuring relative degrees of global DNA methylation that could potentially be scaled up to higher-throughput formats.


Subject(s)
Biological Assay/methods , Cytosine , DNA Methylation/genetics , Azacitidine/analogs & derivatives , Azacitidine/chemistry , DNA Restriction Enzymes/genetics , Decitabine , Fluorescence , Hep G2 Cells , Humans , Liver/cytology , Liver/metabolism , Primary Cell Culture
8.
J Toxicol Environ Health A ; 80(1): 32-52, 2017.
Article in English | MEDLINE | ID: mdl-27905861

ABSTRACT

Hypomethylation of DNA repeats has been linked to diseases and cancer predisposition. Human studies suggest that higher blood concentrations of environmental contaminants (EC) correlate with levels of hypomethylation of DNA repeats in blood. The objective of this study was to examine the effect of in utero and/or lactational exposure to EC on the methylation of DNA repeats (LINE-1 and identifier element) in Sprague-Dawley rat pups at birth, at postnatal day (PND) 21, and in adulthood (PND78-86). From gestation day 0 to PND20, dams were exposed to a mixture "M" of polychlorinated biphenyls (PCB), pesticides, and methylmercury (MeHg), at 0.5 or 1 mg/kg/d (0.5M and M). At birth, some control (C) and M litters were cross-fostered to create the following in utero/postnatal exposure groups: C/C, M/C, C/M, M/M. Additional dams received 1.8 ng/kg/d of a mixture of aryl-hydrocarbon receptor (AhR) agonists (non-ortho-PCB, PC-dibenzodioxins, and PC-dibenzofurans) without or with 0.5M (0.5MAhR). Measurements of EC residue levels confirmed differences in their accumulation across treatments, age, and tissues. Although induction of hepatic detoxification enzyme activities (cytochrome P-450) demonstrated biological effects of treatments, the assessment of methylation in DNA repeats by sodium bisulfite pyrosequencing of liver, spleen, and thymus samples revealed no marked treatment-related effects but significant tissue- and age-related methylation differences. Further studies are required to determine whether absence of significant observable treatment effects on methylation of DNA repeats in the rat relate to tissue, strain, or species differences.


Subject(s)
DNA Methylation/drug effects , Environmental Pollutants/toxicity , Lactation , Long Interspersed Nucleotide Elements/drug effects , Maternal Exposure/adverse effects , Animals , Female , Male , Methylmercury Compounds/toxicity , Pesticides/toxicity , Polychlorinated Biphenyls/toxicity , Pregnancy , Rats , Rats, Sprague-Dawley , Sequence Analysis, DNA , Sulfites/chemistry
9.
Carcinogenesis ; 36 Suppl 1: S61-88, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26106144

ABSTRACT

Genome instability is a prerequisite for the development of cancer. It occurs when genome maintenance systems fail to safeguard the genome's integrity, whether as a consequence of inherited defects or induced via exposure to environmental agents (chemicals, biological agents and radiation). Thus, genome instability can be defined as an enhanced tendency for the genome to acquire mutations; ranging from changes to the nucleotide sequence to chromosomal gain, rearrangements or loss. This review raises the hypothesis that in addition to known human carcinogens, exposure to low dose of other chemicals present in our modern society could contribute to carcinogenesis by indirectly affecting genome stability. The selected chemicals with their mechanisms of action proposed to indirectly contribute to genome instability are: heavy metals (DNA repair, epigenetic modification, DNA damage signaling, telomere length), acrylamide (DNA repair, chromosome segregation), bisphenol A (epigenetic modification, DNA damage signaling, mitochondrial function, chromosome segregation), benomyl (chromosome segregation), quinones (epigenetic modification) and nano-sized particles (epigenetic pathways, mitochondrial function, chromosome segregation, telomere length). The purpose of this review is to describe the crucial aspects of genome instability, to outline the ways in which environmental chemicals can affect this cancer hallmark and to identify candidate chemicals for further study. The overall aim is to make scientists aware of the increasing need to unravel the underlying mechanisms via which chemicals at low doses can induce genome instability and thus promote carcinogenesis.


Subject(s)
Carcinogenesis/chemically induced , Carcinogens, Environmental/adverse effects , Environmental Exposure/adverse effects , Genomic Instability/drug effects , Hazardous Substances/adverse effects , Neoplasms/chemically induced , Neoplasms/etiology , Animals , Humans
10.
Int J Toxicol ; 31(5): 454-66, 2012.
Article in English | MEDLINE | ID: mdl-22914890

ABSTRACT

Exposure to environmental contaminants induces the activation of cytochrome P450s (CYP) which lead to the hydroxylation of contaminants and endogenous hormones such as estrogens. The hydroxylation of estrogens forms catecholestrogens (CEs), one of them being the mutagenic 4-hydroxyestradiol-17ß (4-OH-E2). Catecholestrogens are transformed by catechol-o-methyltransferases (COMTs) into nonreactive methoxyestrogens. To investigate the hepatic metabolism of estradiol-17ß in female offspring at postnatal day (PND) 21, pregnant rats were dosed daily from gestation day 1 until PND 21 with 2 dose levels of organochlorine pesticides (OCPs; 0.019 or 1.9 mg/kg per d), methylmercury (MeHg; 0.02 or 2 mg/kg per d), polychlorinated biphenyls (PCBs; 0.011 or 1.1 mg/kg per d), or a mixture (M; 0.05 or 5 mg/kg per d) including all 3 groups of chemicals. Concentrations of organochlorines in the mixture M were based on their proportions in serum of the Canadian Arctic population. The messenger RNA (mRNA) expressions of CYP and COMT were analyzed by quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR). High-performance thin layer chromatography and phosphor imaging were used to measure the transformation of (14)C substrates into estrogen metabolites. The low-dose treatments or the MeHg groups had no effect. The high-dose OCP, PCB, and M group increased the production of 2-OH-E2 and 6α-OH-E2, while only the PCB and M groups increased the 2-OH-CE/methoxyestrogen ratio. In all groups, the cytosolic COMT activity exceeded the microsomal production rate of 4-OH-E2. Although the M treatment included the PCB and OCP mixtures, it did not modify the estrogen metabolism more than did the PCB mixture alone. This endocrine disruption information contributes to our understanding of chemical interactions in the toxicology of chemical mixtures.


Subject(s)
Endocrine Disruptors/toxicity , Environmental Pollutants/toxicity , Estradiol/metabolism , Hydrocarbons, Chlorinated/toxicity , Methylmercury Compounds/toxicity , Pesticides/toxicity , Animals , Catechol O-Methyltransferase/genetics , Cytochrome P-450 Enzyme System/genetics , Female , Gene Expression Regulation, Enzymologic/drug effects , Liver/drug effects , Liver/metabolism , Maternal-Fetal Exchange , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Sprague-Dawley
11.
Int J Toxicol ; 30(3): 334-47, 2011 May.
Article in English | MEDLINE | ID: mdl-21444927

ABSTRACT

This investigation reports the effects of various terminal procedures, and how they modified the responses to a toxicant (polychlorinated biphenyls [A1254], 130 mg/kg/day × 5 days) administered by gavage to Sprague-Dawley male rats. Terminal procedures included exsanguination via the abdominal aorta under anesthesia (isoflurane inhalation or Equithesin injection), decapitation with or without anesthesia, or narcosis induced by carbon dioxide inhalation. Effects of repeated anesthesia were also tested. Terminal procedures induced confounding stress responses, particularly when Equithesin was used. The terminal procedures modified the conclusions about effects of A1254 on the concentrations of corticosterone, insulin, glucagon, glucose, alkaline phosphatase, lactate dehydrogenase, uric acid, and blood urea nitrogen, from nonstatistically significant to significant changes, and in the case of luteinizing hormone from a statistically significant increase to a significant decrease. Investigations of effects of toxicants should be designed and interpreted considering potential changes induced by the selection of a terminal procedure.


Subject(s)
Anesthetics/pharmacology , Polychlorinated Biphenyls/toxicity , Animals , Clinical Chemistry Tests , Cytochrome P-450 Enzyme System/metabolism , Glucose/metabolism , Liver/drug effects , Liver/enzymology , Male , Pituitary Hormones, Anterior/blood , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Thyroid Hormones/blood
12.
Int J Toxicol ; 28(4): 294-307, 2009.
Article in English | MEDLINE | ID: mdl-19636072

ABSTRACT

DNA methylation is one of the epigenetic mechanisms that regulates gene expression, chromosome structure, and stability. Our objective was to determine whether the DNA methylation system could be a target following in utero and postnatal exposure to human blood contaminants. Pregnant rats were dosed daily from gestation day 1 until postnatal day 21 with 2 dose levels of either organochlorine pesticides (OCP; 0.019 or 1.9 mg/kg/day), methylmercury chloride (MeHg; 0.02 or 2 mg/kg/day), polychlorinated biphenyls (PCBs; 0.011 or 1.1 mg/kg/day), or a mixture (Mix; 0.05, or 5 mg/kg/day) including all 3 groups of chemicals. Livers from 1 female offspring per litter were collected at postnatal day 29. Hepatic analysis revealed that the mRNA abundance for DNA methyltransferase (DNMT)-1, -3a, and -3b were significantly reduced by the high dose of PCB, that the high dose of MeHg also reduced mRNA levels for DNMT-1, and -3b, but that OCP had no significant effects compared with control. The high dose of PCB and Mix reduced the abundance of the universal methyl donor S-adenosylmethionine, and Mix also reduced global genome DNA methylation (5-methyl-deoxycytidine/5-methyl-deoxycytidine + deoxycytidine). The latter is consistent with pyrosequencing methylation analysis, revealing that the high-dose groups (except OCP) generally decreased the methylation of CpG sites (position -63 to -29) in the promoter of the tumor suppressor gene p16(INK4a). Overall, these hepatic results suggest that the DNA methylation system can be affected by exposure to high doses of blood contaminants, and that OCP is the least potent chemical group from the investigated mixtures.


Subject(s)
DNA Methylation/drug effects , Hydrocarbons, Chlorinated/toxicity , Liver/drug effects , Methylmercury Compounds/toxicity , Pesticides/toxicity , Polychlorinated Biphenyls/toxicity , Sexual Maturation , Animals , Base Sequence , Chromatography, Liquid , DNA Primers , Dose-Response Relationship, Drug , Female , Liver/metabolism , Pregnancy , Rats , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Electrospray Ionization , Spectrophotometry, Ultraviolet
13.
Int J Toxicol ; 24(2): 111-27, 2005.
Article in English | MEDLINE | ID: mdl-16036770

ABSTRACT

The postnatal period is a critical phase of development and a time during which humans are exposed to higher levels of persistent organic pollutants (POPs), than during subsequent periods of life. There is a paucity of information describing effects of postnatal exposure to environmentally relevant mixtures of POPs, such as polychlorinated biphenyls (PCBs), p,p'-dichlorodiphenyltrichloroethane (DDT), and p,p'-dichlorodiphenyldichloroethene (DDE). To provide data useful for the risk assessment of postnatal exposure to POPs, mixtures containing 19 PCBs, DDT, and DDE were prepared according to their concentrations previously measured in the milk of Canadian women, and dose-response effects were tested on the proliferation of MCF7-E3 cells in vitro, and in vivo experiments. Female neonates were exposed by gavage at postnatal days (PNDs) 1, 5, 10, 15, and 20 with dosages equivalent to 10, 100, and 1000 times the estimated human exposure level over the first 24 days of life. The MCF7-E3 cells showed a 227% increase in the AlamarBlue proliferation index, suggesting estrogen-like properties of the mixture, but this was not confirmed in vivo, given the absence of uterotrophic effects at PND21. An increase (511%) in hepatic ethoxyresorufin-o-deethylase activity at the dose 100 x was the most sensitive endpoint among those measured at PND21 (organ weight, mammary gland and ovarian morphometry, hepatic enzyme inductions, serum thyroxine and pituitary hormones). In liver samples from older female rats (previously involved in a mammary tumor study [Desaulniers et al., Toxicol. Sci. 75:468-480, 2001]), hepatic metabolism of 14C-estradiol-17beta (E2) at PND55 to PND62 was significantly higher in the 1000x compared to the control group, but hepatic detoxification enzyme activities had already returned to control values. The production of hepatic 2-hydroxy-E2 decreased, whereas that of estrone increased with age. In conclusion, the smallest dose of the mixture to induce significant effects was 100x, and mixture-induced changes in the hepatic metabolism of estrogens might be a sensitive indicator of persistent effects.


Subject(s)
Aging , Cell Proliferation/drug effects , Environmental Pollutants/toxicity , Liver/drug effects , Mammary Glands, Animal/drug effects , Ovary/drug effects , Administration, Oral , Aging/drug effects , Aging/metabolism , Aging/pathology , Animals , Animals, Newborn , Cell Line, Tumor , DDT/toxicity , Dichlorodiphenyl Dichloroethylene/toxicity , Dose-Response Relationship, Drug , Estradiol/metabolism , Female , Humans , Liver/metabolism , Mammary Glands, Animal/pathology , Ovary/pathology , Pituitary Hormones/blood , Polychlorinated Biphenyls/toxicity , Rats , Rats, Sprague-Dawley , Thyroxine/blood
14.
J Toxicol Environ Health A ; 67(18): 1457-75, 2004 Sep 24.
Article in English | MEDLINE | ID: mdl-15371232

ABSTRACT

There are concerns that early life exposure to organochlorines, including aryl hydrocarbon receptor (AhR) agonists, may lead to long-term effects and increase the risk of developing breast cancer. Our objective was to test if postnatal exposure to a mixture of 2,3,7,8-tetrachlorodibenzodioxin (TCDD)-like chemicals would modulate the development of methylnitrosourea (MNU)-induced mammary tumors. Females received by gavage a mixture containing 3 non-ortho-polychlorinated biphenyls (PCBs), 6 polychlorinated dibenzodioxins (PCDDs), and 7 polychlorinated dibenzofurans (PCDFs), at 1, 5, 10, 15, and 20d of age. The doses were equivalent to 0, 1, 10, 100, or 1000 times the amount ingested through breast milk by a human infant during its first 24 d of life. Subgroups of 1000 x reated rats and controls were sacrificed at 21 d of age for assessment of mammary-gland development, cell death, and proliferation. Mammary-tumor development was assessed in MNU (30 mg/kg body weight ip at 50 days of age)-induced rats pre-exposed to the mixture (MNU-0, MNU-1, MNU-10, MNU-100, MNU-1000). Rats were sacrificed when their mammary tumors reached 1 cm in diameter, or when the rats reached > or = 32 wk of age. Mammary-gland whole mounts were analyzed with all palpable and microscopic lesions (n = 1563) histologically classified and grouped as benign, intraductal proliferations, or malignant. There were no marked effects on age at onset of puberty (vaginal opening) and estrous cyclicity. Despite a significant decrease in proliferating cell nuclear antigen (PCNA)-positive mammary cells in 1000 x treated 21-d-old rats, there were no long-term dose-response effects on mammary-gland morphology and tumor development. In conclusion, postnatal exposure to the mixture of AhR agonists had no significant effects on the development of MNU-initiated mammary tumors.


Subject(s)
Alkylating Agents , Animals, Newborn , Disease Models, Animal , Environmental Exposure/adverse effects , Environmental Pollutants/toxicity , Mammary Neoplasms, Experimental/chemically induced , Methylnitrosourea , Polychlorinated Dibenzodioxins/analogs & derivatives , Polychlorinated Dibenzodioxins/toxicity , Receptors, Aryl Hydrocarbon/agonists , Age Factors , Animals , Benzofurans/toxicity , Body Weight/drug effects , Dibenzofurans, Polychlorinated , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Environmental Exposure/analysis , Estrous Cycle/drug effects , Immunohistochemistry , Mammary Neoplasms, Experimental/pathology , Milk/adverse effects , Milk/chemistry , Polychlorinated Biphenyls/toxicity , Proliferating Cell Nuclear Antigen/drug effects , Random Allocation , Rats , Rats, Sprague-Dawley , Risk Factors
15.
Toxicol Sci ; 67(1): 131-43, 2002 May.
Article in English | MEDLINE | ID: mdl-11961226

ABSTRACT

Human populations throughout the world are exposed daily to low levels of environmental contaminants. The consequences of potential interactions of these compounds to human endocrine, reproductive, and immune function remain unknown. The current study examines the effects of subchronic oral exposure to a complex mixture of ubiquitous persistent environmental contaminants that have been quantified in human reproductive tissues. The dosing solution used in this study contained organochlorines (2,3,7,8-tetrachlorodibenzo-p-dioxin [TCDD], polychlorinated biphenyls [PCBs],p,p'-dichlorodiphenoxydichloroethylene [p,p'-DDE],p,p-dichlorodiphenoxytrichloroethane [p,p'-DDT], dieldrin, endosulfan, methoxychlor, hexachlorobenzene, and other chlorinated benzenes, hexachlorocyclohexane, mirex and heptachlor) as well as metals (lead and cadmium). Each chemical was included in the mixture at the minimum risk level (MRL) or tolerable daily intake (TDI) as determined by the U.S. EPA or ATSDR or, for TCDD, at the no observable effect level (NOEL) used to calculate the TDI. Sexually mature male rats were exposed to this complex mixture at 1, 10, 100, and 1000 times the estimated safe levels daily for 70 days. On day 71, all animals were sacrificed and a variety of physiological systems assessed for toxic effects. Evidence of hepatotoxicity was seen in the significant enlargement of the liver in the 1000x group, reduced serum LDH activity (100x), and increased serum cholesterol and protein levels (both 1000x). Hepatic EROD activities were elevated in animals exposed to10x and above. The mixture caused decreased proliferation of splenic T cells at the highest dose and had a biphasic effect on natural killer cell lytic activity with an initial increase in activity at 1x followed by a decrease to below control levels in response to 1000x. No treatment-related effects were seen on bone marrow micronuclei, daily sperm production, serum LH, FSH, or prolactin levels or weights of most organs of the reproductive tract. The weights of the whole epididymis and of the caput epididymis were significantly decreased at 10x and higher doses, although no effect was seen on cauda epididymal weight. The sperm content of the cauda epididymis was increased at the 1x level but not significantly different from control at higher dose levels. A slight, but significant, increase in the relative numbers of spermatids was seen in the animals from the 1000x group with a trend towards reduced proportion of diploid cells at the same dose. Only minor, nondose related changes were seen in parameters related to condensation of chromatin, as determined by flow cytometry, in epididymal sperm. We conclude that the mixture induced effects on the liver and kidney and on general metabolism at high doses but caused only minor effects on immune function, reproductive hormone levels, or general indices of reproductive function measures. These data suggest that additive or synergistic effects of exposure to contaminants resulting in residue levels representative of contemporary human tissue levels are unlikely to result in adverse effects on immune function or reproductive physiology in male rats.


Subject(s)
Hazardous Substances/toxicity , Immune System/drug effects , Liver/drug effects , Reproduction/drug effects , Xenobiotics/toxicity , Animals , Body Weight/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Dose-Response Relationship, Drug , Drug Combinations , Female , Insecticides/toxicity , Liver/enzymology , Liver/pathology , Male , Metals, Heavy/toxicity , Micronucleus Tests , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Rats , Rats, Sprague-Dawley , Toxicity Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...