Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Regul Toxicol Pharmacol ; 149: 105598, 2024 May.
Article in English | MEDLINE | ID: mdl-38548044

ABSTRACT

In 2022 the World Health Organization (WHO) published updated 'Toxic Equivalence Factors' (TEFs) for a wide variety of chlorinated dioxins, dibenzofurans and PCBs [collectively referred to as 'dioxin-like chemicals'; DLCs) that interact with the aryl hydrocarbon receptor (AHR)]. Their update used sophisticated statistical analysis of hundreds of published studies that reported estimation of 'Relative Effective Potency' (REP) values for individual DLC congeners. The weighting scheme used in their assessment of each study favored in vivo over in vitro studies and was based largely on rodent studies. In this Commentary, we highlight the large body of published studies that demonstrate large species differences in AHR-ligand activation and provide supporting evidence for our position that the WHO 2022 TEF values intended for use in human risk assessment of DLC mixtures will provide highly misleading overestimates of 'Toxic Equivalent Quotients' (TEQs), because of well-recognized striking differences in AHR ligand affinities between rodent (rat, mouse) and human. The data reviewed in our Commentary support the position that human tissue-derived estimates of REP/TEF values for individual DLC congeners, although uncertain, will provide much better, more realistic estimates of potential activation of the human AHR, when exposure to complex DLC mixtures occurs.


Subject(s)
Receptors, Aryl Hydrocarbon , Species Specificity , Receptors, Aryl Hydrocarbon/metabolism , Animals , Humans , Ligands , Risk Assessment , Dioxins/toxicity , Polychlorinated Biphenyls/toxicity , Rats , Mice
3.
J Nutr ; 144(11): 1850-7, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25165394

ABSTRACT

BACKGROUND: Isothiocyanates in cruciferous vegetables modulate signaling pathways critical to carcinogenesis, including nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), a central regulator of inflammation. Glutathione S-transferase (GST) M1 and GSTT1 metabolize isothiocyanates; genetic variants may result in differences in biologic response. OBJECTIVE: The objective of this study was to test whether consumption of cruciferous or cruciferous plus apiaceous vegetables altered serum concentrations of interleukin (IL)-6, IL-8, C-reactive protein (CRP), tumor necrosis factor (TNF) α, and soluble TNF receptor (sTNFR) I and II, and whether this response was GSTM1/GSTT1 genotype dependent. METHODS: In a randomized crossover trial, healthy men (n = 32) and women (n = 31) aged 20-40 y consumed 4 14-d controlled diets: basal (vegetable-free), single-dose cruciferous (1xC) [7 g vegetables/kg body weight (BW)], double-dose cruciferous (2xC) (14 g/kg BW), and cruciferous plus apiaceous (carrot family) (1xC+A) vegetables (7 and 4 g/kg BW, respectively), with a 21-d washout period between each intervention. Urinary isothiocyanate excretion was also evaluated as a marker of systemic isothiocyanate exposure. Fasting morning blood and urine samples were collected on days 0 and 14 and analyzed. RESULTS: IL-6 concentrations were significantly lower on day 14 of the 2xC and 1xC+A diets than with the basal diet [-19% (95% CI: -30%, -0.1%) and -20% (95% CI: -31%, -0.7%), respectively]. IL-8 concentrations were higher after the 1xC+A diet (+16%; 95% CI: 4.2%, 35.2%) than after the basal diet. There were no effects of diet on CRP, TNF-α, or sTNFRI or II. There were significant differences between GSTM1-null/GSTT1+ individuals for several biomarkers in response to 1xC+A compared with basal diets (CRP: -37.8%; 95% CI: -58.0%, -7.4%; IL-6: -48.6%; 95% CI: -49.6%, -12.0%; IL-8: 16.3%; 95% CI: 6.7%, 57.7%) and with the 2xC diet compared with the basal diet (IL-8: -33.2%; 95% CI: -43.0%, -1.4%; sTNFRI: -7.5%; 95% CI: -12.7%, -2.3%). There were no significant reductions in biomarker concentrations in response to diet among GSTM1+/GSTT1+ or GSTM1-null/GSTT1-null individuals. Twenty-four-hour urinary isothiocyanate excretion was not associated with any of the inflammation markers overall; however, IL-6 was inversely associated with total isothiocyanate excretion in GSTM1-null/GSTT1-null individuals (ß = -0.12; 95% CI: -0.19, -0.05). CONCLUSIONS: In this young, healthy population, consumption of cruciferous and apiaceous vegetables reduced circulating IL-6; however, results for other biomarkers of inflammation were not consistent.


Subject(s)
Brassicaceae , Diet , Inflammation/metabolism , Vegetables , Adult , Biomarkers , Cross-Over Studies , Female , Gene Expression Regulation , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Inflammation/blood , Male , Young Adult
4.
Toxicol Appl Pharmacol ; 266(1): 122-31, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23153560

ABSTRACT

Sulforaphane (SFN), is an effective in vitro antagonist of ligand activation of the human pregnane and xenobiotic receptor (PXR). PXR mediated CYP3A4 up-regulation is implicated in adverse drug-drug interactions making identification of small molecule antagonists a desirable therapeutic goal. SFN is not an antagonist to mouse or rat PXR in vitro; thus, normal rodent species are not suitable as in vivo models for human response. To evaluate whether SFN can effectively antagonize ligand activation of human PXR in vivo, a three-armed, randomized, crossover trial was conducted with 24 healthy adults. The potent PXR ligand - rifampicin (300mg/d) was given alone for 7days in arm 1, or in daily combination with 450µmol SFN (Broccoli Sprout extract) in arm 2; SFN was given alone in arm 3. Midazolam as an in vivo phenotype marker of CYP3A was administered before and after each treatment arm. Rifampicin alone decreased midazolam AUC by 70%, indicative of the expected increase in CYP3A4 activity. Co-treatment with SFN did not reduce CYP3A4 induction. Treatment with SFN alone also did not affect CYP3A4 activity in the cohort as a whole, although in the subset with the highest basal CYP3A4 activity there was a statistically significant increase in midazolam AUC (i.e., decrease in CYP3A4 activity). A parallel study in humanized PXR mice yielded similar results. The parallel effects of SFN between humanized PXR mice and human subjects demonstrate the predictive value of humanized mouse models in situations where species differences in ligand-receptor interactions preclude the use of a native mouse model for studying human ligand-receptor pharmacology.


Subject(s)
Brassica , Plant Extracts/pharmacology , Receptors, Steroid/antagonists & inhibitors , Receptors, Steroid/metabolism , Thiocyanates/pharmacology , Adult , Animals , Cross-Over Studies , Female , Humans , Isothiocyanates , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plant Extracts/isolation & purification , Pregnane X Receptor , Sulfoxides , Thiocyanates/isolation & purification , Treatment Outcome , Young Adult
5.
Am J Pathol ; 175(1): 54-65, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19498008

ABSTRACT

Parkinson disease (PD) typically affects the cortical regions during the later stages of disease, with neuronal loss, gliosis, and formation of diffuse cortical Lewy bodies in a significant portion of patients with dementia. To identify novel proteins involved in PD progression, we prepared synaptosomal fractions from the frontal cortices of pathologically verified PD patients at different stages along with age-matched controls. Protein expression profiles were compared using a robust quantitative proteomic technique. Approximately 100 proteins displayed significant differences in their relative abundances between PD patients at various stages and controls; three of these proteins were validated using independent techniques. One of the confirmed proteins, glutathione S-transferase Pi, was further investigated in cellular models of PD, demonstrating that its level was intimately associated with several critical cellular processes that are directly related to neurodegeneration in PD. These results have, for the first time, suggested that the levels of glutathione S-transferase Pi may play an important role in modulating the progression of PD.


Subject(s)
Brain Chemistry , Glutathione S-Transferase pi/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology , Animals , Blotting, Western , Disease Progression , Female , Humans , Immunohistochemistry , MAP Kinase Kinase 4/metabolism , Male , Mice , Polymerase Chain Reaction , Signal Transduction/physiology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transfection
6.
Neurotoxicol Teratol ; 81: 106916, 2020.
Article in English | MEDLINE | ID: mdl-32698050

ABSTRACT

Response variability across the lifespan is an important consideration in toxicology and risk assessment, and the toxic effects of drugs and chemicals during adolescence need more research. This paper summarizes a workshop presented in March 2019, at the Society of Toxicology Annual Meeting in Baltimore, Maryland, that brought together experts in research on drug dependence and toxicity related to nicotine, cannabis, cocaine, and other illicit drugs during adolescence. The goal of the workshop was to address the following issues: (1) Do the effects of adolescent exposure differ from the same exposure in adults? (2) Are there unique biological markers of adolescent brain development? If so, what are they and how reliable are they? (3) Since multiple factors influence substance use disorder, can we disentangle risk factors for abuse and/or toxicity? What are the underlying biological susceptibilities that lead to dependence and neurotoxicity? What are the social, psychosocial and environmental factors that contribute to abuse susceptibilities? This paper reviews drug policy and national trends in adolescent substance use; the public health consequences of e-cigarettes; rat models of adolescent-onset nicotine self-administration and persisting effects of gestational nicotine; sex-dependent effects of delta-9-tetrahydrocannabinol on adolescent brain-behavior relationships; and translational approaches for identifying adolescent risk factors for transition to drug dependence. There is strong evidence that drug exposure prior to adulthood has longer lasting effects on behavior and the underlying neural circuitry. These effects, which are sex-dependent and influenced by stress, may be candidates as predictors of adolescent vulnerability. A major challenge to determining if adolescents have a unique susceptibility to dependence is whether and to what extent the human data allow distinction between the increased risk due to biological immaturity, an underlying biological susceptibility to dependence, or psychosocial and environmental factors for substance dependence. Factors important to consider for development of animal models include the timing and pattern of exposure as it relates to adolescence; age of assessment, and direct comparison with similar effects following exposures to adults to demonstrate that these effects are unique to adolescence. Here we provide a roadmap for further research into what makes adolescent brain development unique.


Subject(s)
Adolescent Behavior/drug effects , Biological Factors/pharmacology , Brain/drug effects , Nicotine/pharmacology , Adolescent , Animals , Electronic Nicotine Delivery Systems , Humans , Substance-Related Disorders/metabolism
7.
Cancer Epidemiol Biomarkers Prev ; 17(8): 2012-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18708392

ABSTRACT

Testicular germ cell carcinoma (TGCC) is the most common malignancy among men ages 20 to 34 years. Although the pathogenesis of TGCC is poorly understood, suboptimal androgen levels or impaired androgen signaling may play a role. Some persistent organochlorine pesticides commonly found in human tissue possess antiandrogenic properties. We examined whether the risk of TGCC is associated with serum levels of 11 organochlorine pesticides, including p,p'-DDE, and whether the p,p'-DDE-TGCC association is modified by CAG or GGN repeat polymorphisms in the androgen receptor gene. We conducted a population-based case-control study among 18- to 44-year-old male residents of three Washington State counties. Cases (n = 246) were diagnosed during 1999 to 2003 with a first, primary TGCC. Controls (n = 630) were men of similar age with no history of TGCC from the same population identified through random-digit telephone dialing. Questionnaires elicited information on demographic, medical, and lifestyle factors. A blood specimen provided serum for gas chromatography-high-resolution mass spectrometry analysis of organochlorine pesticide residues and DNA for genotyping. We observed no clear patterns between TGCC risk and concentrations of any of the organochlorines measured, nor did we observe that the risk associated with p,p'-DDE was modified by androgen receptor CAG (<23 versus > or =23 repeats) or GGN (<17 versus > or =17 repeats) genotype. This study does not provide support for the hypothesis that adult exposure to organochlorine pesticides is associated with risk of TGCC. Due to uncertainty regarding how well organochlorine levels measured in adulthood reflect exposures during early life, further research is needed using exposure measurements collected in utero or during infancy.


Subject(s)
Hydrocarbons, Chlorinated/toxicity , Neoplasms, Germ Cell and Embryonal/chemically induced , Pesticides/toxicity , Testicular Neoplasms/chemically induced , Adolescent , Adult , Case-Control Studies , Environmental Exposure/adverse effects , Genotype , Humans , Hydrocarbons, Chlorinated/blood , Interviews as Topic , Male , Microsatellite Repeats , Neoplasms, Germ Cell and Embryonal/blood , Neoplasms, Germ Cell and Embryonal/epidemiology , Neoplasms, Germ Cell and Embryonal/genetics , Pesticides/blood , Regression Analysis , Risk , Surveys and Questionnaires , Testicular Neoplasms/blood , Testicular Neoplasms/epidemiology , Testicular Neoplasms/genetics , Washington/epidemiology
8.
Crit Rev Toxicol ; 38 Suppl 2: 1-125, 2008.
Article in English | MEDLINE | ID: mdl-18726789

ABSTRACT

This review examines the large body of toxicological and epidemiological information on human exposures to chlorpyrifos, with an emphasis on the controversial potential for chlorpyrifos to induce neurodevelopmental effects at low doses. The results of this review demonstrate that the use of urinary 3,5,6-trichlorpyridinol (TCPy), a metabolite of chlorpyrifos as a biomarker of nonoccupational exposure is problematic and may overestimate nonoccupational exposures to chlorpyrifos by 10-to 20-fold because of the widespread presence of both TCPy and chlorpyrifos-methyl in the food supply. Current "background" (nonoccupational) levels of exposure to chlorpyrifos are several orders of magnitude lower than those required to inhibit plasma cholinesterase activity, which is a more sensitive target than nervous system cholinesterase. However, several in vitro studies have identified putative neurodevelopmental mechanisms that are altered at concentrations of chlorpyrifos below those that inhibit cholinesterases. Although one human cohort study reported an association between maternal and cord blood chlorpyrifos levels and several measures of neurodevelopment, two other cohort studies that utilized urinary TCPy as a surrogate for chlorpyrifos exposure did not demonstrate an association. Although the weight of the scientific evidence demonstrates that current levels of chlorpyrifos exposure will not have any adverse effects on neurodevelopment that might result from inhibition of nervous system cholinesterases, several recent studies propose alternative mechanisms. Thus, further in vivo investigation on neurodevelopment in an appropriate animal model is needed; additional epidemiological studies may be warranted if a suitable, chlorpyrifos-exposed cohort can be identified and more rigorous measures of exposure are utilized.


Subject(s)
Chlorpyrifos/toxicity , Cholinesterase Inhibitors/toxicity , Environmental Exposure/adverse effects , Environmental Pollutants/toxicity , Insecticides/toxicity , Nervous System/drug effects , Age Factors , Animals , Chlorpyrifos/pharmacokinetics , Chlorpyrifos/pharmacology , Cholinesterase Inhibitors/pharmacokinetics , Cholinesterase Inhibitors/pharmacology , Environmental Pollutants/pharmacokinetics , Environmental Pollutants/pharmacology , Humans , Insecticides/pharmacokinetics , Insecticides/pharmacology , Nervous System/embryology , Nervous System/growth & development
9.
Mol Cell Biol ; 25(14): 5823-33, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15988000

ABSTRACT

Aflatoxin B1 (AFB1) is a human hepatotoxin and hepatocarcinogen produced by the mold Aspergillus flavus. In humans, AFB1 is primarily bioactivated by cytochrome P450 1A2 (CYP1A2) and 3A4 to a genotoxic epoxide that forms N7-guanine DNA adducts. A series of yeast haploid mutants defective in DNA repair and cell cycle checkpoints were transformed with human CYP1A2 to investigate how these DNA adducts are repaired. Cell survival and mutagenesis following aflatoxin B1 treatment was assayed in strains defective in nucleotide excision repair (NER) (rad14), postreplication repair (PRR) (rad6, rad18, mms2, and rad5), homologous recombinational repair (HRR) (rad51 and rad54), base excision repair (BER) (apn1 apn2), nonhomologous end-joining (NHEJ) (yku70), mismatch repair (MMR) (pms1), translesion synthesis (TLS) (rev3), and checkpoints (mec1-1, mec1-1 rad53, rad9, and rad17). Together our data suggest the involvement of homologous recombination and nucleotide excision repair, postreplication repair, and checkpoints in the repair and/or tolerance of AFB1-induced DNA damage in the yeast model. Rev3 appears to mediate AFB1-induced mutagenesis when error-free pathways are compromised. The results further suggest unique roles for Rad5 and abasic endonuclease-dependent DNA intermediates in regulating AFB1-induced mutagenicity.


Subject(s)
Aflatoxin B1/toxicity , Cytochrome P-450 Enzyme System/metabolism , DNA Damage , DNA Repair/physiology , Saccharomyces cerevisiae/genetics , Aflatoxin B1/metabolism , Base Pair Mismatch/genetics , Carcinogens/metabolism , Carcinogens/toxicity , Cytochrome P-450 Enzyme System/genetics , DNA Repair/genetics , DNA Replication/genetics , Humans , Mutation , Recombination, Genetic/genetics , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology
10.
Curr Protoc Toxicol ; 73: 14.18.1-14.18.28, 2017 Aug 04.
Article in English | MEDLINE | ID: mdl-28777442

ABSTRACT

The liver and the kidney are key toxicity target organs during drug development campaigns, as they typically carry the burden of drug transport and metabolism. Primary hepatocytes and proximal tubule epithelial cells grown in traditional in vitro 2-D culture systems do not maintain transporter and metabolic functions, thus limiting their utility for nonclinical toxicology investigations. We have developed a renal and hepatic microphysiological system (MPS) platform that uses a commercially available MPS device as the core cell culture platform for our methodologies. We describe protocols for isolating and propagating human proximal epithelial cells and how to seed and culture a renal MPS to recapitulate the human proximal tubule. We present two methods to culture hepatocytes within an MPS and the steps required to connect a renal MPS to a liver MPS. © 2017 by John Wiley & Sons, Inc.


Subject(s)
Kidney/drug effects , Liver/drug effects , Toxicity Tests , Cell Culture Techniques , Humans
11.
Toxicol In Vitro ; 40: 170-183, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28089783

ABSTRACT

The liver is the main site for drug and xenobiotics metabolism, including inactivation or bioactivation. In order to improve the predictability of drug safety and efficacy in clinical development, and to facilitate the evaluation of the potential human health effects from exposure to environmental contaminants, there is a critical need to accurately model human organ systems such as the liver in vitro. We are developing a microphysiological system (MPS) based on a new commercial microfluidic platform (Nortis, Inc.) that can utilize primary liver cells from multiple species (e.g., rat and human). Compared to conventional monolayer cell culture, which typically survives for 5-7days or less, primary rat or human hepatocytes in an MPS exhibited higher viability and improved hepatic functions, such as albumin production, expression of hepatocyte marker HNF4α and canaliculi structure, for up to 14days. Additionally, induction of Cytochrome P450 (CYP) 1A and 3A4 in cryopreserved human hepatocytes was observed in the MPS. The acute cytotoxicity of the potent hepatotoxic and hepatocarcinogen, aflatoxin B1, was evaluated in human hepatocytes cultured in an MPS, demonstrating the utility of this model for acute hepatotoxicity assessment. These results indicate that MPS-cultured hepatocytes provide a promising approach for evaluating chemical toxicity in vitro.


Subject(s)
Cell Culture Techniques , Chemical and Drug Induced Liver Injury , Drug Evaluation, Preclinical/methods , Toxicity Tests/methods , Adult , Aflatoxin B1/toxicity , Animals , Cell Survival/drug effects , Cells, Cultured , Child , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP3A/metabolism , Female , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Male , Middle Aged , Rats , Young Adult
12.
JCI Insight ; 2(22)2017 11 16.
Article in English | MEDLINE | ID: mdl-29202460

ABSTRACT

Environmental exposures pose a significant threat to human health. However, it is often difficult to study toxicological mechanisms in human subjects due to ethical concerns. Plant-derived aristolochic acids are among the most potent nephrotoxins and carcinogens discovered to date, yet the mechanism of bioactivation in humans remains poorly understood. Microphysiological systems (organs-on-chips) provide an approach to examining the complex, species-specific toxicological effects of pharmaceutical and environmental chemicals using human cells. We microfluidically linked a kidney-on-a-chip with a liver-on-a-chip to determine the mechanisms of bioactivation and transport of aristolochic acid I (AA-I), an established nephrotoxin and human carcinogen. We demonstrate that human hepatocyte-specific metabolism of AA-I substantially increases its cytotoxicity toward human kidney proximal tubular epithelial cells, including formation of aristolactam adducts and release of kidney injury biomarkers. Hepatic biotransformation of AA-I to a nephrotoxic metabolite involves nitroreduction, followed by sulfate conjugation. Here, we identify, in a human tissue-based system, that the sulfate conjugate of the hepatic NQO1-generated aristolactam product of AA-I (AL-I-NOSO3) is the nephrotoxic form of AA-I. This conjugate can be transported out of liver via MRP membrane transporters and then actively transported into kidney tissue via one or more organic anionic membrane transporters. This integrated microphysiological system provides an ex vivo approach for investigating organ-organ interactions, whereby the metabolism of a drug or other xenobiotic by one tissue may influence its toxicity toward another, and represents an experimental approach for studying chemical toxicity related to environmental and other toxic exposures.


Subject(s)
Aristolochic Acids/toxicity , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Animals , Biomarkers , Biotransformation , Carcinogens/toxicity , Dicumarol/metabolism , Epithelial Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Kidney/injuries , Male , Multidrug Resistance-Associated Proteins/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Nephrons/drug effects , Nephrons/metabolism , Pathology, Molecular/instrumentation , Pathology, Molecular/methods , Rats , Rats, Sprague-Dawley , Renal Insufficiency, Chronic , Xenobiotics
13.
Mutat Res ; 593(1-2): 121-42, 2006 Jan 29.
Article in English | MEDLINE | ID: mdl-16122766

ABSTRACT

Aflatoxin B1 (AFB(1)) is a potent human hepatotoxin and hepatocarcinogen produced by the mold Aspergillus flavus. In human, AFB(1) is bioactivated by cytochrome P450 (CYP450) enzymes, primarily CYP1A2, to the genotoxic epoxide that forms N(7)-guanine DNA adducts. To characterize the transcriptional responses to genotoxic insults from AFB(1), a strain of Saccharomyces cerevisiae engineered to express human CYP1A2 was exposed to doses of AFB(1) that resulted in minimal lethality, but substantial genotoxicity. Flow cytometric analysis demonstrated a dose and time dependent S phase delay under the same treatment conditions, indicating a checkpoint response to DNA damage. Replicate cDNA microarray analyses of AFB(1) treated cells showed that about 200 genes were significantly affected by the exposure. The genes activated by AFB(1)-treatment included RAD51, DUN1 and other members of the DNA damage response signature reported in a previous study with methylmethane sulfonate and ionizing radiation [A.P. Gasch, M. Huang, S. Metzner, D. Botstein, S.J. Elledge, P.O. Brown, Genomic expression responses to DNA-damaging agents and the regulatory role of the yeast ATR homolog Mec1p, Mol. Biol. Cell 12 (2001) 2987-3003]. However, unlike previous studies using highly cytotoxic doses, environmental stress response genes [A.P. Gasch, P.T. Spellman, C.M. Kao, O. Carmel-Harel, M.B. Eisen, G. Storz, D. Botstein, P.O. Brown, Genomic expression programs in the response of yeast cells to environmental changes, Mol. Biol. Cell 11 (2000) 4241-4257] were largely unaffected by our dosing regimen. About half of the transcripts affected are also known to be cell cycle regulated. The most strongly repressed transcripts were those encoding the histone genes and a group of genes that are cell cycle regulated and peak in M phase and early G1. These include most of the known daughter-specific genes. The rapid and coordinated repression of histones and M/G1-specific transcripts cannot be explained by cell cycle arrest, and suggested that there are additional signaling pathways that directly repress these genes in cells under genotoxic stress.


Subject(s)
Aflatoxin B1/toxicity , Cytochrome P-450 CYP1A2/genetics , DNA, Complementary/genetics , Mutagens/toxicity , Oligonucleotide Array Sequence Analysis , Cell Cycle/drug effects , DNA Repair , DNA Replication , Humans , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/drug effects
14.
Food Chem Toxicol ; 44(9): 1474-84, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16762476

ABSTRACT

In humans, apiaceous vegetables (carrots, parsnips, celery, parsley, etc.) inhibit cytochrome P-450 1A2, a biotransformation enzyme known to activate several procarcinogens, including aflatoxin B1 (AFB). We evaluated eight phytochemicals from apiaceous vegetables for effects on human cytochrome P-450 1A2 (hCYP1A2) activity using a methoxyresorufin O-demethylase (MROD) assay and a trp-recombination assay. Saccharomyces cerevisiae was used for heterologous CYP1A2 expression and this yeast strain is also diploid and auxotrophic for tryptophan due to mutations in the trp5 alleles. When these two alleles undergo AFB-induced mitotic recombination, gene conversion occurs, allowing yeast to grow in the absence of tryptophan. The apiaceous constituents psoralen, 5-methoxypsoralen (5-MOP), 8-methoxypsoralen (8-MOP), and apigenin were potent inhibitors of hCYP1A2-mediated MROD activity in yeast microsomes, whereas quercetin was a modest hCYP1A2 inhibitor. Naringenin, caffeic acid, and chlorogenic acid did not inhibit hCYP1A2-mediated MROD activity. The 2-h pretreatment of intact yeast cells with psoralen, 5-MOP, and 8-MOP significantly improved cell survival after subsequent 4-h AFB treatment and reduced hCYP1A2-mediated mutagenicity of AFB. Apigenin also significantly decreased mutagenicity. These results suggest that in vivo CYP1A2 inhibition by apiaceous vegetables may be due to the phytochemicals present and imply that apiaceous vegetable intake may be chemopreventive by inhibiting CYP1A2-mediated carcinogen activation.


Subject(s)
Aflatoxin B1/pharmacokinetics , Apiaceae , Cytochrome P-450 CYP1A2 Inhibitors , Enzyme Inhibitors/pharmacology , Mutagens/pharmacokinetics , Plant Extracts/pharmacology , Poisons/pharmacokinetics , Aflatoxin B1/toxicity , Apiaceae/chemistry , Biotransformation , Cell Survival/drug effects , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/chemistry , Flavonoids/pharmacology , Furocoumarins/pharmacology , Humans , Inhibitory Concentration 50 , Microsomes/drug effects , Microsomes/enzymology , Mutagens/toxicity , Oxidoreductases/metabolism , Plant Extracts/chemistry , Poisons/toxicity , Recombination, Genetic , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics
15.
Brain Pathol ; 26(1): 75-81, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26095919

ABSTRACT

Mortalin, an essential mitochondrial chaperone protein, has previously been implicated in the pathogenesis of a wide array of diseases, including neurodegenerative conditions such as Parkinson's disease (PD) and Alzheimer's disease. Previous reports have consistently described mortalin protein levels to be lower in the brain tissue of patients with neurodegenerative disease, with expression demonstrated to be lower in neurons of post-mortem PD brain specimens. However, to date, mortalin expression has not yet been evaluated in astrocytes of post-mortem brain tissue from either normal or PD subjects. Mortalin expression was demonstrated in mouse primary astrocyte cultures by Western blot and quantitative polymerase chain reaction (PCR). Furthermore, confocal microscopy studies in human post-mortem tissue indicated co-localization of mortalin within astrocytes. Utilizing a quantitative immunofluorescence staining approach, the protein was found to be moderately reduced (∼35%) in this cell type in the substantia nigra pars compacta, but not structures of the corpus striatum, in PD subjects as compared to age-/gender-matched controls. These findings highlight the potential contribution of disrupted astroglial function in the pathogenesis of PD.


Subject(s)
Astrocytes/metabolism , HSP70 Heat-Shock Proteins/metabolism , Mesencephalon/metabolism , Mesencephalon/pathology , Parkinson Disease/pathology , Aged , Aged, 80 and over , Animals , Animals, Newborn , Case-Control Studies , Cells, Cultured , Female , Glial Fibrillary Acidic Protein/metabolism , HSP70 Heat-Shock Proteins/genetics , Humans , Male , Mice , Middle Aged , RNA, Messenger/metabolism
16.
Environ Health Perspect ; 123(10): 909-18, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25978799

ABSTRACT

BACKGROUND: The Superfund Research Program (SRP) is an academically based, multidisciplinary, translational research program that for 25 years has sought scientific solutions to health and environmental problems associated with hazardous waste sites. SRP is coordinated by the National Institute of Environmental Health Sciences (NIEHS). It supports multi-project grants, undergraduate and postdoctoral training programs, individual research grants, and Small Business Innovation Research (SBIR) and Technology Transfer Research (STTR) grants. RESULTS: SRP has had many successes: discovery of arsenic's toxicity to the developing human central nervous system; documentation of benzene toxicity to hematologic progenitor cells in human bone marrow; development of novel analytic techniques such as the luciferase expression assay and laser fragmentation fluorescence spectroscopy; demonstration that PCBs can cause developmental neurotoxicity at low levels and alter the genomic characteristics of sentinel animals; elucidation of the neurodevelopmental toxicity of organophosphate insecticides; documentation of links between antimicrobial agents and alterations in hormone response; discovery of biological mechanisms through which environmental chemicals may contribute to obesity, atherosclerosis, diabetes, and cancer; tracking the health and environmental effects of the attacks on the World Trade Center and Hurricane Katrina; and development of novel biological and engineering techniques to facilitate more efficient and lower-cost remediation of hazardous waste sites. CONCLUSION: SRP must continue to address the legacy of hazardous waste in the United States, respond to new issues caused by rapid advances in technology, and train the next generation of leaders in environmental health science while recognizing that most of the world's worst toxic hot spots are now located in low- and middle-income countries.


Subject(s)
Environmental Health/history , Hazardous Waste Sites/history , Hazardous Waste/analysis , National Institute of Environmental Health Sciences (U.S.)/history , Public Health/history , History, 20th Century , History, 21st Century , Humans , Translational Research, Biomedical , United States
17.
Toxicol Sci ; 143(2): 256-67, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25628403

ABSTRACT

FutureTox II, a Society of Toxicology Contemporary Concepts in Toxicology workshop, was held in January, 2014. The meeting goals were to review and discuss the state of the science in toxicology in the context of implementing the NRC 21st century vision of predicting in vivo responses from in vitro and in silico data, and to define the goals for the future. Presentations and discussions were held on priority concerns such as predicting and modeling of metabolism, cell growth and differentiation, effects on sensitive subpopulations, and integrating data into risk assessment. Emerging trends in technologies such as stem cell-derived human cells, 3D organotypic culture models, mathematical modeling of cellular processes and morphogenesis, adverse outcome pathway development, and high-content imaging of in vivo systems were discussed. Although advances in moving towards an in vitro/in silico based risk assessment paradigm were apparent, knowledge gaps in these areas and limitations of technologies were identified. Specific recommendations were made for future directions and research needs in the areas of hepatotoxicity, cancer prediction, developmental toxicity, and regulatory toxicology.


Subject(s)
Computer Simulation , In Vitro Techniques , Toxicology/methods , Toxicology/trends , Congresses as Topic , Predictive Value of Tests , Societies, Scientific , United States
18.
Nanotoxicology ; 9(3): 336-43, 2015 May.
Article in English | MEDLINE | ID: mdl-24983898

ABSTRACT

Quantum dots (Qdots) are semiconductor nanoparticles with size-tunable fluorescence capabilities with diverse applications. Qdots typically contain cadmium or other heavy metals, hence raising concerns of their potential toxicity, especially in occupational settings where inhalation of nanomaterials may increase the risk of lung disease. Accordingly, we assessed the effects of tri-n-octylphosphine oxide, poly(maleic anhydride-alt-1-tetradecene) (TOPO-PMAT) coated CdSe/ZnS Qdots on mouse lung epithelial cells and macrophages. Mouse tracheal epithelial cells (MTEC), grown as organotypic cultures, bone marrow-derived macrophages (BMDM), and primary alveolar macrophages (AM) were derived from C57BL/6J or A/J mice and treated with TOPO-PMAT CdSe/ZnS Qdots (10-160 nM) for up to 24 h. Cadmium analysis showed that Qdots remained in the apical compartment of MTEC cultures, whereas they were avidly internalized by AM and BMDM, which did not differ between strains. In MTEC, Qdots selectively induced expression (mRNA and protein) of neutrophil chemokines CXCL1 and CXCL2 but only low to no detectable levels of other factors assessed. In contrast, 4 h exposure to Qdots markedly increased expression of CXCL1, IL6, IL12, and other pro-inflammatory factors in BMDM. Higher inflammatory response was seen in C57BL/6J than in A/J BMDM. Similar expression responses were observed in AM, although overall levels were less robust than in BMDM. MTEC from A/J mice were more sensitive to Qdot pro-inflammatory effects while macrophages from C57BL/6J mice were more sensitive. These findings suggest that patterns of Qdot-induced pulmonary inflammation are likely to be cell-type specific and genetic background dependent.


Subject(s)
Cadmium Compounds/toxicity , Cytokines/metabolism , Inflammation Mediators/metabolism , Lung/drug effects , Macrophages, Alveolar/drug effects , Quantum Dots , Selenium Compounds/toxicity , Sulfides/toxicity , Zinc Compounds/toxicity , Animals , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Lung/cytology , Lung/metabolism , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Inbred C57BL
19.
Cancer Epidemiol Biomarkers Prev ; 12(6): 527-33, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12814998

ABSTRACT

Glutathione S-transferase (GST) enzymes detoxify therapeutic drugs and reactive oxidants, so GST polymorphisms may influence survival after diagnosis of cancer. We evaluated survival according to GST polymorphisms in a population-based series of lung cancer patients. The study subjects (n = 274) were men diagnosed with lung cancer from 1993 through 1996 who participated in a case control study and provided a blood sample for genotyping. The presence of the GSTM1 and GSTT1 genes were assayed by multiplex PCR. Genotype at the GSTP1 Ile(105)Val substitution was determined by PCR and oligonucleotide ligation assay. The study subjects were followed for vital status through 2000, and overall survival was evaluated in Kaplan-Meier survival functions and Cox proportional hazards models. Subjects with the GSTM1 null genotype had shorter survival; the proportion of GSTM1 null subjects surviving at 5 years was 0.20 [95% confidence interval (CI) 0.14-0.27], compared with 0.29 (95% CI 0.22-0.37) for GSTM1 present subjects. The relative risk of death associated with GSTM1 null genotype, adjusted for stage at diagnosis and histology, was 1.36, 95% CI 1.04-1.80. There was no association between GSTT1 or GSTP1 genotype and survival in the overall study population, nor in a subgroup of patients treated with chemotherapy (n = 130). For GSTM1, our results are consistent with a previous study, which also observed that the GSTM1-null genotype, which confers susceptibility to lung cancer, was associated with shorter survival. Future studies of lung cancer survival should take into account GSTM1 genotype as well as investigate underlying mechanisms.


Subject(s)
Carcinoma, Large Cell/genetics , Carcinoma, Large Cell/mortality , Carcinoma, Small Cell/genetics , Carcinoma, Small Cell/mortality , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/mortality , Glutathione Transferase/genetics , Isoenzymes/genetics , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Polymorphism, Genetic/genetics , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Carcinoma, Large Cell/drug therapy , Carcinoma, Small Cell/drug therapy , Carcinoma, Squamous Cell/drug therapy , Case-Control Studies , Female , Follow-Up Studies , Genotype , Glutathione S-Transferase pi , Glutathione Transferase/drug effects , Humans , Isoenzymes/drug effects , Lung Neoplasms/drug therapy , Male , Middle Aged , Odds Ratio , Polymorphism, Genetic/drug effects , Proportional Hazards Models , Smoking/drug therapy , Smoking/genetics , Smoking/mortality , Statistics as Topic , Survival Analysis , Treatment Outcome , Washington/epidemiology
20.
Environ Health Perspect ; 111(8): 1055-64, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12826477

ABSTRACT

Interest is increasing in the role of variations in the human genome (polymorphisms) in modifying the effect of exposures to environmental health hazards (often referred to as gene-environment interaction), which render some individuals or groups in the population more or less likely to develop disease after exposure. This review is intended for an audience of environmental health practitioners and students and is designed to raise awareness about this rapidly growing field of research by presenting established and novel examples of gene-environment interaction that illustrate the major theme of effect modification. Current data gaps are identified and discussed to illustrate limitations of past research and the need for the application of more robust methods in future research projects. Two primary benefits of incorporating genetics into the existing environmental health research framework are illustrated: a) the ability to detect different levels of risk within the population, and b) greater understanding of etiologic mechanisms. Both offer opportunities for developing new methods of disease prevention. Finally, we describe a basic framework for researchers interested in pursuing health effects research that incorporates genetic polymorphisms.


Subject(s)
Environmental Exposure , Environmental Health , Genetic Predisposition to Disease , Polymorphism, Genetic , Humans , Research/trends , Research Design
SELECTION OF CITATIONS
SEARCH DETAIL