Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Adv Nutr ; 13(6): 2098-2114, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36084013

ABSTRACT

National health and nutrition monitoring is an important federal effort in the United States and Canada, and the basis for many of their nutrition and health policies. Understanding of child exposures through human milk (HM) remains out of reach due to lack of current and representative data on HM's composition and intake volume. This article provides an overview of the current national health and nutrition monitoring activities for HM-fed children, HM composition (HMC) and volume data used for exposure assessment, categories of potential measures in HM, and associated variability factors. In this Perspective, we advocate for a framework for collection and reporting of HMC data for national health and nutrition monitoring and programmatic needs, including a shared vision for a publicly available Human Milk Composition Data Repository (HMCD-R) to include essential metadata associated with HMC. HMCD-R can provide a central, integrated platform for researchers and public health officials for compiling, evaluating, and sharing HMC data. The compiled compositional and metadata in HMCD-R would provide pertinent measures of central tendency and variability and allow use of modeling techniques to approximate compositional profiles for subgroups, providing more accurate exposure assessments for purposes of monitoring and surveillance. HMC and related metadata could facilitate understanding the complexity and variability of HM composition, provide crucial data for assessment of infant and maternal nutritional needs, and inform public health policies, food and nutrition programs, and clinical practice guidelines.


Subject(s)
Milk, Human , Nutritional Status , Infant , Child , Humans , United States , Canada
2.
Mycotoxin Res ; 34(4): 241-255, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29971579

ABSTRACT

The presence of the mycotoxin ochratoxin A (OTA) in cereal grains is due to the growth of toxigenic Penicillium mold on stored crops. Human exposure to OTA is higher in infants, toddlers, and children than in adolescents and adults, based on exposure assessments of ng OTA consumed/kg body weight/day. Ochratoxin A is nephrotoxic and teratogenic in animals, but its effects on juveniles exposed during the reproduction and development period have not been studied. To address this, Fischer rats were exposed to 0, 0.16, 0.4, 1.0, or 2.5 mg OTA/kg diet throughout breeding, gestation, and lactation and its adverse effects were assessed in adult rats and their offspring on postnatal day (PND) 21. There were no effects on implantation but post-implantation fetotoxicity was observed in the 2.5 mg/kg dose group, corresponding to a calculated dose of 167.0 µg/kg bw/day in dams. Adverse effects on body and kidney weights and on clinical parameters indicative of renal toxicity were significant in adult rats exposed to 1.0 mg OTA/kg diet (55.2 and 73.3 µg/kg bw/day in adult males and females, respectively) and in PND21 rats at the 0.4 mg/kg dose (33.9 µg/kg bw/day in dams), suggesting that weanling rats were more sensitive to OTA than adults. Overall, nephrotoxicity was the primary effect of OTA in weanling rats exposed throughout gestation and lactation at sub-fetotoxic concentrations in diet.


Subject(s)
Ochratoxins/toxicity , Poisoning/pathology , Pregnancy Complications/pathology , Renal Insufficiency/pathology , Teratogens/toxicity , Abnormalities, Drug-Induced/epidemiology , Abnormalities, Drug-Induced/pathology , Administration, Oral , Animals , Disease Models, Animal , Female , Ochratoxins/administration & dosage , Pregnancy , Rats, Inbred F344 , Renal Insufficiency/chemically induced
3.
Food Chem Toxicol ; 110: 395-401, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29107027

ABSTRACT

The chemical amaranth (AM) is permitted as a colouring agent in a variety of foods. Safety was established based on chronic rodent studies. AM and its metabolite naphthionic acid (NA) can be absorbed through the intestine, exposing circulating immune cells including splenocytes. An AM feeding study in rats demonstrated an increase in blood lymphocytes. Yet, in contrast, AM inhibited the delayed-type hypersensitivity reaction to antigen. DO11.10 mice express a T Cell Receptor specific for ovalbumin323-339 peptide (OVAp) presented by I-Ad MHCII. DO11.10 splenocytes were cultured to evaluate mechanisms by which AM and NA modulate immune cell function in vitro. Exposure to OVAp alone for 72 h induced cell proliferation, and combination with 2 or 20 µg/mL AM increased IFN-γ. Cytotoxicity was evident at higher concentrations of AM (200 and 2000 µg/mL) and NA (2000 µg/mL) in combination with OVAp, as both cell number and cytokine secretion decreased. At 200 µg/mL AM with OVAp, immunotoxicity gene expression was modified and OVAp-specific KJ1-26+ CD28+ cells became enriched. The equivalent dose of NA did not modify those parameters. Using an antigen-specific model in vitro, lower concentrations of AM potentiated pro-inflammatory cytokine production, and higher concentrations of AM and NA demonstrated cytotoxicity.


Subject(s)
Amaranth Dye/pharmacology , Food Coloring Agents/pharmacology , Immunologic Factors/pharmacology , Spleen/immunology , Animals , Cells, Cultured , Female , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocytes/drug effects , Lymphocytes/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Naphthalenesulfonates/pharmacology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Spleen/cytology , Spleen/drug effects
4.
Toxicology ; 377: 1-13, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27932249

ABSTRACT

The brominated flame retardant TBECH is used as an additive to delay ignition and inhibit fires in construction materials and consumer goods. Trends in human exposure are not clear, although humans may be exposed to TBECH via indoor dust and air. In birds and fish there is some evidence of disruption in endocrine and reproductive parameters due to TBECH. In vitro studies indicate that TBECH is an androgen receptor agonist. In this study rats were exposed to 0, 10, 50, 250, 1250 or 5000mg/kg technical TBECH for 28days in diet, corresponding to 0, 0.9, 4.2, 21.3, 98.0 or 328.9mg TBECH/kg bw/d in males and 0, 0.8, 3.9, 19.4, 91.7 or 321.4mg TBECH/kg bw/d in females. Dose-dependent increases in α- and ß- TBECH were detected in serum, liver and adipose. Rats in the 5000mg/kg group lost weight rapidly and were euthanized after 15-18days. At study termination rats displayed dose-dependent clinical and histopathological changes consistent with mild hepatic and renal inflammation. In male rats, evidence of gender-specific alpha2u-globulin nephropathy was not considered predictive of renal toxicity in humans. Frank immunosuppression or inappropriate immunostimulation were not apparent, nor was there a primary effect of TBECH on adaptive immunity. Some evidence of hormone disruption was observed, including changes in serum testosterone levels in males and changes in serum T3 and T4 levels in females. Apparent increases in thyroid follicular cell hypertrophy and hyperplasia in male and female rats were not statistically significant. Benchmark dose (BMD) modelling indicated that clinical changes indicative of mild nephrotoxicity and increased blood monocyte numbers indicative of inflammation and tissue damage were the most sensitive outcomes of TBECH exposure that could be modelled. Preliminary evidence of hormone disruption supports the need for rodent studies using more sensitive models of growth, development and reproduction.


Subject(s)
Cyclohexanes/administration & dosage , Cyclohexanes/toxicity , Diet/adverse effects , Flame Retardants/administration & dosage , Flame Retardants/toxicity , Animals , Body Weight/drug effects , Body Weight/physiology , Cyclohexanes/blood , Dose-Response Relationship, Drug , Female , Flame Retardants/metabolism , Male , Organ Size/drug effects , Organ Size/physiology , Rats , Rats, Inbred F344
5.
Nanotoxicology ; 10(10): 1422-1430, 2016 12.
Article in English | MEDLINE | ID: mdl-27534448

ABSTRACT

Humans could become exposed to carbon black nanoparticles (CBNPs) in consumer products or an occupational setting. In rodent models, acute respiratory, subcutaneous, and direct immune cell exposure to CBNPs has been shown to enhance allergic sensitization to co-administered ovalbumin (OVA) protein from chicken egg. However, little is known about the effects of ingested CBNPs on immunological responses and oral tolerance to food antigens. We hypothesized that ingestion of CBNPs would enhance the development of food allergy to OVA. Allergy prone DO11.10 mice were orally exposed to CBNPs every second day for 2 weeks (total dose 10.8 (LOW) or 108 µg (HI)), with and without (±) co-administered OVA. Systemic immune parameters were measured at necropsy. Exposure to OVA resulted in significant increases in serum anti-OVA IgG1, anti-OVA IgM, and anti-OVA IgA antibodies relative to vehicle control. Immunophenotyping revealed a reduction in the number of OVA-specific CD4+ T helper cells upon OVA ± CBNPHI treatment in the spleen. Yet, secretion of the allergy-associated Th2 cytokines IL-4, IL-9, and IL-13 was greater in OVA323-339 peptide-pulsed splenocytes from OVA + CBNPHI-treated mice compared with control. Transcriptome analysis at necropsy of splenocytes from OVA + CBNPHI dose mice compared with OVA mice revealed increases in the allergy associated genes Il4 and Stat6 and decreases in Csf3r and Retnlg. Although oral exposure to high-dose CBNPs did not impact OVA-specific antibody production relative to OVA, we did observe increased expression of genes and cytokines associated with allergy in peripheral splenocytes. This work suggests that CBNP gastrointestinal exposure may potentiate allergy pathways.


Subject(s)
Food Hypersensitivity , Nanoparticles/toxicity , Ovalbumin/immunology , Receptors, Antigen, T-Cell/genetics , Soot/toxicity , Administration, Oral , Animals , Cytokines/metabolism , Female , Food Hypersensitivity/etiology , Food Hypersensitivity/genetics , Food Hypersensitivity/immunology , Immunoglobulins/blood , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Ovalbumin/genetics , Spleen/drug effects , Spleen/immunology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , Th2 Cells/drug effects , Th2 Cells/immunology
6.
Environ Toxicol Pharmacol ; 41: 201-10, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26724606

ABSTRACT

Perfluorooctanesulfonate (PFOS) has been widely used in a variety of industrial and commercial applications as a surfactant and stain repellent. PFOS causes liver damage (including liver tumors) in experimental animals, primarily via interaction with PPARα and CAR/PXR. We investigated the involvement of microRNAs (miRNAs) in PFOS-induced hepatotoxicity, and mechanisms involved in abnormal thyroid hormone (TH) homeostasis, in the livers of adult male rats exposed in feed to 50mg PFOS/kg diet for 28 days. PFOS-treated rats exhibited expected histopathological and clinical chemistry changes, and global gene expression changes consistent with the involvement of PPARα and CAR/PXR. Thirty-eight miRNAs were significantly altered. Three members of the miR-200 family were the most increased, while miR-122-5p and miR-21-5p were the most decreased, in PFOS-treated rats. Expression of the miR-23b-3p/27b-3p/24-3p cluster also decreased in PFOS-treated animals. Pathway analysis of miRNAs and associated gene expression changes suggests involvement of epithelial to mesenchymal transition (EMT), which is a primary process of tumor cell motility and cancer metastasis. Our analysis also revealed transcripts that may mediate PFOS-induced effects on TH homeostasis including: activation of the CAR/PXR pathway, phase II/III enzymes, and deiodinase. These changes are consistent with low serum TH due to enhanced metabolic clearance of TH. However, most TH hepatic target genes were not altered in a manner consistent with reduced TH signaling, suggesting that PFOS exposure did not induce functional hypothyroidism. Collectively, the study suggests an important role for miRNAs in PFOS-induced hepatotoxicity and provides insight into the effects of PFOS on TH homeostasis.


Subject(s)
Alkanesulfonic Acids/adverse effects , Fluorocarbons/adverse effects , Liver/drug effects , MicroRNAs/genetics , Thyroid Hormones/blood , Transcriptome/drug effects , Animals , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Homeostasis/drug effects , Liver/chemistry , Male , Potassium, Dietary/adverse effects , Rats , Rats, Sprague-Dawley
7.
Food Chem Toxicol ; 84: 250-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26363308

ABSTRACT

Microbial detoxification of deoxynivalenol (DON) represents a new approach to treating DON-contaminated grains. A bacterium Devosia mutans 17-2-E-8 was capable of completely transforming DON into a major product 3-epi-DON and a minor product 3-keto-DON. Evaluation of toxicities of these DON-transformation products is an important part of hazard characterization prior to commercialization of the biotransformation application. Cytotoxicities of the products were demonstrated by two assays: a MTT bioassay assessing cell viability and a BrdU assay assessing DNA synthesis. Compared with DON, the IC50 values of 3-epi-DON and 3-keto-DON were respectively 357 and 3.03 times higher in the MTT bioassay, and were respectively 1181 and 4.54 times higher in the BrdU bioassay. Toxicological effects of 14-day oral exposure of the B6C3F1 mouse to DON and 3-epi-DON were also investigated. Overall, there were no differences between the control (free of toxin) and the 25 mg/kg bw/day or 100 mg/kg bw/day 3-epi-DON treatments in body and organ weights, hematology and organ histopathology. However, in mice exposed to DON (2 mg/kg bw/day), white blood cell numbers and serum immunoglobulin levels were altered relative to controls, and lesions were observed in adrenals, thymus, stomach, spleen and colon. Taken together, in vitro and in vivo studies indicate that 3-epi-DON is substantially less toxic than DON.


Subject(s)
Hyphomicrobiaceae/metabolism , Nucleic Acid Synthesis Inhibitors/toxicity , Trichothecenes/toxicity , Administration, Oral , Animals , Caco-2 Cells , Cell Survival/drug effects , Crosses, Genetic , DNA/biosynthesis , Dose-Response Relationship, Drug , Female , Humans , Inactivation, Metabolic , Kinetics , Mice , NIH 3T3 Cells , Nucleic Acid Synthesis Inhibitors/administration & dosage , Nucleic Acid Synthesis Inhibitors/chemistry , Nucleic Acid Synthesis Inhibitors/metabolism , Random Allocation , Stereoisomerism , Toxicity Tests, Subacute , Trichothecenes/administration & dosage , Trichothecenes/chemistry , Trichothecenes/metabolism
8.
Article in English | MEDLINE | ID: mdl-26247304

ABSTRACT

In an investigation of deoxynivalenol (DON)-transformation products by Devosia mutans 17-2-E-8, the major product was identified as 3-epi-DON. This DON-transformation product was analysed by liquid chromatography and identified by congruent retention time and UV/Vis spectrum, as well as mass spectrometric data. Nuclear magnetic resonance (NMR) experiments including correlation spectroscopy (COSY), heteronuclear single quantum coherence (HSQC) and nuclear overhauser effect (NOE) were conducted for structural characterisation of 3-epi-DON. High-speed counter-current chromatography (HSCCC) was applied to scale up the separation of 3-epi-DON from DON in a D. mutans 17-2-E-8 culture. From the culture where 100 mg DON was applied, 56 mg of 3-epi-DON (purity of 96.8%) was obtained from the HSCCC. The purified 3-epi-DON will be used for toxicological characterisation studies of this chemical.


Subject(s)
Hyphomicrobiaceae/chemistry , Trichothecenes/chemistry , Trichothecenes/isolation & purification , Chromatography, High Pressure Liquid , Countercurrent Distribution , Magnetic Resonance Spectroscopy , Mass Spectrometry , Molecular Conformation , Spectrophotometry, Ultraviolet
9.
Mutat Res ; 773: 63-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25847422

ABSTRACT

Aflatoxin B1 (AFB1) is produced by species of Aspergillus, and is a known human carcinogen. AFB1-induced oxidative DNA damage, specifically 8-hydroxy-2-deoxyguanosine (8-OHdG) lesions, has been demonstrated in both animal models and in humans, and is repaired by base excision repair (BER). The tumour suppressor gene p53 is implicated in the regulation of DNA repair, and heterozygous p53 knockouts have an attenuated nucleotide excision repair response to AFB1. Male heterozygous p53 knockout mice and their wild-type controls were exposed to 0, 0.2 or 1.0ppm AFB1 for 26 weeks in their diet. BER activity of lung and liver was assessed with an in vitro assay, using 8-OHdG-damaged plasmid DNA as a substrate. BER activity did not differ between livers or lungs from untreated wild-type versus heterozygous p53 knockout mice. In wild-type mice, repair was 65% lower in liver extracts from mice exposed to 1.0ppm AFB1 than in liver extracts from mice exposed to 0.2ppm AFB1 (p<0.05), but not significantly lower than that in liver extracts from control mice. AFB1 did not affect BER in lung extracts from wild-type mice, or in lung and liver extracts from heterozygous p53 knockout mice. In liver and lung, AFB1 exposure did not alter levels of 8-oxoguanine glycosylase protein, a key enzyme in the repair of 8-OHdG, and did not cause hepatotoxicity, as indicated by plasma alanine aminotransferase levels. In conclusion, chronic exposure to AFB1 did not affect BER in lungs or livers of heterozygous p53 knockout mice. BER activity was lower in livers from p53 wild type mice exposed to 1.0ppm AFB1 versus those exposed to 0.2ppm AFB1, an effect that was not attributable to liver cell death or altered levels of 8-oxoguanine glycosylase.


Subject(s)
Aflatoxin B1/toxicity , DNA Repair , Liver/drug effects , Lung/drug effects , Tumor Suppressor Protein p53/genetics , Animals , Genotype , Health Status , Liver/metabolism , Lung/metabolism , Male , Mice , Mice, Knockout , Tumor Suppressor Protein p53/physiology
10.
Toxicol Pathol ; 43(5): 715-29, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25694087

ABSTRACT

Exposure to the mycotoxin ochratoxin A (OTA) causes nephropathy in domestic animals and rodents and renal tumors in rodents and poultry. Humans are exposed to OTA by consuming foods made with contaminated cereal grains and other commodities. Management of human health risks due to OTA exposure depends, in part, on establishing a mode of action (MOA) for OTA carcinogenesis. To further investigate OTA's MOA, p53 heterozygous (p53+/-) and p53 homozygous (p53+/+) mice were exposed to OTA in diet for 26 weeks. The former are susceptible to tumorigenesis upon chronic exposure to genotoxic carcinogens. OTA-induced renal damage but no tumors were observed in either strain, indicating that p53 heterozygosity conferred little additional sensitivity to OTA. Renal changes included dose-dependent increases in cellular proliferation, apoptosis, karyomegaly, and tubular degeneration in proximal tubules, which were consistent with ochratoxicosis. The lowest observed effect level for renal changes in p53+/- and p53+/+ mice was 200 µg OTA/kg bw/day. Based on the lack of tumors and the severity of renal and body weight changes at a maximum tolerated dose, the results were interpreted as suggestive of a primarily nongenotoxic (epigenetic) MOA for OTA carcinogenesis in this mouse model.


Subject(s)
Ochratoxins/toxicity , Tumor Suppressor Protein p53/genetics , Animals , Eating/drug effects , Immunohistochemistry , Kidney/drug effects , Leukocytes/drug effects , Liver/drug effects , Male , Mice , Mice, Knockout , Organ Size/drug effects , Toxicity Tests, Chronic
11.
Nanotoxicology ; 9(4): 523-42, 2015 May.
Article in English | MEDLINE | ID: mdl-25119418

ABSTRACT

Engineered metal/mineral, lipid and biochemical macromolecule nanomaterials (NMs) have potential applications in food. Methodologies for the assessment of NM digestion and bioavailability in the gastrointestinal tract are nascent and require refinement. A working group was tasked by the International Life Sciences Institute NanoRelease Food Additive project to review existing models of the gastrointestinal tract in health and disease, and the utility of these models for the assessment of the uptake of NMs intended for food. Gastrointestinal digestion and absorption could be addressed in a tiered approach using in silico computational models, in vitro non-cellular fluid systems and in vitro cell culture models, after which the necessity of ex vivo organ culture and in vivo animal studies can be considered. Examples of NM quantification in gastrointestinal tract fluids and tissues are emerging; however, few standardized analytical techniques are available. Coupling of these techniques to gastrointestinal models, along with further standardization, will further strengthen methodologies for risk assessment.


Subject(s)
Digestion , Food , Gastrointestinal Tract/physiology , Models, Biological , Nanostructures , Animals , Humans
12.
J Nanopart Res ; 16: 2440, 2014.
Article in English | MEDLINE | ID: mdl-24944520

ABSTRACT

The increasing use of zinc oxide nanoparticles (ZnO-NPs) has raised concerns about their potential hazards to human and environmental health. In this study, the characterization and cytotoxicity of two ZnO-NPs products (Z-COTE and Z-COTE HP1) were investigated. The zinc content of Z-COTE and Z-COTE HP1 was 82.5 ± 7.3 and 80.1 ± 3.5 %, respectively. Both ZnO-NP samples contained sub-cytotoxic levels of iron and copper, and silicon was detected from the surface coating of Z-COTE HP1. All samples were highly agglomerated, and the primary particles appeared as variable polyhedral structures. There was no significant difference in size distribution or average diameter of Z-COTE (53 ± 23 nm) and Z-COTE HP1 (54 ± 26 nm). A dose-dependent cytotoxicity was observed 24 h after exposure to ZnO-NPs, and monocytes were more sensitive than lung epithelial cells or lymphoblasts in both human and mouse cells. There was a significant difference in cytotoxicity between nano- and fine-forms, but only at the threshold cytotoxic dose with cellular metabolism assays. Compared to uncoated ZnO-NPs, the surface coating with triethoxycaprylylsilane marginally attenuated cellular oxidative stress and protected cellular metabolic activity. These results demonstrate the importance of model cell type, dose selection, and cytotoxicity assessment methodology to accurately evaluate the potential toxicity of various nanoparticles in vitro.

13.
Toxicol Sci ; 138(2): 322-32, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24449417

ABSTRACT

Agglomerated carbon black nanoparticles (CBNPs) administered via respiratory or subcutaneous routes have been shown to promote allergic sensitization to coadministered ovalbumin (OVA) protein in rodents. In the present study, we aimed to model and elucidate the mechanism of this adjuvanticity using an in vitro assay based on T cell sensitization to ovalbumin323₋339 peptide (OVA(p)). CBNP base particles of 22 and 39 nm were characterized and termed CBNP22 and CBNP39 powders. Splenic leukocytes derived from transgenic DO11.10 mice were exposed to suspensions of media alone, concanavalin A mitogen, CBNP agglomerates smaller than 220 nm, OVA(p) alone, OVA(p) + anti-CD28 costimulant, OVA(p) + cyclosporin A immunosuppressant, or OVA(p) + CBNPs. Samples were analyzed at 72 h post-exposure. Proliferation rate, a marker of cellular mitosis, was assessed. Polymerase chain reaction arrays were used to assess genes involved in allergic response pathways. The mitogen control, costimulatory control, and immunosuppressive control chemicals modified the T helper cell proliferation rate. CBNP22 mildly reduced proliferation at 12 µg/ml, but CBNP39 did not. Gene expression analysis of cells treated with OVA(p) showed that coincubation with 12 µg/ml CBNP22 enhanced gene expression of interleukin-4 (IL-4), IL-10, and IL-13, all allergy-associated Th2 cytokines. Coincubation of OVA(p) with 12 µg/ml CBNP39 significantly enhanced IL-13 gene expression concurrent with downregulation of the Th1-associated transcription factor Stat4. IL-4 and IL-13 protein secretion reflected the mRNA trends. The changes were consistently higher in cells exposed to CBNP22 than CBNP39, suggesting that smaller particle size, higher surface area, and higher purity were associated with the direct adjuvant effect on Th2 cells in this genetically susceptible model of OVA allergy.


Subject(s)
Adjuvants, Immunologic/toxicity , Hypersensitivity/immunology , Nanoparticles/chemistry , Ovalbumin/immunology , Soot/toxicity , Th2 Cells/drug effects , Adjuvants, Immunologic/chemistry , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Female , Flow Cytometry , Hypersensitivity/etiology , Male , Mice , Mice, Inbred Strains , Microscopy, Electron, Scanning , Particle Size , Porosity , Primary Cell Culture , Soot/chemistry , Surface Properties , Th2 Cells/immunology
14.
Toxicol Appl Pharmacol ; 275(2): 96-103, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24380836

ABSTRACT

Aflatoxin B1(AFB1) is biotransformed in vivo into an epoxide metabolite that forms DNA adducts that may induce cancer if not repaired. p53 is a tumor suppressor gene implicated in the regulation of global nucleotide excision repair (NER). Male heterozygous p53 knockout (B6.129-Trp53(tm1Brd)N5, Taconic) and wild-type mice were exposed to 0, 0.2 or 1.0 ppm AFB1 for 26 weeks. NER activity was assessed with an in vitro assay, using AFB1-epoxide adducted plasmid DNA as a substrate. For wild-type mice, repair of AFB1-N7-Gua adducts was 124% and 96% greater in lung extracts from mice exposed to 0.2 ppm and 1.0 ppm AFB1respectively, and 224% greater in liver extracts from mice exposed to 0.2 ppm AFB1( p<0.05). In heterozygous p53 knockout mice, repair of AFB1-N7-Gua was only 45% greater in lung extracts from mice exposed to 0.2 ppm AFB1 (p<0.05), and no effect was observed in lung extracts from mice treated with 1.0 ppm AFB1or in liver extracts from mice treated with either AFB1concentration. p53 genotype did not affect basal levels of repair. AFB1exposure did not alter repair of AFB1-derived formamidopyrimidine adducts in lung or liver extracts of either mouse genotype nor did it affect XPA or XPB protein levels. In summary, chronic exposure to AFB1increased NER activity in wild-type mice, and this response was diminished in heterozygous p53 knockout mice, indicating that loss of one allele of p53 limits the ability of NER to be up-regulated in response to DNA damage.


Subject(s)
Aflatoxin B1/toxicity , DNA Repair/drug effects , DNA Repair/genetics , Tumor Suppressor Protein p53/genetics , Up-Regulation , Alleles , Animals , Biotransformation , DNA Damage/drug effects , Genotype , Liver/drug effects , Liver/metabolism , Lung/drug effects , Lung/metabolism , Male , Mice , Mice, Knockout , Tumor Suppressor Protein p53/metabolism , Xeroderma Pigmentosum Group A Protein/genetics , Xeroderma Pigmentosum Group A Protein/metabolism
15.
J Toxicol Environ Health A ; 76(24): 1333-45, 2013.
Article in English | MEDLINE | ID: mdl-24283475

ABSTRACT

Arsenobetaine (ASB) is the major form of arsenic (As) in seafood sources such as molluscs and fish. Limited data demonstrated that ASB toxicity in mammals is minimal; however, data on possible reproductive effects are lacking. This study investigated the tissue distribution and developmental effects of ASB during pregnancy, early postnatal life, and development to adulthood. Pregnant rats were randomly assigned to 3 cohorts and gavaged daily from gestational day 8 (GD8) with ASB in deionized water at 0, 0.1, 1, or 10 mg/kg body weight (bw)/d. Cohort 1 dams were sacrificed on GD20 (n = 6 per dose group), cohort 2 dams and pups were sacrificed on postnatal day 13 (PND13; n = 4 dams per dose group), and cohort 3 pups (n = 2 dams per dose group) were sacrificed on PND90. Residue analysis detected significant levels of ASB in livers of cohort 1 dams and lower levels in cohort 1 GD20 fetuses, as well as in cohort 2 male and female offspring, indicating placental transfer from the maternal circulation in utero. Trace amounts of ASB in dams' milk were found only in the 10-mg/kg bw/d dose cohort 2 (PND13), demonstrating that lactational transfer was limited. ASB levels in liver varied during pregnancy, lactation, and postweaning, with levels falling rapidly as these physiological states progress. Although transfer of ASB through the placenta to the fetuses and to a limited extent through milk was confirmed, ASB exposure during pregnancy and lactation appeared to produce no teratogenic or deleterious effects on reproductive development.


Subject(s)
Arsenicals/adverse effects , Liver/metabolism , Maternal Exposure/adverse effects , Milk/chemistry , Prenatal Exposure Delayed Effects/chemically induced , Spermatids/drug effects , Testosterone/blood , Administration, Oral , Animals , Animals, Newborn , Chromatography, Liquid , Dose-Response Relationship, Drug , Female , Fetus , Lactation , Liver/drug effects , Male , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/physiopathology , Random Allocation , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Electrospray Ionization
16.
Crit Rev Toxicol ; 43 Suppl 2: 1-24, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24164514

ABSTRACT

There is disagreement internationally across major regulatory jurisdictions on the relevance and utility of whole food (WF) toxicity studies on GM crops, with no harmonization of data or regulatory requirements. The scientific value, and therefore animal ethics, of WF studies on GM crops is a matter addressable from the wealth of data available on commercialized GM crops and WF studies on irradiated foods. We reviewed available GM crop WF studies and considered the extent to which they add to the information from agronomic and compositional analyses. No WF toxicity study was identified that convincingly demonstrated toxicological concern or that called into question the adequacy, sufficiency, and reliability of safety assessments based on crop molecular characterization, transgene source, agronomic characteristics, and/or compositional analysis of the GM crop and its near-isogenic line. Predictions of safety based on crop genetics and compositional analyses have provided complete concordance with the results of well-conducted animal testing. However, this concordance is primarily due to the improbability of de novo generation of toxic substances in crop plants using genetic engineering practices and due to the weakness of WF toxicity studies in general. Thus, based on the comparative robustness and reliability of compositional and agronomic considerations and on the absence of any scientific basis for a significant potential for de novo generation of toxicologically significant compositional alterations as a sole result of transgene insertion, the conclusion of this review is that WF animal toxicity studies are unnecessary and scientifically unjustifiable.


Subject(s)
Food Safety/methods , Food, Genetically Modified/toxicity , Plants, Genetically Modified/toxicity , Animal Experimentation/ethics , Animal Experimentation/legislation & jurisprudence , Animals , Genetic Engineering/methods , Humans , Reproducibility of Results , Risk Assessment/ethics , Risk Assessment/methods , Toxicity Tests/ethics , Toxicity Tests/methods
17.
Paediatr Perinat Epidemiol ; 27(4): 415-25, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23772943

ABSTRACT

BACKGROUND: The Maternal-Infant Research on Environmental Chemicals (MIREC) Study was established to obtain Canadian biomonitoring data for pregnant women and their infants, and to examine potential adverse health effects of prenatal exposure to priority environmental chemicals on pregnancy and infant health. METHODS: Women were recruited during the first trimester from 10 sites across Canada and were followed through delivery. Questionnaires were administered during pregnancy and post-delivery to collect information on demographics, occupation, life style, medical history, environmental exposures and diet. Information on the pregnancy and the infant was abstracted from medical charts. Maternal blood, urine, hair and breast milk, as well as cord blood and infant meconium, were collected and analysed for an extensive list of environmental biomarkers and nutrients. Additional biospecimens were stored in the study's Biobank. The MIREC Research Platform encompasses the main cohort study, the Biobank and follow-up studies. RESULTS: Of the 8716 women approached at early prenatal clinics, 5108 were eligible and 2001 agreed to participate (39%). MIREC participants tended to smoke less (5.9% vs. 10.5%), be older (mean 32.2 vs. 29.4 years) and have a higher education (62.3% vs. 35.1% with a university degree) than women giving birth in Canada. CONCLUSIONS: The MIREC Study, while smaller in number of participants than several of the international cohort studies, has one of the most comprehensive datasets on prenatal exposure to multiple environmental chemicals. The biomonitoring data and biological specimen bank will make this research platform a significant resource for examining potential adverse health effects of prenatal exposure to environmental chemicals.


Subject(s)
Environmental Pollutants/adverse effects , Infant Welfare , Maternal Exposure/adverse effects , Prenatal Exposure Delayed Effects/chemically induced , Adolescent , Adult , Biomarkers , Canada , Cohort Studies , Environmental Exposure/adverse effects , Environmental Monitoring/methods , Female , Humans , Infant , Male , Pregnancy , Surveys and Questionnaires , Young Adult
18.
Environ Toxicol ; 28(4): 215-28, 2013 Apr.
Article in English | MEDLINE | ID: mdl-21544923

ABSTRACT

Brominated diphenyl ethers (BDEs) are persistent environmental contaminants found in human blood, tissues, and milk. To assess the impact of the commercial BDE mixture DE-71 on the developing immune system in relation to hepatic and thyroid changes, adult (F0) rats were exposed to DE-71 by gavage at doses of 0, 0.5, 5, or 25 mg/kg body weight (bw)/d for 21 weeks. F0 rats were bred and exposure continued through gestation, lactation and postweaning. F1 pups were weaned and exposed to DE-71 by gavage from postnatal day (PND) 22 to 42. On PND 42, half of the F1 rats were assessed for toxicologic changes. The remaining F1 rats were challenged with the T-dependent antigen keyhole limpet hemocyanin (KLH) and immune function was assessed on PND 56. Dose-dependent increases in total BDE concentrations were detected in the liver and adipose of all F0 and F1 rats. In F0 rats, increased liver weight, hepatocellular hypertrophy, and decreased serum thyroxine (T4) were characteristic of DE-71 exposure. In F1 rats perinatal DE-71 exposure caused a nondose-dependent increase in body weight and dose-dependent increases in liver weight and hepatocellular hypertrophy. Serum T3 and T4 levels were decreased. In spleen from DE-71 exposed rats the area occupied by B cells declined while the area occupied by T cells increased; however, cellular and humoral immune responses to KLH challenge were not altered. Thus hepatic and thyroid changes in rats exposed perinatally to DE-71 were associated with altered splenic lymphocyte populations, an effect which has been linked to hypothyroidism.


Subject(s)
Environmental Pollutants/toxicity , Halogenated Diphenyl Ethers/toxicity , Immune System/drug effects , Prenatal Exposure Delayed Effects , Adult , Animals , Body Weight/drug effects , Environmental Pollutants/analysis , Environmental Pollutants/immunology , Female , Halogenated Diphenyl Ethers/analysis , Halogenated Diphenyl Ethers/immunology , Humans , Immune System/embryology , Immune System/growth & development , Lactation , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Maternal Exposure/adverse effects , Milk/chemistry , Organ Size/drug effects , Paternal Exposure/adverse effects , Pregnancy , Rats , Rats, Sprague-Dawley , Reproduction/drug effects , Spleen/drug effects , Spleen/pathology , Thyroid Hormones/blood , Time Factors , Tissue Distribution , Weaning
19.
Food Chem Toxicol ; 50(10): 3604-13, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22841953

ABSTRACT

The fungal toxin fumonisin B1 (FB1) is a potential human carcinogen based on evidence of renal carcinogenicity in rats and hepatocarcinogenicity in mice. The toxicity and carcinogenicity of FB1 is linked to ceramide synthase inhibition. Based on this mechanism of action and on lack of evidence of genotoxicity, FB1 is considered a non-genotoxic carcinogen. The p53 heterozygous (p53+/-) mouse is a cancer-prone model used for carcinogenesis. The effects of chronic dietary FB1 exposure were characterized in p53+/- mice to confirm non-genotoxicity using a model which is more sensitive to genotoxic than non-genotoxic carcinogens and to clarify the relationship between p53 expression, altered sphingolipid metabolism, and FB1-induced carcinogenesis. Responses to FB1 were similar in p53+/- and p53+/+ mice after 26 weeks exposure to 0, 5, 50 or 150 mg FB1/kg diet, supporting a non-genotoxic mechanism of action. Hepatic adenomas and cholangiomas were observed in mice exposed to 150 mg/kg FB1. For a 10% increase in hepatic megalocytosis, the estimated 95% lower confidence limit of the benchmark dose (BMDL10) ranged from 0.15 and 1.11 mg FB1/kg bw/day. Based on similar responses in p53+/- and p53+/+ mice, p53 and related pathways play a secondary role in responses to FB1 toxicity and carcinogenesis.


Subject(s)
Fumonisins/toxicity , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Adenoma, Bile Duct/chemically induced , Adenoma, Liver Cell/chemically induced , Animals , Chemical and Drug Induced Liver Injury/pathology , Diet , Dose-Response Relationship, Drug , Drug Administration Schedule , Heterozygote , Homozygote , Liver/cytology , Liver/drug effects , Liver Neoplasms/chemically induced , Mice , Mice, Transgenic , Random Allocation
20.
J Biol Chem ; 284(8): 4786-95, 2009 Feb 20.
Article in English | MEDLINE | ID: mdl-19095642

ABSTRACT

Fumonisin B(1) (FB(1)) is a mycotoxin that inhibits ceramide synthases (CerS) and causes kidney and liver toxicity and other disease. Inhibition of CerS by FB(1) increases sphinganine (Sa), Sa 1-phosphate, and a previously unidentified metabolite. Analysis of the latter by quadrupole-time-of-flight mass spectrometry assigned an m/z = 286.3123 in positive ionization mode, consistent with the molecular formula for deoxysphinganine (C(18)H(40)NO). Comparison with a synthetic standard using liquid chromatography, electrospray tandem mass spectrometry identified the metabolite as 1-deoxysphinganine (1-deoxySa) based on LC mobility and production of a distinctive fragment ion (m/z 44, CH(3)CH=NH (+)(2)) upon collision-induced dissociation. This novel sphingoid base arises from condensation of alanine with palmitoyl-CoA via serine palmitoyltransferase (SPT), as indicated by incorporation of l-[U-(13)C]alanine into 1-deoxySa by Vero cells; inhibition of its production in LLC-PK(1) cells by myriocin, an SPT inhibitor; and the absence of incorporation of [U-(13)C]palmitate into 1-[(13)C]deoxySa in LY-B cells, which lack SPT activity. LY-B-LCB1 cells, in which SPT has been restored by stable transfection, however, produce large amounts of 1-[(13)C]deoxySa. 1-DeoxySa was elevated in FB(1)-treated cells and mouse liver and kidney, and its cytotoxicity was greater than or equal to that of Sa for LLC-PK(1) and DU-145 cells. Therefore, this compound is likely to contribute to pathologies associated with fumonisins. In the absence of FB(1), substantial amounts of 1-deoxySa are made and acylated to N-acyl-1-deoxySa (i.e. 1-deoxydihydroceramides). Thus, these compounds are an underappreciated category of bioactive sphingoid bases and "ceramides" that might play important roles in cell regulation.


Subject(s)
Enzyme Inhibitors/pharmacology , Fumonisins/pharmacology , Kidney/enzymology , Lipid Metabolism/drug effects , Liver/enzymology , Oxidoreductases/antagonists & inhibitors , Sphingosine/analogs & derivatives , Animals , Chlorocebus aethiops , Humans , Mice , Oxidoreductases/metabolism , Sphingosine/metabolism , Swine , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...