Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Toxicol Sci ; 164(1): 218-228, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29596651

ABSTRACT

The effects of body fat mass on the elimination of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) was examined in mice. When male C57BL/6J mice are fed a high-fat, simple carbohydrate diet (HFD) for 13 weeks, they develop an obese phenotype. In contrast, A/J mice fed an HFD do not become obese. After 13 weeks on a normal diet (ND) or HFD, male C57BL/6J and A/J mice received a single dose by gavage of 0.1 or 5.0 µg of 2,3,7,8-tetrachloro[1,6-3H] dibenzo-p-dioxin per kg body weight. Using classical pharmacokinetics, the blood elimination half-life of TCDD was approximately 10 and 2 times longer in the C57BL/6J on the HFD compared with the mice on the ND at 0.1 and 5.0 µg/kg doses, respectively. The diet did not increase the blood half-life of TCDD in the A/J mice, which did not get obese. Using a physiologically based pharmacokinetic model for TCDD that incorporated experimentally derived percent body fat mass and tissue partition coefficients, as well as data on hepatic sequestration, did not provide accurate predictions to the data and could not explain the increase in half-life of TCDD in the HFD groups. This work demonstrates that obesity influences the half-life of TCDD, but other undetermined factors are involved in its elimination because the increase in body fat mass, decreases in cytochrome P4501A2, and altered partition coefficients could not completely explain the prolonged half-life.


Subject(s)
Models, Biological , Obesity/metabolism , Polychlorinated Dibenzodioxins/pharmacokinetics , Animals , Computer Simulation , Cytochrome P-450 CYP1A2/genetics , Diet, High-Fat/adverse effects , Gene Expression/drug effects , Half-Life , Inactivation, Metabolic , Liver/drug effects , Liver/metabolism , Lung/drug effects , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Polychlorinated Dibenzodioxins/blood , Species Specificity , Tissue Distribution
2.
Toxicol Sci ; 123(1): 256-63, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21705717

ABSTRACT

Hexabromocyclododecane (HBCD) is a mixture of three stereoisomers alpha (α), beta (ß), and gamma (γ). γ-HBCD dominates the mixture (∼70%), and despite α-HBCD's minor contribution to global HBCD production and usage (∼10%), it is the dominant congener found in most biotic samples worldwide. Evidence of toxicity and lack of stereoisomer studies drives the importance of understanding HBCD toxicokinetics in potentially susceptible populations. The majority of public health concern has focused on hazardous effects resulting from exposure of infants and young children to HBCD due to reports on adverse developmental effects in rodent studies, in combination with human exposure estimates suggesting that nursing infants and young children have the highest exposure to HBCD. This study was designed to investigate differences in the disposition of both γ-HBCD and α-HBCD in infantile mice reported to be susceptible to the HBCD commercial mixture. The tissue distribution of α-[(14)C]HBCD- and γ-[(14)C]HBCD-derived radioactivity was monitored in C57BL/6 mice following a single oral dose of either compound (3 mg/kg) after direct gavage at postnatal day 10. Mice were held up to 7 days in shoebox cages after which pups were sacrificed, tissue collected, and internal dosimetry was measured. Developing mice exposed to α-HBCD had an overall higher body burden than γ-HBCD at every time point measured; at 4 days postexposure, they retained 22% of the α-HBCD administered dose, whereas pups exposed to γ-HBCD retained 10%. Total body burden in infantile mice after exposure to γ-HBCD was increased 10-fold as compared with adults. Similarly, after exposure to α-HBCD, infantile mice contained 2.5-fold higher levels than adult. These differences lead to higher concentrations of the HBCD diastereomers at target tissues during critical windows of development. The results indicate that the toxicokinetics of the two HBCD diastereomers differ between developing and adult mice; whereas distribution patterns are similar, concentrations of each HBCD diastereomer's-derived radioactivity are higher in the pup's liver, fat, kidney, brain, blood, muscle, and lungs than in the adult's. This study suggests that developmental stage may be a risk factor for the harmful effects of α-HBCD and γ-HBCD, when developing animals may be more sensitive to effects and have increased body burden.


Subject(s)
Flame Retardants/pharmacokinetics , Hydrocarbons, Brominated/pharmacokinetics , Age Factors , Animals , Body Burden , Female , Flame Retardants/toxicity , Hydrocarbons, Brominated/toxicity , Male , Mice , Mice, Inbred C57BL , Stereoisomerism , Tissue Distribution
3.
Toxicol Sci ; 121(2): 234-44, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21441408

ABSTRACT

Alpha-hexabromocyclododecane (α-HBCD) is an emerging persistent organic pollutant present in the hexabromocyclododecane (HBCD) commercial mixture. HBCD is used as an additive flame retardant in a wide variety of household consumer products. Three main stereoisomers, alpha (α), beta (ß), and gamma (γ), comprise roughly 10, 10, and 80% of the mixture, respectively. Despite its small contribution to HBCD global production and usage, α-HBCD is the major stereoisomer found in wildlife and human tissues including breast milk and blood in North America, European Union, and Asia. No mammalian or human data are currently available regarding the toxicokinetics of α-HBCD. This study was conducted in an effort to fully characterize the absorption, distribution, metabolism, and elimination of α-HBCD following a single and repeated exposure with respect to dose, time, and route of administration in female C57BL/6 mice. Results indicate that ∼90% of the administered dose (3 mg/kg) was absorbed after oral exposure. Disposition was (1) dictated by lipophilicity, as adipose, liver, muscle, and skin were major depots and (2) was dose dependent with nonlinear accumulation at higher doses. Elimination, both whole-body and from individual tissues, was biphasic. α-HBCD-derived radioactivity was excreted in the feces as parent and metabolites, whereas urine only contained metabolites. Presence of polar metabolites in the blood and urine were a major factor in determining the rapid initial whole-body half-life after a single oral exposure. Initial half-lives were ∼1-3 days and much longer terminal half-lives of 17 days were observed, suggesting the potential for α-HBCD bioaccumulation. A 10-day repeated study supports α-HBCD bioaccumulation potential. Stereoisomerization previously observed after exposure to γ-HBCD was not seen after exposure of α-HBCD. The toxicokinetic behavior reported here has important implications for the extrapolation of toxicological studies of the commercial HBCD mixture to the assessment of risk of α-HBCD which is the major stereoisomer found in wildlife and people.


Subject(s)
Dose-Response Relationship, Drug , Flame Retardants/pharmacokinetics , Hydrocarbons, Brominated/pharmacokinetics , Administration, Oral , Animals , Biotransformation , Chromatography, Liquid , Chromatography, Thin Layer , Endocrine Disruptors/pharmacokinetics , Female , Injections, Intravenous , Mass Spectrometry , Mice , Mice, Inbred C57BL , Nonlinear Dynamics , Risk Assessment , Time Factors , Tissue Distribution
4.
Toxicol Sci ; 117(2): 282-93, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20562218

ABSTRACT

Hexabromocyclododecane-gamma (γ-HBCD) is the predominate diastereoisomer in the commercial HBCD mixture used as a flame retardant in a wide variety of consumer products. Three main diastereoisomers, alpha (α), beta (ß), and gamma (γ), comprise the mixture. Despite the γ-diastereoisomer being the major diastereoisomer in the mixture and environmental samples, the α-diastereoisomer predominates human tissue and wildlife. This study was conducted to characterize absorption, distribution, metabolism, and excretion parameters of γ-HBCD with respect to dose and time following a single acute exposure and repeated exposure in adult female C57BL/6 mice. Results suggest that 85% of the administered dose (3 mg/kg) was absorbed after po exposure. Disposition was dose independent and did not significantly change after 10 days of exposure. Liver was the major depot (< 0.3% of dose) 4 days after treatment followed by blood, fat, and then brain. γ-HBCD was rapidly metabolized and eliminated in the urine and feces. For the first time, in vivo stereoisomerization was observed of the γ-diastereoisomer to the ß-diastereoisomer in liver and brain tissues and to the α- and ß-diastereoisomer in fat and feces. Polar metabolites in the blood and urine were a major factor in determining the initial whole-body half-life (1 day) after a single po exposure. Elimination, both whole-body and from individual tissues, was biphasic. Initial half-lives were approximately 1 day, whereas terminal half-lives were up to 4 days, suggesting limited potential for γ-diastereoisomer bioaccumulation. The toxicokinetic behavior reported here has important implications for the extrapolation of toxicological studies of the commercial HBCD mixture to the assessment of risk.


Subject(s)
Flame Retardants/pharmacokinetics , Flame Retardants/toxicity , Hydrocarbons, Brominated/pharmacokinetics , Hydrocarbons, Brominated/toxicity , Administration, Oral , Animals , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Half-Life , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Stereoisomerism
5.
Toxicol Appl Pharmacol ; 241(1): 119-26, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19695277

ABSTRACT

Numerous metabolism studies have demonstrated that the toxic contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is poorly metabolized. A hallmark feature of TCDD exposure is induction of hepatic CYP1A2 and subsequent sequestration leading to high liver-to-fat concentration ratios. This study was initiated to determine whether TCDD was inherently poorly metabolized or unavailable for metabolism because of sequestration to CYP1A2. [(3)H]TCDD was administered as a single, oral dose (0.1 and 10 microg/kg) to 12 male C57BL/6N mice or 12 CYP1A2 (-/-) mice. At 96 h, less than 5% of the dose was eliminated in the urine of all groups, and TCDD detected in urine was bound to mouse major urinary protein (mMUP). Feces were the major elimination pathway (24-31% of dose), and fecal extracts and non-extractables were quantitated by HPLC for metabolites. No great differences in urinary or fecal elimination (% dose) were observed between the high and low dose treatments. TCDD concentrations were the highest in adipose tissue for CYP1A2 knockout mice but in liver for C57BL/6N mice supporting the role of hepatic CYP1A2 in the sequestration of TCDD. Overall metabolism between parental and knockout strains showed no statistical differences at either the high or low doses. The data suggested that metabolism of TCDD is inherently slow, due principally to CYP1A1, and that hepatic CYP1A2 is not an active participant in the metabolism of TCDD in male mice. Rather, CYP1A2 governs the pharmacokinetics of TCDD by making it unavailable for hepatic CYP1A1 through sequestration and attenuating extrahepatic tissue disposition.


Subject(s)
Cytochrome P-450 CYP1A2/metabolism , Environmental Pollutants/pharmacokinetics , Liver/metabolism , Polychlorinated Dibenzodioxins/pharmacokinetics , Adipose Tissue/metabolism , Administration, Oral , Animals , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP1A2/genetics , Dose-Response Relationship, Drug , Environmental Pollutants/administration & dosage , Feces/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Polychlorinated Dibenzodioxins/administration & dosage , Proteins/metabolism , Tissue Distribution
6.
Toxicol Sci ; 107(1): 27-39, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18978342

ABSTRACT

Previous studies demonstrated that perinatal exposure to polybrominated diphenyl ethers (PBDEs), a major class of brominated flame retardants, may affect thyroid hormone (TH) concentrations by inducing hepatic uridinediphosphate-glucoronosyltransferases (UGTs). This study further examines effects of the commercial penta mixture, DE-71, on genes related to TH metabolism at different developmental time points in male rats. DE-71 is predominately composed of PBDE congeners 47, 99, 100, 153, 154 with low levels of brominated dioxin and dibenzofuran contaminants. Pregnant Long-Evans rats were orally administered 1.7 (low), 10.2 (mid), or 30.6 (high) mg/kg/day of DE-71 in corn oil from gestational day (GD) 6 to postnatal day (PND) 21. Serum and liver were collected from male pups at PND 4, 21, and 60. Total serum thyroxine (T(4)) decreased to 57% (mid) and 51% (high) on PND 4, and 46% (mid) dose and 25% (high) on PND 21. Cyp1a1, Cyp2b1/2, and Cyp3a1 enzyme and mRNA expression, regulated by aryl hydrocarbon receptor, constitutive androstane receptor, and pregnane xenobiotic receptor, respectively, increased in a dose-dependent manner. UGT-T(4) enzymatic activity significantly increased, whereas age and dose-dependent effects were observed for Ugt1a6, 1a7, and 2b mRNA. Sult1b1 mRNA expression increased, whereas that of transthyretin (Ttr) decreased as did both the deiodinase I (D1) enzyme activity and mRNA expression. Hepatic efflux transporters Mdr1 (multidrug resistance), Mrp2 (multidrug resistance-associated protein), and Mrp3 and influx transporter Oatp1a4 mRNA expression increased. In this study the most sensitive responses to PBDEs following DE-71 exposure were CYP2B and D1 activities and Cyb2b1/2, d1, Mdr1, Mrp2, and Mrp3 gene expression. All responses were reversible by PND 60. In conclusion, deiodination, active transport, and sulfation, in addition to glucuronidation, may be involved in disruption of TH homeostasis due to perinatal exposure to DE-71 in male rat offspring.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Gene Expression Regulation, Developmental/drug effects , Halogenated Diphenyl Ethers/toxicity , Thyroxine/analysis , Analysis of Variance , Animals , Animals, Newborn , Aryl Hydrocarbon Hydroxylases/genetics , Female , Flame Retardants/administration & dosage , Flame Retardants/toxicity , Gene Expression Regulation/drug effects , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Halogenated Diphenyl Ethers/administration & dosage , Male , Microsomes, Liver/chemistry , Pregnancy , Rats , Rats, Long-Evans , Sulfotransferases/genetics , Sulfotransferases/metabolism , Thyroxine/blood , Triiodothyronine/analysis , Triiodothyronine/blood
7.
Environ Sci Technol ; 42(7): 2694-700, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18505018

ABSTRACT

Household dust has been implicated as a major source of polybrominated diphenyl ether (PBDE) exposure in humans. This finding has important implications for young children, who tend to ingest more dust than adults and may be more susceptible to some of the putative developmental effects of PBDEs. Absorption parameters of PBDEs from ingested dust are unknown; therefore, the objectives of this study were to determine and to compare the uptake of PBDEs from either household dust (NIST Standard Reference Material 2585) or a corn oil solution. Male rats were administered dust or corn oil doses at 1 or 6 microg of PBDEs kg(-1) body wt in the diet for 21 days (n = 4 rats per group). The concentrations of 15 PBDEs were measured in adipose tissue and liver from each treatment group and showed that bioconcentration was congener dependent, but for the majority of congeners, the concentrations did not differ with either dose level or dose vehicle. Hepatic Cyp2b1 and 2b2 mRNA expression increased in rats receiving the higher PBDE doses, suggesting potential effects on metabolic activity. Retention of PBDEs in tissues ranged from <5% of the dose for BDE-209 to 70% for BDEs-47, 100, and 153 but generally did not differ between the high dust and high oil treatment groups. Excretion via the feces was significantly lower in the high oil dosed rats suggesting differences in absorption, excretion, and/or metabolism. The present study shows that PBDEs in dust are readily bioavailable and are biologically active, as indicated by increased transcription of hepatic enzymes.


Subject(s)
Dust , Oils , Polybrominated Biphenyls/metabolism , Animals , Ethers , Male , Polybrominated Biphenyls/pharmacokinetics , Rats , Rats, Sprague-Dawley
8.
Toxicol Appl Pharmacol ; 226(3): 244-50, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17964624

ABSTRACT

Polybrominated diphenyl ethers (PBDEs) are a class of polyhalogenated aromatic compounds commercially used as fire retardants in consumer products. These compounds have been shown to decrease thyroid hormone concentrations in rodents after acute exposures. This study examines the ability of 2,2',4,4'-tetrabromodiphenyl ether (BDE 47) to decrease circulating thyroid hormone concentrations and pairs this with BDE 47-induced effects on genes involved in thyroid hormone homeostasis. Female C57BL/6 mice (9 weeks old) were orally administered 3, 10, or 100 mg/kg/day of BDE 47 for 4 days. Animals were euthanized 24 h after the final dose (day 5) and liver, kidney, and serum were collected for analysis. BDE 47 caused a significant 43% decrease at 100 mg/kg/day in serum total thyroxine (T(4)) concentrations. There was no increase in hepatic T(4)-glucuronidation activity, but significant increases in hepatic Ugt1a1, Ugt1a7, and Ugt2b5 mRNA expression accompany significant decreases in T(4) concentrations at 100 mg/kg/day of BDE 47. Induction of PROD activity occurred at the lowest dose (3 mg/kg/day). Cyp2b10 mRNA expression also increased significantly at 10 and 100 mg/kg/day. These key findings show that BDE activates the nuclear receptor, CAR. Decreases in Mdr1a mRNA expression also occurred at the lowest dose administered (3 mg/kg/day BDE 47). BDE 47 exposure also decreased hepatic transthyretin and monocarboxylate transporter 8 (Mct8) mRNA expression, suggesting that while induction of UGTs may be partly responsible for T(4) decreases, other mechanisms are also involved. No effects were seen in the kidney. We conclude that changes in hepatic UGTs and transporters may be involved in decreases in circulating T(4) following BDE 47 exposure.


Subject(s)
Endocrine Disruptors/toxicity , Environmental Pollutants/toxicity , Flame Retardants/toxicity , Polybrominated Biphenyls/toxicity , Thyroid Gland/drug effects , Thyroxine/blood , ATP Binding Cassette Transporter, Subfamily B/biosynthesis , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Animals , Aryl Hydrocarbon Hydroxylases/biosynthesis , Aryl Hydrocarbon Hydroxylases/genetics , Cytochrome P-450 CYP2B1/biosynthesis , Cytochrome P-450 CYP2B1/genetics , Cytochrome P450 Family 2 , Dose-Response Relationship, Drug , Enzyme Induction , Female , Gene Expression Regulation, Enzymologic/drug effects , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Halogenated Diphenyl Ethers , Liver/drug effects , Liver/enzymology , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Monocarboxylic Acid Transporters , Prealbumin/genetics , Prealbumin/metabolism , Protein Kinases/biosynthesis , Protein Kinases/genetics , RNA, Messenger/metabolism , Steroid Hydroxylases/biosynthesis , Steroid Hydroxylases/genetics , Symporters , Thyroid Gland/metabolism
9.
Toxicol Sci ; 94(1): 28-37, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16936226

ABSTRACT

The congener profiles of polybrominated diphenyl ethers (PBDEs) in human and wildlife samples are dominated by brominated diphenyl ether (BDE) congeners 47, 99, 100, 153, and 154, all of which are components of the commercial pentaBDE mixtures commonly used in a variety of flammable consumer products. Very little information is available on the toxicokinetics of these congeners and no studies are available directly comparing these BDE congeners in mice. Therefore, the objective of this study was to compare the distribution, metabolism, and excretion of BDEs 47, 99, 100 and 153. Female C57BL/6 mice were administered a single dose of BDE (1 mg/kg: 2.1, 1.9, 1.9, and 1.8 mumol/kg, respectively) intravenously. Excretion was monitored daily, and terminal tissue disposition was examined 5 days following exposure. All BDE congeners in this study distribute with similar patterns into lipophilic tissues; however, tissue concentrations 5 days following exposure were much higher for BDE-153 than for 100, 99, and 47, respectively. Excretion rates were inversely related to tissue concentrations as BDE-47 was the most rapidly excreted congener, followed by BDE-99, -100, and -153. Differences in tissue concentrations were largely driven by congener-specific urinary elimination rates which were associated with protein binding in the urine. While the overall rate of metabolism appeared to be low, analysis of metabolites in daily feces samples revealed that BDE-99 was the most rapidly metabolized congener in this study. The results of this study demonstrate that congener substitution plays a role in the distribution, metabolism, and excretion of PBDEs in mice and it is therefore important to consider the differential toxicokinetic parameters associated with each congener when assessing the risk to human health from these PBDE congeners.


Subject(s)
Phenyl Ethers/pharmacokinetics , Polybrominated Biphenyls/pharmacokinetics , Animals , Carbon Radioisotopes , Dose-Response Relationship, Drug , Feces/chemistry , Female , Halogenated Diphenyl Ethers , Injections, Intravenous , Mice , Mice, Inbred C57BL , Molecular Structure , Phenyl Ethers/chemistry , Phenyl Ethers/toxicity , Polybrominated Biphenyls/chemistry , Polybrominated Biphenyls/toxicity , Structure-Activity Relationship , Time Factors , Tissue Distribution , Urine/chemistry
10.
Toxicol Sci ; 90(2): 309-16, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16407092

ABSTRACT

Despite its minor contribution to global polybrominated diphenyl ether (PBDE) production and usage, 2,2',4,4'-tetrabromodiphenyl ether (BDE 47) is the dominant congener found in most biotic samples in North America. The majority of public health concern has focused on potential hazardous effects resulting from exposure of infants and young children to BDE 47 because of previous studies reporting adverse developmental effects in rodent studies, in combination with human exposure estimates suggesting that nursing infants and young children have the highest exposure to BDE 47. This study was designed with two objectives: (1) to investigate the disposition of BDE 47 in infantile mice reported to be susceptible to BDE 47 and (2) to investigate the disposition and excretion of BDE 47 at various developmental stages in an attempt to further identify the mechanism responsible for rapid urinary excretion. The disposition of (14)C-BDE 47 was monitored in C57BL/6 mice following a single oral dose of BDE 47 (1 mg/kg) at different stages of development. The results show that the toxicokinetics of BDE 47 are different in developing mice than in adult mice; whereas disposition patterns are similar, concentrations of BDE 47 are higher in pups because they have a reduced capacity to excrete BDE 47. These differences lead to higher concentrations of BDE 47 at target tissues during critical windows of development.


Subject(s)
Animals, Newborn/metabolism , Flame Retardants/pharmacokinetics , Hydrocarbons, Brominated/pharmacokinetics , Phenyl Ethers/pharmacokinetics , Animals , Environmental Pollutants/blood , Environmental Pollutants/pharmacokinetics , Environmental Pollutants/urine , Female , Halogenated Diphenyl Ethers , Hydrocarbons, Brominated/blood , Hydrocarbons, Brominated/urine , Male , Mice , Mice, Inbred C57BL , Phenyl Ethers/blood , Phenyl Ethers/urine , Polybrominated Biphenyls , Pregnancy , Tissue Distribution
11.
Environ Int ; 29(6): 855-60, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12850101

ABSTRACT

This article reviews the state of the science regarding the health effects of polybrominated dibenzo-p-dioxins (PBDDs) and dibenzofurans (PBDFs). While thousands of articles have been published on the health effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin and related polychlorinated dibenzodioxins (PCDDs) and dibenzofurans (PCDFs), little is know about the brominated and mixed chloro/bromo homologs. Available literature suggests that brominated compounds have similar toxicity profiles to their chlorinated homologs. However, further research investigating health effects will only be useful if exposure scenarios truly exist. Current exposure data is extremely limited, posing a major data gap in assessing potential risk of these chemicals. The rapid increase in the use of brominated flame retardants has raised the level of environmental concern regarding PBDDs/PBDFs as it is likely that human, as well as wildlife, exposure to brominated dioxins and furans will increase with their use.


Subject(s)
Benzofurans/toxicity , Dioxins/toxicity , Hydrocarbons, Brominated/chemistry , Hydrocarbons, Brominated/toxicity , Environmental Exposure , Public Health
SELECTION OF CITATIONS
SEARCH DETAIL
...