Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Radiat Res ; 182(5): 556-72, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25251702

ABSTRACT

We present here a methodology for health risk assessment adopted by the World Health Organization that provides a framework for estimating risks from the Fukushima nuclear accident after the March 11, 2011 Japanese major earthquake and tsunami. Substantial attention has been given to the possible health risks associated with human exposure to radiation from damaged reactors at the Fukushima Daiichi nuclear power station. Cumulative doses were estimated and applied for each post-accident year of life, based on a reference level of exposure during the first year after the earthquake. A lifetime cumulative dose of twice the first year dose was estimated for the primary radionuclide contaminants ((134)Cs and (137)Cs) and are based on Chernobyl data, relative abundances of cesium isotopes, and cleanup efforts. Risks for particularly radiosensitive cancer sites (leukemia, thyroid and breast cancer), as well as the combined risk for all solid cancers were considered. The male and female cumulative risks of cancer incidence attributed to radiation doses from the accident, for those exposed at various ages, were estimated in terms of the lifetime attributable risk (LAR). Calculations of LAR were based on recent Japanese population statistics for cancer incidence and current radiation risk models from the Life Span Study of Japanese A-bomb survivors. Cancer risks over an initial period of 15 years after first exposure were also considered. LAR results were also given as a percentage of the lifetime baseline risk (i.e., the cancer risk in the absence of radiation exposure from the accident). The LAR results were based on either a reference first year dose (10 mGy) or a reference lifetime dose (20 mGy) so that risk assessment may be applied for relocated and non-relocated members of the public, as well as for adult male emergency workers. The results show that the major contribution to LAR from the reference lifetime dose comes from the first year dose. For a dose of 10 mGy in the first year and continuing exposure, the lifetime radiation-related cancer risks based on lifetime dose (which are highest for children under 5 years of age at initial exposure), are small, and much smaller than the lifetime baseline cancer risks. For example, after initial exposure at age 1 year, the lifetime excess radiation risk and baseline risk of all solid cancers in females were estimated to be 0.7 · 10(-2) and 29.0 · 10(-2), respectively. The 15 year risks based on the lifetime reference dose are very small. However, for initial exposure in childhood, the 15 year risks based on the lifetime reference dose are up to 33 and 88% as large as the 15 year baseline risks for leukemia and thyroid cancer, respectively. The results may be scaled to particular dose estimates after consideration of caveats. One caveat is related to the lack of epidemiological evidence defining risks at low doses, because the predicted risks come from cancer risk models fitted to a wide dose range (0-4 Gy), which assume that the solid cancer and leukemia lifetime risks for doses less than about 0.5 Gy and 0.2 Gy, respectively, are proportional to organ/tissue doses: this is unlikely to seriously underestimate risks, but may overestimate risks. This WHO-HRA framework may be used to update the risk estimates, when new population health statistics data, dosimetry information and radiation risk models become available.


Subject(s)
Fukushima Nuclear Accident , Neoplasms, Radiation-Induced/etiology , Adolescent , Adult , Age Factors , Aged , Child , Child, Preschool , Female , Humans , Infant , Japan , Male , Middle Aged , Radiation Dosage , Risk , Time Factors , Young Adult
2.
Rev Sci Tech ; 32(2): 459-67, 2013 Aug.
Article in English | MEDLINE | ID: mdl-24547649

ABSTRACT

Foodborne diseases are a multi-sectoral public health risk closely linked with the agricultural and animal health sectors. Many foodborne diseases are zoonotic in nature. The World Health Organization (WHO) seeks to measure for the first time the real impact of foodborne diseases through the advice of its independent expert body, the Foodborne Disease Burden Epidemiology Reference Group (FERG). Through the FERG, the WHO works on both assembling and appraising existing data as well as supporting countries in conducting their own studies into the national burden of foodborne disease. This is complemented by efforts to ensure thatthe findings are meaningful and useable to policy-makers and other research end-users to implement informed policy and interventions. For the Initiative to operate effectively and achieve its objectives, linkages and collaboration at all levels, especially at the human-animal interface, need to be fostered.


Subject(s)
Food Safety/methods , Foodborne Diseases/epidemiology , Population Surveillance/methods , World Health Organization , Zoonoses/prevention & control , Animals , Humans , Public Health Administration , Zoonoses/epidemiology
3.
World Health Organ Tech Rep Ser ; (947): 1-225, back cover, 2007.
Article in English | MEDLINE | ID: mdl-18551832

ABSTRACT

This report represents the conclusions of a Joint FAO/WHO Expert Committee convened to evaluate the safety of various food additives, including flavouring agents, with a view to recommending acceptable daily intakes (ADIs) and to preparing specifications for identity and purity. The Committee also evaluated the risk posed by two food contaminants, with the aim of advising on risk management options for the purpose of public health protection. The first part of the report contains a general discussion of the principles governing the toxicological evaluation and assessment of intake of food additives (in particular flavouring agents) and contaminants. A summary follows of the Committee's evaluations of technical, toxicological and intake data for certain food additives (acidified sodium chlorite, asparaginase from Aspergillus oryzae expressed in Aspergillus oryzae, carrageenan and processed Eucheuma seaweed, cyclotetraglucose and cyclotetraglucose syrup, isoamylase from Pseudomonas amyloderamosa, magnesium sulfate, phospholipase A1 from Fusarium venenatum expressed in Aspergillus oryzae, sodium iron(III) ethylenediaminetetraacetic acid (EDTA) and steviol glycosides); eight groups of related flavouring agents (linear and branched-chain aliphatic, unsaturated, unconjugated alcohols, aldehydes, acids and related esters; aliphatic acyclic and alicyclic terpenoid tertiary alcohols and structurally related substances; simple aliphatic and aromatic sulfides and thiols; aliphatic acyclic dials, trials and related substances; aliphatic acetals; sulfur-containing heterocyclic compounds; aliphatic and aromatic amines and amides; and aliphatic alicyclic linear alpha, beta -unsaturated di- and trienals and related alcohols, acids and esters); and two food contaminants (aflatoxin and ochratoxin A). Specifications for the following food additives were revised: maltol and ethyl maltol, nisin preparation, pectins, polyvinyl alcohol, and sucrose esters of fatty acids. Specifications for the following flavouring agents were revised: maltol and ethyl maltol, maltyl isobutyrate, 3-acetyl-2,5-dimethylfuran and 2,4,5-trimethyl-delta-oxazoline (Nos 1482, 1506 and 1559), and monomenthyl glutarate (No. 1414), as well as the method of assay for the sodium salts of certain flavouring agents. Annexed to the report are tables summarizing the Committee's recommendations for intakes and toxicological evaluations of the food additives and contaminants considered.


Subject(s)
Consumer Product Safety , Food Additives/adverse effects , Food Additives/analysis , Food Contamination/analysis , Nutrition Policy , Animals , Flavoring Agents/adverse effects , Flavoring Agents/analysis , Food Coloring Agents/adverse effects , Food Coloring Agents/analysis , Humans , Risk Assessment , Risk Management , Safety , United Nations , World Health Organization
4.
Food Chem Toxicol ; 44(10): 1636-50, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16891049

ABSTRACT

The European Food Safety Authority (EFSA) and the World Health Organization (WHO), with the support of the International Life Sciences Institute, European Branch (ILSI Europe), organized an international conference on 16-18 November 2005 to discuss how regulatory and advisory bodies evaluate the potential risks of the presence in food of substances that are both genotoxic and carcinogenic. The objectives of the conference were to discuss the possible approaches for risk assessment of such substances, how the approaches may be interpreted and whether they meet the needs of risk managers. ALARA (as low as reasonably achievable) provides advice based solely on hazard identification and does not take into account either potency or human exposure. The use of quantitative low-dose extrapolation of dose-response data from an animal bioassay raises numerous scientific uncertainties related to the selection of mathematical models and extrapolation down to levels of human exposure. There was consensus that the margin of exposure (MOE) was the preferred approach because it is based on the available animal dose-response data, without extrapolation, and on human exposures. The MOE can be used for prioritisation of risk management actions but the conference recognised that it is difficult to interpret it in terms of health risk.


Subject(s)
Carcinogens/toxicity , Food/standards , Mutagens/toxicity , Animals , Carcinogenicity Tests , Europe , Foodborne Diseases/etiology , Foodborne Diseases/genetics , Humans , Mutagenicity Tests , Risk Assessment , World Health Organization
6.
Food Chem Toxicol ; 40(2-3): 237-82, 2002.
Article in English | MEDLINE | ID: mdl-11893399

ABSTRACT

Hazard characterisation of low molecular weight chemicals in food and diet generally use a no-observed-adverse-effect level (NOAEL) or a benchmark dose as the starting point. For hazards that are considered not to have thresholds for their mode of action, low-dose extrapolation and other modelling approaches may be applied. The default position is that rodents are good models for humans. However, some chemicals cause species-specific toxicity syndromes. Information on quantitative species differences is used to modify the default uncertainty factors applied to extrapolate from experimental animals to humans. A central theme for extrapolation is unravelling the mode of action for the critical effects observed. Food can be considered as an extremely complex and variable chemical mixture. Interactions among low molecular weight chemicals are expected to be rare given that the exposure levels generally are far below their NOAELs. Hazard characterisation of micronutrients must consider that adverse effects may arise from intakes that are too low (deficiency) as well as too high (toxicity). Interactions between different nutrients may complicate such hazard characterisations. The principle of substantial equivalence can be applied to guide the hazard identification and hazard characterisation of macronutrients and whole foods. Macronutrients and whole foods must be evaluated on a case-by-case basis and cannot follow a routine assessment protocol.


Subject(s)
Hazardous Substances/toxicity , Micronutrients/adverse effects , Animals , Dose-Response Relationship, Drug , European Union , Hazardous Substances/administration & dosage , Humans , Micronutrients/administration & dosage , Models, Animal , Molecular Weight , No-Observed-Adverse-Effect Level , Risk Assessment/methods , Rodentia , Species Specificity
7.
Toxicol Pathol ; 28(6): 761-9, 2000.
Article in English | MEDLINE | ID: mdl-11127289

ABSTRACT

2,3,7,8,-Tetrachlorodibenzo-p-dioxin (TCDD) has been classified as a known human carcinogen, and epidemiologic studies identify the lung as one of the target organs. Few experimental studies have attempted to characterize pulmonary effects of TCDD exposure. In this study, we characterize the induction of lesions in the lung by chronic oral TCDD exposure in diethylnitrosamine (DEN)-initiated or noninitiated female Sprague-Dawley rats. Two or 18 weeks after initiation, rats were treated with TCDD continuously for 14, 30, or 60 weeks by biweekly oral gavage (1,750 ng TCDD/kg) at a dose equivalent to 125 ng/kg body weight per day (controls received corn oil). To assess the time dependence and reversibility of potential changes, some groups included withdrawal periods of 16 or 30 weeks after 30 weeks of TCDD treatment. TCDD treatment alone for 60 weeks caused significant increases in alveolar-bronchiolar (AB) metaplasia. TCDD treatment of DEN-initiated animals for 60 weeks resulted in a significant increase in bronchiolar epithelial hyperplasia. These increases were not observed in animals treated with TCDD for 30 weeks followed by corn oil for 30 weeks, indicating that the development of these lesions required continuous exposure to TCDD. AB hyperplasia increased in an age-dependent manner after DEN initiation but was unaffected by TCDD treatment. Expression of the aromatic hydrocarbon receptor (AHR) and induction of CYP1A1 was observed only in bronchiolar Clara and ciliated cells, indicating that the mechanism of induction of AB metaplasia may be mediated by the AHR. TCDD elimination half-life was monophasic in the lung, and serum and was estimated to be 39.7 days and 44.6 days, respectively, independent of age, tissue TCDD concentration, or body weight. This is the first report to identify the AB region as a target for TCDD-induced metaplastic and proliferative changes after chronic oral exposure.


Subject(s)
Bronchi/drug effects , Carcinogens/toxicity , Polychlorinated Dibenzodioxins/toxicity , Pulmonary Alveoli/drug effects , Respiratory Mucosa/drug effects , Adenoma/chemically induced , Adenoma/pathology , Age Factors , Animals , Bronchi/metabolism , Bronchi/pathology , Carcinogenicity Tests , Carcinogens/administration & dosage , Carcinogens/pharmacokinetics , Carcinoma/chemically induced , Carcinoma/pathology , Cytochrome P-450 CYP1A1/metabolism , Diethylnitrosamine/toxicity , Drug Administration Schedule , Female , Half-Life , Hyperplasia , Lung Neoplasms/chemically induced , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Metaplasia , Polychlorinated Dibenzodioxins/administration & dosage , Precancerous Conditions/chemically induced , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Rats , Rats, Sprague-Dawley , Receptors, Aryl Hydrocarbon/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Tissue Distribution
8.
Cancer Res ; 60(19): 5414-9, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11034082

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a multispecies reproductive toxicant, and it has been recently classified by IARC as a known human carcinogen. Here, we report that TCDD promotes the development of ovarian tumors in an initiation-promotion model in female Sprague Dawley rats. Rats were initiated with diethylnitrosamine (DEN) or vehicle at 70 days of age. Starting 2 or 18 weeks after initiation, rats were exposed biweekly to TCDD at a daily average dose of 125 ng/kg/day for 14, 30, or 60 weeks continuously or for 30 weeks plus withdrawal periods of 16 or 30 weeks. Fifteen of 76 (20%) rats initiated with DEN and promoted with TCDD for various lengths of time developed ovarian sex cord-stromal tumors of Sertoli cell type, whereas no ovarian tumors developed in 86 rats used as vehicle controls or that received DEN alone or TCDD alone. The highest tumor incidence occurred in 6 of 14 rats (43%) after 60 weeks of continuous TCDD after DEN initiation. One of six rats developed a tumor by 30 weeks of exposure. Because most effects of TCDD can be attributed to its activation of the aryl hydrocarbon receptor (AhR), the presence and localization of AhR was determined in the rat ovary and in the ovarian tumors by reverse transcription-PCR, immunohistochemistry, and in situ hybridization. AhR was localized to oocytes, granulosa and thecal cells of growing follicles, surface epithelial cells, and epithelial cells lining single tubules in ovaries from adult control Sprague Dawley rats. Neoplastic cells in the ovarian tumors were also positive for both AhR message and protein. These results indicate that the ability of TCDD to cause ovarian tumors is dependent on initiation, length of promotion, and age of the animal when exposed and evaluated. The tumor type induced by TCDD in this experimental system is the same histological subtype as that reported from an early study of youngsters exposed during an industrial accident in Seveso, Italy.


Subject(s)
Carcinogens, Environmental/toxicity , Ovarian Neoplasms/chemically induced , Polychlorinated Dibenzodioxins/toxicity , Animals , Carcinogens , Diethylnitrosamine , Drug Administration Schedule , Drug Synergism , Environmental Pollutants/toxicity , Estradiol/blood , Female , Ovarian Neoplasms/pathology , Ovary/drug effects , Progesterone/blood , Rats , Rats, Sprague-Dawley , Receptors, Aryl Hydrocarbon/physiology , Sertoli Cell Tumor/chemically induced , Sertoli Cell Tumor/pathology
9.
Toxicol Sci ; 54(2): 330-7, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10774815

ABSTRACT

In this study, we investigated the time course of promotion of tumors and putatively preneoplastic altered hepatic foci in the livers of diethylnitrosamine (DEN)-initiated female Sprague-Dawley rats. These rats had been treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) under different dosing regimens, but we used the same administered biweekly dose of 1.75 microg/kg of body weight. Animals were treated continuously for up to 60 weeks, or continuously for 30 weeks, followed by cessation of treatment for up to 30 weeks. In addition, TCDD treatment in these groups was begun either 2 or 18 weeks after initiation with DEN. Liver tumors were only observed in animals after 60 weeks on the study and were increased by continuous TCDD treatment, relative to controls. The incidence of hepatocellular adenoma and carcinoma combined, in animals treated with TCDD for 30 weeks followed by no TCDD treatment for 30 weeks (17%), was lower than in animals receiving either TCDD (79%) or vehicle control (corn oil) alone (55%) for 60 weeks. The lower liver-tumor incidence after cessation of TCDD treatment paralleled time-dependent decreases in the volume fraction occupied by placental glutathione S-transferase-positive altered hepatic foci and the number of foci per unit volume, but not the mean focus volume that exhibited a time-dependent increase after cessation of TCDD treatment. Cessation of TCDD treatment led to reductions in liver TCDD levels, and these changes were reflected in a cessation of reduced body weight because of TCDD treatment. These data indicate that liver-tumor promotion by TCDD in female rats is dependent upon continuous exposure to TCDD, and that alterations in patterns of TCDD exposure can have significant effects on tumor incidence not reflected by standard measures of dioxin exposure.


Subject(s)
Adenoma/chemically induced , Carcinogens/toxicity , Carcinoma/chemically induced , Liver Neoplasms, Experimental/chemically induced , Polychlorinated Dibenzodioxins/toxicity , Precancerous Conditions/chemically induced , Animals , Carcinogens/administration & dosage , Carcinogens/pharmacokinetics , Diethylnitrosamine/toxicity , Drug Administration Schedule , Female , Glutathione Transferase/metabolism , Half-Life , Image Processing, Computer-Assisted , Liver/drug effects , Liver/enzymology , Liver/pathology , Liver Neoplasms, Experimental/enzymology , Liver Neoplasms, Experimental/pathology , Organ Size/drug effects , Polychlorinated Dibenzodioxins/administration & dosage , Polychlorinated Dibenzodioxins/pharmacokinetics , Precancerous Conditions/enzymology , Precancerous Conditions/pathology , Rats , Rats, Sprague-Dawley
10.
Carcinogenesis ; 19(8): 1427-35, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9744539

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a potent tumor promoter in two-stage initiation-promotion models and induces cell proliferation and development of enzyme-altered hepatic foci. It is believed that increased cell proliferation is a necessary step in carcinogenesis. Therefore, the analysis of the effect of TCDD on cell proliferation in rat liver may aid in the understanding of the mechanism of hepatocarcinogenesis induced by TCDD. The aim of this study was to investigate the time course and reversibility of cell proliferation in non-initiated and diethylnitrosamine-initiated female rats exposed biweekly to a daily averaged dose of 125 ng TCDD/kg/day for up to 60 weeks. In addition we evaluated the suitability of different dose metrics for the evaluation of TCDD-induced changes in cell proliferation and CYP1A1 enzyme induction. Cell proliferation was measured as the incorporation of 5-bromo-2'-deoxyuridine (BrdU) into hepatocytes undergoing replicative DNA synthesis. Mean BrdU labeling indices in TCDD-treated animals were not increased over controls after 14 weeks exposure, but were increased 8- and 2-fold after 30 and 60 weeks' treatment respectively, despite similar liver levels of TCDD at all these times (23-30 p.p.b.). In comparison, CYP1A1 activity, as measured by ethoxyresorufin deethylase activity, was significantly induced at all times points analyzed. Sixteen weeks following cessation of TCDD treatment, labeling indices were still significantly elevated over controls, but after 30 weeks of withdrawal, labeling indices were no different from controls, indicating that TCDD-induced changes in cell proliferation were reversible. Dosimetric analysis indicated that rat liver tissue burden was suitable for prediction of CYP1A1 expression but not cell proliferation and that the area under the curve was unsuitable for prediction of both TCDD-induced changes in CYP1A1 expression and cell proliferation.


Subject(s)
Cytochrome P-450 CYP1A1/metabolism , Liver/drug effects , Polychlorinated Dibenzodioxins/metabolism , Animals , Bromodeoxyuridine/metabolism , Carcinogens , Cell Division/drug effects , Diethylnitrosamine , Enzyme Induction , Female , Liver/metabolism , Organ Size/drug effects , Polychlorinated Dibenzodioxins/toxicity , ROC Curve , Rats , Rats, Sprague-Dawley
11.
Carcinogenesis ; 17(12): 2609-15, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9006096

ABSTRACT

The differential display technique was used to identify genes whose expression was regulated by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Expression of a novel sequence was up-regulated in a dose-dependent fashion in liver of Sprague-Dawley male rats exposed to both chronic and acute treatment with TCDD, as measured by densitometry of Northern blot analyses (P < 0.01). A rapid amplification of cDNA ends (RACE) procedure was used to isolate a 1.8 kb cDNA from a rat liver cDNA preparation. This cloned cDNA, called 25-Dx, was sequenced and found to encode a peptide of 223 amino acids. In control rats, the 25-Dx gene was expressed at high levels in lung and liver. A hydrophobic domain of 14 residues followed by a proline-rich domain, both located in the N-terminal region, showed 71% homology with the transmembrane domain of the precursor for the interleukin-6 receptor and a conserved consensus sequence found in the cytokine/growth factor/prolactin receptor superfamily respectively.


Subject(s)
DNA, Complementary/isolation & purification , Liver/drug effects , Polychlorinated Dibenzodioxins/toxicity , Amino Acid Sequence , Animals , Base Sequence , Liver/metabolism , Male , Molecular Sequence Data , Polymerase Chain Reaction , Protein Biosynthesis , Rats , Rats, Sprague-Dawley
12.
Cancer Lett ; 98(2): 219-25, 1996 Jan 02.
Article in English | MEDLINE | ID: mdl-8556712

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a more potent hepatocarcinogen in female than in male or ovariectomized rats. A possible mechanism for this increased sensitivity is through enhanced metabolic activation of estrogens by TCDD-induced enzymes leading to oxidative damage in the cell. As a marker for oxidative DNA damage, 8-oxo-deoxyguanosine (8-oxo-dG) was quantitated in livers of intact and ovariectomized Sprague-Dawley rats chronically treated with TCDD (125 ng/kg per day) with and without diethylnitrosamine as initiator. Elevated levels of 8-oxo-dG were detected in a significantly greater number of the intact compared to ovariectomized TCDD-treated rats. Expression of CYP1B1 mRNA, a newly identified cytochrome P450 with proposed estrogen hydroxylase activity, was highly induced by TCDD. The results are consistent with the hypothesis that increased metabolism of endogenous estrogens to catechols by TCDD-induced enzymes may lead to increased oxidative DNA damage and hence contribute to TCDD-mediated hepatocarcinogenicity in female rats.


Subject(s)
Cocarcinogenesis , DNA Damage , DNA/metabolism , Liver Neoplasms, Experimental/chemically induced , Liver/drug effects , Ovary/physiology , Polychlorinated Dibenzodioxins/toxicity , Animals , Carcinogens/toxicity , DNA/drug effects , Deoxyguanosine/metabolism , Diethylnitrosamine/toxicity , Estradiol/physiology , Female , Liver/metabolism , Liver Neoplasms, Experimental/metabolism , Ovariectomy , Oxidation-Reduction , Rats , Rats, Sprague-Dawley
13.
Clin Chem ; 41(12 Pt 2): 1829-34, 1995 Dec.
Article in English | MEDLINE | ID: mdl-7497640

ABSTRACT

Numerous xenobiotics regulate cellular functions by altering transcription of target genes. Use of sensitive and specific biomarkers based on gene transcript levels may help clarify the shape of the dose-response curve in the low-dose region associated with human exposures to environmental concentrations of chemicals. We have quantified gene transcription induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin in both animal models and humans with the use of Northern analysis and PCR-based methods. In addition, we describe a rapid and sensitive in vitro assay that we have used to screen chemicals and biological samples for their ability to alter gene transcription. Whereas some of the endpoints in our studies such as cytochrome P-450 1A1 are predictive indicators of exposure and dose, other gene responses such as growth factors are more complex and represent a critical event, progression, or adaptation to a pathological alteration. In conclusion, measurement of toxicant-induced gene transcription will contribute to the usefulness of biomarkers in addressing issues of human health and environmentally induced disease.


Subject(s)
Biomarkers , Gene Expression Regulation, Enzymologic/drug effects , Polychlorinated Dibenzodioxins/adverse effects , Transcription, Genetic/drug effects , Animals , Blotting, Northern , Cytochrome P-450 CYP1A1 , Cytochrome P-450 Enzyme System/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Environmental Exposure/adverse effects , Environmental Health , ErbB Receptors/drug effects , ErbB Receptors/genetics , Gene Expression Regulation, Enzymologic/genetics , Humans , Oxidoreductases/metabolism , Polymerase Chain Reaction , Transcription, Genetic/genetics
14.
Carcinogenesis ; 16(11): 2807-11, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7586202

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a potent tumor promoter in two-stage models of hepatocarcinogenesis. This study focuses on the persistence or reversibility of TCDD-mediated changes in livers after 30 weeks of treatment and cessation of treatment. Diethylnitrosamine (DEN) initiated animals (175 mg/kg) were promoted bi-weekly with TCDD at a dose equivalent to 125 ng/kg/day for 30 weeks without or with a following waiting period of 32 weeks before necropsy. 2,3,7,8-Tetrachlorodibenzo-p-dioxin liver concentration decreased 300-fold above background. Induction of CYP1A1 dependent enzyme activity decreased according to TCDD tissue levels. In contrast, cell proliferation, as measured by BrdU-labeling index, was still 2.8-fold increased over controls in the TCDD group with waiting period compared to a 4-fold increase over controls at the end of the 30 week dosing period. Enzyme altered hepatic foci expressing the placental form of glutathione S-transferase decreased in number but the remaining foci were significantly increased in size and the percent of liver occupied by foci was higher at the end of the waiting period as compared to livers at the end of the dosing period. Liver tumor incidence at the end of the waiting period was 71% (5 of 7 animals) and the livers showed an increase in bile duct lesions with only mild toxicity. There was pronounced bile duct proliferation in DEN/TCDD treated animals after the waiting period with intense expression of TGF alpha in bile duct epithelial cells at detected by immunohistochemical methods. In comparison, at the end of the 30 week dosing period the livers showed more severe toxicity and only mild bile duct proliferation. Also, one small hepatocellular adenoma was observed. It is concluded that as opposed to CYP1A1 induction the more complex biological responses, cell proliferation and selective growth of certain preneoplastic foci, are persistent after prolonged TCDD treatment within the experimental framework of our study.


Subject(s)
Carcinogens/toxicity , Liver Neoplasms, Experimental/chemically induced , Liver/drug effects , Polychlorinated Dibenzodioxins/toxicity , Animals , Cell Division/drug effects , Diethylnitrosamine , Female , Glutathione Transferase/metabolism , Liver/enzymology , Liver/pathology , Polychlorinated Dibenzodioxins/metabolism , Rats , Rats, Sprague-Dawley
15.
Toxicol Appl Pharmacol ; 132(2): 237-44, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7540335

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a multisite carcinogen. Although the hepatocarcinogenic actions of TCDD have received the most attention, it has been demonstrated in several rodent carcinogenicity bioassays that TCDD causes a dose-related increase in thyroid follicular cell adenomas and carcinomas. The purpose of the present experiment was to investigate the dose-response relationship for thyroid function alterations in female Sprague-Dawley rats following chronic treatment with TCDD. TCDD was administered via oral gavage biweekly for 30 weeks at average daily equivalent doses of 0.1-125 ng/kg/day, thereby more than encompassing the dose range historically used in previous TCDD rodent bioassays. The endpoints examined include serum levels of thyroxine (T4), triiodothyronine (T3), and thyroid-stimulating hormone (TSH). In addition, the induction of the dioxin-responsive genes UDP-glucuronosyltransferase-1 (UGT1) and cytochrome P450 1A1 (CYP1A1) in liver were measured using reverse-transcriptase-polymerase chain reaction (RT-PCR). In agreement with previous hypotheses, TCDD appears to alter thyroid function via a secondary mechanism, namely increased excretion of T4-glucuronide resulting from TCDD induction of UGT1. The observed follicular cell hyperplasia and hypertrophy are consistent with the observed elevated TSH levels and may represent the early stages in the progression of thyroid carcinogenesis. Therefore, TCDD induces alterations in thyroid hormone function, probably as a result of chronic perturbations of liver-pituitary-thyroid axis.


Subject(s)
Polychlorinated Dibenzodioxins/toxicity , Thyroid Hormones/biosynthesis , Thyrotropin/biosynthesis , Animals , Antithyroid Agents/metabolism , Base Sequence , Cytochrome P-450 Enzyme System/genetics , Enzyme Induction/genetics , Female , Gene Expression Regulation/genetics , Glucuronosyltransferase/genetics , Molecular Sequence Data , Polychlorinated Dibenzodioxins/metabolism , Polymerase Chain Reaction , RNA, Messenger/metabolism , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Thyroid Gland/drug effects , Thyroid Gland/pathology , Thyroxine/antagonists & inhibitors , Thyroxine/biosynthesis , Triiodothyronine/antagonists & inhibitors , Triiodothyronine/biosynthesis
16.
Fundam Appl Toxicol ; 23(3): 465-9, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7835546

ABSTRACT

Polychlorinated dibenzo-p-dioxins (PCDDs), dibenzofurans (PCDFs), and biphenyls belong to a class of compounds, the polyhalogenated aromatic hydrocarbons (PHAHs), which are ubiquitous environmental contaminants. Due to the existence of a common mechanism of action, i.e., binding to the Ah receptor, the activity of members of this class of compounds is generally expressed relative to the prototypical 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) as toxic equivalency factors (TEFs). In the present studies we examined the presence of liver of untreated PCDFs in standard laboratory feed and in the liver of untreated rats at three different ages (60, 140, and 200 days) in terms of concentration and in toxic equivalents (TEQs, TEF x concentration). Feed was shown to contain trace amounts of PCDDs and PCDFs and control rat liver was shown to contain several PCDD and PCDF congeners in terms of concentration of congener and concentration of TEQs contributed by that congener. The total concentration of TEQs increased with increasing age in rat liver, going from 20 ppt TEQ at 60 days to 78 ppt TEQ at 200 days of age. This accumulation in dioxin-like activity was due primarily to PCDFs. In particular the congener 2,3,4,7,8-pentachlorodibenzofuran accrued in untreated rat liver accounting for approximately 80% of the total TEQ at 200 days of age. These studies affirm the pervasive presence of PHAHs and suggest prudence in evaluating chronic rat studies in which interference from background levels of PCDDs and PCDFs may be a factor.


Subject(s)
Benzofurans/pharmacokinetics , Liver/metabolism , Polychlorinated Dibenzodioxins/analogs & derivatives , Age Factors , Animals , Cytochrome P-450 Enzyme System/physiology , Dibenzofurans, Polychlorinated , Female , Polychlorinated Dibenzodioxins/pharmacokinetics , Rats , Rats, Sprague-Dawley
18.
Cancer Res ; 54(1): 62-8, 1994 Jan 01.
Article in English | MEDLINE | ID: mdl-8261464

ABSTRACT

The purpose of the present experiments was to examine dose-response relationships for induction of hepatic mRNA following a single administration of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) to rats. The induction of cytochrome P450-1A1 (CYP1A1) mRNA is compared to other "dioxin-responsive" genes including UDP-glucuronosyltransferase I, plasminogen activator inhibitor 2, and transforming growth factor alpha using a sensitive reverse transcriptase-polymerase chain reaction-based method. Sample-to-sample variability in amplification is a concern in using polymerase chain reaction to quantitate biological responses. However, in the present study recombinant RNA templates were synthesized to use as internal standards in both the reverse transcription and the polymerase chain reaction steps. The induction of CYP1A1 mRNA was extremely sensitive to TCDD treatment with increases observed at doses as low as 1 ng/kg body weight. The induction of CYP1A1 mRNA correlated highly (R2 > 0.90) with an increase in ethoxyresorufin-o-deethylase activity, a CYP1A1-associated enzyme activity. However, induction of CYP1A1 mRNA levels was detected at lower TCDD doses than was ethoxyresorufin-o-deethylase activity, reflecting the greater sensitivity of the reverse transcription-polymerase chain reaction approach to detect transcriptional activation of the CYP1A1 gene. UDP-glucuronosyltransferase I mRNA was increased over control (5-fold) but required 1000-times higher TCDD doses (1 microgram/kg) to result in a significant increase than did CYP1A1. Plasminogen activator inhibitor 2 and transforming growth factor alpha mRNA, both previously shown to be induced by TCDD in human keratinocytes, were not increased in rat liver. Hence, these studies reaffirm that TCDD acts through classical receptor mechanisms with gene-to-gene differences in responsiveness. The reverse transcription-polymerase chain reaction method developed to measure mRNA for dioxin-responsive genes in rat liver will allow for measuring multigene and tissue responses to TCDD and other xenobiotics with high sensitivity, reproducibility, and adaptability and should increase our understanding of various dose-response relationships.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Liver/enzymology , Polychlorinated Dibenzodioxins/toxicity , RNA, Messenger/biosynthesis , Animals , Base Sequence , Cytochrome P-450 CYP1A1 , Dose-Response Relationship, Drug , Enzyme Induction/drug effects , Female , Glucuronosyltransferase/biosynthesis , Liver/drug effects , Molecular Sequence Data , Oxidoreductases/biosynthesis , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley
19.
Environ Health Perspect ; 101(7): 634-42, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8143597

ABSTRACT

The present study examines the dose-response relationship for 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) promotion of histologic and biochemical parameters by using a two-stage model for hepatocarcinogenesis in female Sprague-Dawley rats initiated with a single intraperitoneal dose of 175 mg of diethylnitrosamine (DEN)/kg body weight at 70 days of age. Starting 2 weeks after initiation, treatment groups of 8-10 rats were given TCDD by gavage in corn oil once every 2 weeks for 30 weeks. Doses were 3.5, 10.7, 35.7, and 125 ng TCDD/kg body weight/day. A significant body weight reduction was present in the noninitiated group that received 125 ng TCDD. Relative liver weight was statistically increased in initiated rats treated with > or = 10.7 ng TCDD and in noninitiated rats treated with > or = 35.7 ng TCDD. Histopathologic evidence of cytotoxicity was dose-related in all TCDD-treated groups. There was a statistically significant dose response in the bromodeoxyuridine (BrdU) S-phase labeling index (LI) in the DEN-initiated rats (p < 0.01) and a marginally significant trend in the saline-treated rats (p = 0.10), but proliferating cell nuclear antigen S-phase LI and growth fraction within altered hepatic foci showed no increase. Among the DEN-initiated groups there was a significant increase in glutathione S-transferase altered hepatic foci stereological parameters in the 125 ng TCDD group. This study demonstrates that dose-response relationships for TCDD's effects on cell proliferation growth of altered hepatic foci are different from previously reported effects on P450 gene expression, indicating that different biological or biochemical responses may exhibit different dose-response relationships.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Liver Neoplasms, Experimental/chemically induced , Polychlorinated Dibenzodioxins , Animals , Body Weight , Cell Division , Diethylnitrosamine , Dose-Response Relationship, Drug , Female , Glutathione Transferase/metabolism , Isoenzymes/metabolism , Liver/metabolism , Liver/pathology , Liver Neoplasms, Experimental/pathology , Organ Size , Osmolar Concentration , Placenta/enzymology , Polychlorinated Dibenzodioxins/metabolism , Rats , Rats, Sprague-Dawley
20.
Carcinogenesis ; 14(9): 1885-93, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8403215

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a potent liver tumor promoter in rats, with females being more sensitive than males. The epidermal growth factor receptor (EGFR) pathway has been implicated in altered cell growth patterns induced by tumor promoters. We investigated hepatic EGFR levels in a two-stage initiation promotion model. The TCDD doses were chosen to encompass the dose range administered in a previous chronic bioassay currently used to determine the cancer potency commonly used for human health risk assessments. TCDD was administered biweekly by oral gavage to female Sprague-Dawley rats for 30 weeks following initiation by a single dose of diethylnitrosamine (DEN). TCDD-mediated decreased EGF receptor levels were demonstrated in intact but not ovariectomized animals, consistent with previous tumor data. Likewise, previous studies have shown that TCDD induces cell proliferation in intact rats but not ovariectomized rats. We report a significant dose-dependent decrease in plasma membrane EGF receptor maximum binding capacity in both initiated and non-initiated intact rats at TCDD doses equivalent to 3.5, 10.7, 35.7 and 125 ng/kg/day. There was a significant correlation between EGF receptor effects and liver TCDD concentration. The decrease in plasma membrane EGFR determined by equilibrium binding was confirmed quantitatively by EGF stimulation of EGFR autophosphorylation as well as qualitatively by immunohistochemical detection in control and treated rats. These results demonstrate that the observed down modulation of the EGFR by TCDD is ovarian-dependent and is a sensitive effect induced at dose levels associated with TCDD hepatocarcinogenicity in rodent bioassays.


Subject(s)
ErbB Receptors/metabolism , Liver/metabolism , Polychlorinated Dibenzodioxins/pharmacology , Analysis of Variance , Animals , Cell Division/drug effects , Cell Membrane/metabolism , Diethylnitrosamine , Dose-Response Relationship, Drug , Down-Regulation/drug effects , ErbB Receptors/analysis , ErbB Receptors/drug effects , Female , Liver/chemistry , Liver/cytology , Liver Neoplasms, Experimental/chemically induced , Ovariectomy , Phosphorylation , Polychlorinated Dibenzodioxins/toxicity , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...