Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Data Brief ; 27: 104763, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31828184

ABSTRACT

Dietary acrylamide, a thermally induced food contaminant, at a level (2 mg/kg diet) typifying higher occurrence in certain food products - is neither an independent carcinogen nor a tumor promoter in the colon. This is evidenced by our previous studies using the medium-term azoxymethane (AOM)-induced colon tumorigenesis assay in F344 rats and the human colon tumor xenograft model in athymic nude (nu/nu) mice (https://doi.org/10.1371/journal.pone.0073916) [1]. In addition, we found that acrylamide may act as a colon co-carcinogen in association with a known carcinogen (AOM) in F344 rats. Furthermore, exposure to acrylamide at 2 mg/kg in the diet was not associated with any toxicologically relevant changes in clinical biochemistry, hematology, and apical endpoints in healthy rats (exposed only to saline injections) (https://doi.org/10.1016/j.toxrep.2016.08.010) [2]. Here we report data from our previous investigation [1] on gene expression of cancer pathway targets as well as the methylation status of select tumor suppressor genes. Briefly, mRNA and DNA were extracted from (a) colon mucosae and tumors from F344 rats exposed to AOM or saline and (b) athymic nude (nu/nu) mice bearing human colon tumor xenografts, both exposed to dietary acrylamide at concentrations of 0 or 2 mg/kg diet for 20 and 4 weeks, respectively. RT2 Profiler PCR Cancer PathwayFinder Arrays (Qiagen) and EpiTect Methyl II DNA Restriction kits and PCR Assays (Qiagen) were used to detect cancer-relevant gene expression (84 genes representing 9 pathways) and the methylation status of the CpG islands associated with 22 tumor suppressor genes in colon mucosae, tumors and xenografts. Additionally, RT2 Profiler PCR Arrays (Qiagen) for cell cycle regulation, growth factors, inflammatory cytokines and receptors, and inflammatory response and autoimmunity were used to investigate the gene expression (84 genes in each array) of targets involved in these select cellular pathways in the colon mucosae from AOM-treated F344 rats.

2.
Data Brief ; 7: 460-7, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27014731

ABSTRACT

We previously reported that dietary acrylamide, at doses (10 and 50 mg/kg diet) known to cause rodent tumors, lowered serum total high density lipoprotein and total testosterone, increased serum lipase, and lowered lymphocytes levels together with other hematological parameters in male F344 rats exposed for 10 weeks (doi: 10.1016/j.etap.2014.11.009 [1]). Here we present data related to the role of food-borne acrylamide exposure (at 0, 5, 10 and 50 mg/kg diet) in the presence of low (7% wt/wt) or high (23.9% wt/wt) dietary fat on serum and urinary markers of oxidative stress and inflammation in F344 rats. Briefly, urine and serum samples were collected from the experimental animals a day prior to or at the time of necropsy, respectively and processed for enzyme-linked immunosorbent assay estimations of biochemical markers. Urine samples were analyzed for 8-hydroxydeoxyguanosine and isoprostane, and serum samples for total antioxidant capacity, paraoxonase 1 activity, c-reactive protein, homocysteine, oxidized low-density lipoprotein, intercellular adhesion molecule-1, thromboxin 2, and Nε-(carboxymethyl)lysine.

3.
Toxicol Rep ; 3: 673-678, 2016.
Article in English | MEDLINE | ID: mdl-28959591

ABSTRACT

Understanding the health hazards following exposure to food-borne acrylamide, especially at low levels typified by human diets, is an ongoing food safety issue. We recently published results from a study that aimed to understand the effects of acrylamide short-term exposure at doses known to cause tumors in rodents, demonstrating that a number of key toxicological end points were altered by acrylamide exposure. Additionally, we reported that at much lower doses for 30 weeks of exposure, dietary acrylamide was 'not a complete carcinogen' to the colon in an organ-specific rodent carcinogenesis study but acted as a co-carcinogen along with azoxymethane (AOM, a colon-specific carcinogen). Here, we present toxicological data from a sub-set of this long-term exposure study from animals that received saline (instead of AOM). Briefly, male F344 rats were randomized to receive acrylamide at 0.5, 1.0 and 2.0 mg/kg diet (∼0.02, 0.04, and 0.09 mg/kg BW/day, respectively) or no acrylamide (control), for 30 weeks; all rats were then euthanized and their tissues harvested and processed for toxicological evaluation. We report that at the doses tested, acrylamide did not cause any changes in general well-being, body weight or food intake. Similarly, acrylamide did not cause any biologically relevant change in parameters associated with immunophenotyping, serum biochemistry or hematology. Histopathology assessment of tissues showed no changes except in the testis, where non-specific mild lesions were observed in all the groups, inclusive of the controls. No neuropathological effects of acrylamide were observed in the brain and nerve tissues. Together, these results suggest that acrylamide administered to rats through the diet at low doses for 30 weeks did not cause any toxicologically relevant changes. Given that the doses of acrylamide in the current study are low and are comparable to human dietary exposure, this null-effect study provides data that contribute to the body of scientific evidence relevant to understanding the health effects of acrylamide.

4.
Carcinogenesis ; 36 Suppl 1: S38-60, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26106143

ABSTRACT

The aim of this work is to review current knowledge relating the established cancer hallmark, sustained cell proliferation to the existence of chemicals present as low dose mixtures in the environment. Normal cell proliferation is under tight control, i.e. cells respond to a signal to proliferate, and although most cells continue to proliferate into adult life, the multiplication ceases once the stimulatory signal disappears or if the cells are exposed to growth inhibitory signals. Under such circumstances, normal cells remain quiescent until they are stimulated to resume further proliferation. In contrast, tumour cells are unable to halt proliferation, either when subjected to growth inhibitory signals or in the absence of growth stimulatory signals. Environmental chemicals with carcinogenic potential may cause sustained cell proliferation by interfering with some cell proliferation control mechanisms committing cells to an indefinite proliferative span.


Subject(s)
Carcinogens, Environmental/adverse effects , Cell Proliferation/drug effects , Environmental Exposure/adverse effects , Hazardous Substances/adverse effects , Neoplasms/chemically induced , Neoplasms/etiology , Signal Transduction/drug effects , Animals , Humans
5.
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
6.
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
7.
Environ Toxicol Pharmacol ; 39(1): 85-92, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25473820

ABSTRACT

We recently reported that acrylamide, a known rodent and probable human carcinogen, does not increase the risk of azoxymethane (AOM)-induced rat colon precancerous lesions when administered through the diet. Here, we present toxicological data from non-AOM-injected rats. Briefly, male F344 rats were randomized into four dietary groups and received experimental diets based on AIN-93G formulation and containing acrylamide at 0 (control), 5, 10 or 50mg/kg diet (wt/wt) ad libitum for 10 weeks, after which they were killed and their blood collected for hematological and biochemical markers. Acrylamide at the higher doses (10 and 50mg/kg diet) significantly lowered (p<0.05) serum total high density lipoprotein and total testosterone and increased serum lipase in comparison to the control. Blood hematocrit values and lymphocyte counts were significantly lower (p<0.05) in the high dose acrylamide (50mg/kg diet) group compared to control, with a concomitant decrease in hemoglobin level, mean corpuscular volume and mean corpuscular hemoglobin. These results provide additional hazard characterization data and strengthen the notion that at high doses, acrylamide may involve systemic toxicity potentiating tumorigenesis in experimental animals. Further studies are required to understand the health effects of food-borne acrylamide, especially at the lower exposures typified by human diets.


Subject(s)
Acrylamide/toxicity , Carcinogens/toxicity , Animals , Diet , Hematocrit , Lipase/blood , Lipoproteins, HDL/blood , Lymphocyte Count , Male , Rats, Inbred F344 , Testosterone/blood
8.
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
9.
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
10.
Genes Nutr ; 9(1): 373, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24292949

ABSTRACT

Accumulation of hepatic lipid droplet (HLD) is the hallmark pathology of non-alcoholic fatty liver disease (NAFLD). This study examined the effects of soy isoflavones (ISF) and different amounts of soy proteins on the accumulation of HLD, lipid metabolism and related gene expression in rats. Weanling Sprague-Dawley rats were fed diets containing either 20 % casein protein without (D1) or with (D2) supplemental ISF (50 mg/kg diet) or substitution of casein with increasing amounts of alcohol-washed soy protein isolate (SPI, 5, 10, and 20 %; D3, D4, D5) for 90 days. Dietary casein (20 %) induced accumulation of HLD in female, but not in male rats. Both soy proteins and ISF remarkably prevented the formation of HLD. Soy proteins lowered hepatic total cholesterol and triglyceride in a dose-dependent manner. Interestingly, soy proteins but not ISF significantly increased free fatty acids in the liver of the female rats compared to D1. Proteomic analysis showed that at least 3 enzymes involved in lipogenesis were down-regulated and 7 proteins related to fatty acid ß-oxidation or lipolysis were up-regulated by soy protein over D1. Additionally, 9 differentially expressed proteins identified were related to amino acid metabolism, 5 to glycolysis and 2 to cholesterol metabolism. Dietary ISF and SPI markedly reduced hepatic-peroxisome-proliferator-activated receptor γ2 (PPARγ2) and fat-specific protein 27 (FSP27) in female rats. Overall, this study has shown that partial or full replacement of dietary casein by soy protein or supplementation with soy ISF can effectively prevent the accumulation of HLD. The potential molecular mechanism(s) involved might be due to suppression of lipogenesis and stimulation of lipolysis and down-regulation of PPARγ2 and FSP27. This suggests that consumption of soy foods or supplements might be a useful strategy for the prevention or treatment of fatty liver diseases.

11.
PLoS One ; 8(9): e73916, 2013.
Article in English | MEDLINE | ID: mdl-24040114

ABSTRACT

Acrylamide, a possible human carcinogen, is formed in certain carbohydrate-rich foods processed at high temperature. We evaluated if dietary acrylamide, at doses (0.5, 1.0 or 2.0 mg/kg diet) reflecting upper levels found in human foods, modulated colon tumorigenesis in two rodent models. Male F344 rats were randomized to receive diets without (control) or with acrylamide. 2-weeks later, rats in each group received two weekly subcutaneous injections of either azoxymethane (AOM) or saline, and were killed 20 weeks post-injections; colons were assessed for tumors. Male athymic nude (nu/nu) mice bearing HT-29 human colon adenocarcinoma cells-derived tumor xenografts received diets without (control) or with acrylamide; tumor growth was monitored and mice were killed 4 weeks later. In the F344 rat study, no tumors were found in the colons of the saline-injected rats. However, the colon tumor incidence was 54.2% and 66.7% in the control and the 2 mg/kg acrylamide-treated AOM-injected groups, respectively. While tumor multiplicity was similar across all diet groups, tumor size and burden were higher in the 2 mg/kg acrylamide group compared to the AOM control. These results suggest that acrylamide by itself is not a "complete carcinogen", but acts as a "co-carcinogen" by exacerbating the effects of AOM. The nude mouse study indicated no differences in the growth of human colon tumor xenografts between acrylamide-treated and control mice, suggesting that acrylamide does not aid in the progression of established tumors. Hence, food-borne acrylamide at levels comparable to those found in human foods is neither an independent carcinogen nor a tumor promoter in the colon. However, our results characterize a potential hazard of acrylamide as a colon co-carcinogen in association with known and possibly other environmental tumor initiators/promoters.


Subject(s)
Acrylamide/adverse effects , Colonic Neoplasms/etiology , Colonic Neoplasms/pathology , Diet , Acrylamide/administration & dosage , Animals , Body Weight , Disease Models, Animal , Heterografts , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Rats , Tumor Burden
12.
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
13.
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
14.
Cardiovasc Toxicol ; 12(1): 10-24, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21822669

ABSTRACT

Epidemiological studies demonstrated that human exposure to methylmercury (MeHg) may contribute to the development and progression of metabolic and cardiovascular disorders. However, the mechanisms involved and the role of selenium (Se) and vitamin E (V(E)) supplementation in modulating MeHg cardiovascular toxicities remain unclear. This study examined the effects of Se and V(E) supplementation on MeHg-mediated systemic oxidative stress, antioxidant defense, inflammation, and endothelial dysfunction in an animal model. Male Sprague-Dawley rats were fed a starch-based casein diet or the same diet supplemented with 1 or 3 mg Se/kg diet and with or without 250 or 750 mg V(E)/kg diet. After 28 days of dietary treatment, rats were gavaged with 0 or 3 mg MeHg/kg BW for 14 consecutive days. Results suggested that exposure to MeHg may increase the risk of cardiovascular disease by decreasing circulating paraoxonase-1 activities, increasing serum oxidized low density lipoprotein levels, and associated systemic inflammation and endothelial dysfunction as reflected by increased leukocyte counts and serum levels of intercellular adhesion molecule-1 and monocyte chemotactic protein-1. Se and V(E) supplementation may either alleviate or augment the effects of MeHg, depending on their doses and combinations.


Subject(s)
Cardiovascular Diseases/blood , Cardiovascular Diseases/urine , Dietary Supplements , Methylmercury Compounds/toxicity , Selenium/physiology , Vitamin E/physiology , Animals , Antioxidants/administration & dosage , Biomarkers/blood , Biomarkers/urine , Cardiovascular Diseases/prevention & control , Male , Methylmercury Compounds/antagonists & inhibitors , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Selenium/administration & dosage , Vitamin E/administration & dosage
15.
Food Chem Toxicol ; 49(6): 1373-80, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21421017

ABSTRACT

Acrylamide, a known rodent and a probable human carcinogen, is spontaneously formed in foods cooked at high temperature. We studied the role of dietary acrylamide in modulating the early stages of colon carcinogenesis and assessed if dietary fat level was critical in altering the effects of acrylamide. Male F344 rats were subcutaneously injected with azoxymethane and were simultaneously randomized into 8 dietary groups (n=8 rats/group). Diets were based on AIN-93G semi-synthetic formula modified to contain either low fat (7% corn oil) or high fat (23.9% corn oil) and acrylamide at 0, 5, 10 or 50 mg/kg diet (wt/wt). All rats received the experimental diets ad libitum for 8 weeks, after which they were killed and their colons assessed for aberrant crypt foci (ACF), putative precancerous lesions. Irrespective of dietary fat level, rats with the highest tested dose of acrylamide (50 mg/kg diet) had significantly lower total ACF (p<0.05) and lower large ACF (those with 4 or more crypts/focus; p<0.001) compared with their respective controls (0 mg/kg diet). A significantly lower number of large ACF (p=0.046) was noted in rats treated with 10 mg/kg diet acrylamide exclusively in the high fat group, compared to the high fat control. This short-term bio-assay to test carcinogenicity of dietary acrylamide in the colon demonstrates that acrylamide, when administered through the diet at doses known to cause rat tumors, does not increase the risk of developing azoxymethane-induced precancerous lesions of the colon in rats. On the contrary, a high dose of dietary acrylamide decreased the growth of precancerous lesions in both low and high fat diet regimens in this model.


Subject(s)
Aberrant Crypt Foci/chemically induced , Acrylamide/toxicity , Carcinogens/toxicity , Colon/drug effects , Colorectal Neoplasms/chemically induced , Corn Oil/administration & dosage , Aberrant Crypt Foci/metabolism , Aberrant Crypt Foci/pathology , Acrylamide/classification , Animals , Azoxymethane/toxicity , Biomarkers, Tumor/metabolism , Carcinogens/classification , Caspase 9/metabolism , Colon/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Interactions , Male , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Inbred F344
16.
Nutr Cancer ; 63(1): 139-50, 2011.
Article in English | MEDLINE | ID: mdl-21161820

ABSTRACT

To understand the relationship between the role of soy isoflavones and estrogen receptor (ER)-ß in colon tumorigenesis, we investigated the cellular effects of soy isoflavones (composed of genistein, daidzein, and glycitein) in DLD-1 human colon adenocarcinoma cells with or without ER-ß gene silencing by RNA interference (RNAi). Soy isoflavones decreased the expression of proliferating cell nuclear antigen (PCNA), extracellular signal-regulated kinase (ERK)-1/2, AKT, and nuclear factor (NF)-κB. Soy isoflavones dose-dependently caused G2/M cell cycle arrest and downregulated the expression of cyclin A. This was associated with inhibition of cyclin dependent kinase (CDK)-4 and up-regulation of its inhibitor p21(cip1) expressions. ER-ß gene silencing lowered soy isoflavone-mediated suppression of cell viability and proliferation. ERK-1/2 and AKT expressions were unaltered and NF-κB was modestly upregulated by soy isoflavones after transient knockdown of ER-ß expression. Soy isoflavone-mediated arrest of cells at G2/M phase and upregulation of p21(cip1) expression were not observed when ER-ß gene was silenced. These findings suggest that maintaining the expression of ER-ß is crucial in mediating the growth-suppressive effects of soy isoflavones against colon tumors. Thus upregulation of ER-ß status by specific food-borne ER-ligands such as soy isoflavones could potentially be a dietary prevention or therapeutic strategy for colon cancer.


Subject(s)
Adenocarcinoma/prevention & control , Anticarcinogenic Agents/pharmacology , Colonic Neoplasms/prevention & control , Estrogen Receptor beta/physiology , Isoflavones/pharmacology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/genetics , Genistein/pharmacology , Humans , NF-kappa B/antagonists & inhibitors
17.
J Nutr ; 139(3): 474-81, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19141699

ABSTRACT

We studied the effects of lifetime exposure to dietary soy isoflavones in an azoxymethane (AOM)-induced rat colon cancer model. Male pups born to Sprague-Dawley rats exposed (including during pregnancy and lactation) to soy isoflavones at either no (0 mg = control), low (40 mg), or high (1000 mg) doses/kg diet were weaned and continued receiving their respective parental diets until the end of the study. Weaned rats received 2 subcutaneous injections (15 mg/kg body weight) of AOM 1 wk apart. After 26 wk, rats were killed and the coordinates of colon aberrant crypt foci (ACF) and tumors were determined. Expression of estrogen receptor (ER)-beta was assessed in rat colon tumors and in DLD-1 human colon adenocarcinoma cells exposed to soy isoflavones. Compared with the control, soy isoflavones did not affect incidences or multiplicities of colon ACF or tumors. Low-dose soy isoflavones decreased tumor burden and size compared with the control (P < 0.05). Expression of ERbeta increased in colon tumors of soy isoflavone-treated groups compared with the control. Soy isoflavones dose-dependently arrested the growth of DLD-1 cells and at subcytotoxic levels increased the expression of ERbeta. Our results suggest that pre- and postnatal exposure to dietary soy isoflavones suppresses the growth of colon tumors in male rats. The overexpression of ERbeta in both rat colon tumors and DLD-1 cells caused by soy isoflavones suggests that ERbeta is a critical mediator in mitigating its cancer-preventive effects.


Subject(s)
Adenocarcinoma/prevention & control , Colonic Neoplasms/prevention & control , Estrogen Receptor beta/metabolism , Glycine max/chemistry , Isoflavones/pharmacology , Adenocarcinoma/drug therapy , Animals , Animals, Newborn , Azoxymethane/toxicity , Carcinogens/toxicity , Cell Line, Tumor , Colonic Neoplasms/chemically induced , Colonic Neoplasms/drug therapy , Diet , Estrogen Receptor beta/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isoflavones/chemistry , Male , Pregnancy , Prenatal Exposure Delayed Effects , Random Allocation , Rats , Rats, Sprague-Dawley
18.
Nutr Cancer ; 61(1): 27-35, 2009.
Article in English | MEDLINE | ID: mdl-19116873

ABSTRACT

Cancer chemoprevention is a strategy taken to retard, regress, or resist the multistep process of carcinogenesis, including the blockage of its vital morphogenetic milestones viz. normal-preneoplasia-neoplasia-metastasis. For several reasons, including safety, minimal (or no) toxicity and side-effects, and better availability, alternatives such as naturally occurring phytochemicals that are found in foods are becoming increasingly popular over synthetic drugs. Food saponins have been used in complimentary and traditional medicine against a variety of diseases including several cancers. Diosgenin, a naturally occurring steroid saponin found abundantly in legumes and yams, is a well-known precursor of various synthetic steroidal drugs that are extensively used in the pharmaceutical industry. Over the past decade, a series of preclinical and mechanistic studies have been conducted to understand the role of diosgenin as a chemopreventive/therapeutic agent against several cancers. This review highlights the biological activity of diosgenin that contributes to cancer chemoprevention and control. The anticancer mode of action of diosgenin has been demonstrated via modulation of multiple cell signaling events involving critical molecular candidates associated with growth, differentiation, apoptosis, and oncogenesis. Altogether, these preclinical and mechanistic findings strongly implicate the use of diosgenin as a novel, multitarget-based chemopreventive or therapeutic agent against several cancer types. Future research in this field will help to establish not only whether diosgenin is safe and efficacious as a chemopreventive agent against several human cancers, but also to develop and evaluate standards of evidence for health claims for diosgenin-containing foods as they become increasingly popular and enter the marketplace labeled as functional foods and nutraceuticals.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Diosgenin/therapeutic use , Neoplasms/drug therapy , Neoplasms/prevention & control , Saponins/therapeutic use , Anticarcinogenic Agents/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Division/drug effects , Diosgenin/pharmacology , Humans , Saponins/pharmacology
19.
J Appl Toxicol ; 29(2): 126-40, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18821721

ABSTRACT

Weanling male Sprague-Dawley rats were administered semi-purified isocaloric diet containing soy oil (SO), seal oil (SE), docosahexaenoic acid (DHA), fish oil (FO) or lard (LA) for 28 days, and then gavaged with 0, 1 or 3 mg MeHg kg(-1) body weight per day and fed the same diet for 14 days. Serum and 24 h urine samples were collected on the day of necropsy, and analyzed for markers of kidney function and diseases. Kidney slices were analyzed for para-amino-hippurate (PAH) and tetraethylammonium (TEA) uptake, total mercury and MeHg content, and examined for pathological lesions. Total mercury and MeHg contents increased significantly and dose-dependently in all dietary groups. MeHg significantly increased relative kidney weight in all groups, serum creatinine in all except SO group, serum uric acid in the DHA and LA groups, serum Mg in all except the LA group, and urinary protein in the SO group. MeHg significantly decreased serum urea nitrogen in SE, FO and LA groups, urinary creatinine in the DHA group, PAH uptake in all except the SE group, and TEA uptake in all groups. MeHg caused nephrosis in all dietary groups. MeHg also significantly increased neutrophil counts in all except the SE group, decreased serum albumin and triglyceride in all except the DHA group, and increased serum total cholesterol in all groups, suggesting a nephrotic syndrome-like outcome. These results confirmed that kidney tubules are major targets of MeHg nephrotoxicity. Treatment with dietary fats did not prevent, but rather altered the profile of, nephrotoxicity of MeHg in rats.


Subject(s)
Dietary Fats/pharmacology , Kidney/drug effects , Methylmercury Compounds/toxicity , Animals , Blood Urea Nitrogen , Body Weight/drug effects , Calcium/blood , Cholesterol/blood , Creatinine/blood , Creatinine/urine , Diet , Dietary Fats/classification , Docosahexaenoic Acids/pharmacology , Dose-Response Relationship, Drug , Kidney/metabolism , Kidney/pathology , Magnesium/blood , Male , Metabolic Clearance Rate , Methylmercury Compounds/blood , Methylmercury Compounds/pharmacokinetics , Nephrosis/chemically induced , Nephrosis/pathology , Organ Size/drug effects , Osmolar Concentration , Quaternary Ammonium Compounds/metabolism , Rats , Rats, Sprague-Dawley , Serum Albumin/metabolism , Triglycerides/blood , Uric Acid/blood , gamma-Glutamyltransferase/urine , p-Aminohippuric Acid/metabolism
20.
J Toxicol Environ Health A ; 71(23): 1516-25, 2008.
Article in English | MEDLINE | ID: mdl-18923994

ABSTRACT

Perfluorooctanesulfonate (PFOS) is a stable and environmentally persistent metabolic or degradation product of perfluorooctanyl compounds that were manufactured for a variety of industrial and consumer applications. PFOS itself was sold for use as a surfactant. The structurally related contaminants perfluorooctanoic acid (PFOA), perfluorodecanoic acid (PFDA), and N-ethyl perfluorooctane sulfonamide (N-EtPFOSA) were shown to suppress immune responses in laboratory rodents. Relatively low doses of PFOS were found to be immunosuppressive in mice. To assess effects of PFOS on the rat immune system at doses known to alter hepatic function, changes in the morphology and function of immune tissues and cells were measured in adult rats exposed to PFOS in their diet for 28 d at levels ranging from 2 to 100 mg PFOS/kg diet (corresponding to approximately 0.14 to 7.58 mg/kg body weight [bw]/d) and compared to those receiving control diet. Body weight reductions were significant in male and female rats exposed to 50 and 100 mg PFOS/kg diet. Liver/body weight was significantly increased in females exposed to 2 mg PFOS/kg diet and in males exposed to 20 mg PFOS/kg diet. Female rats exposed to 100 mg PFOS/kg diet exhibited a significant increase in spleen weight relative to body weight; these changes lacked a histologic correlate and were not observed in males. While thymus weights relative to body weights were not affected, numbers of apoptotic lymphocytes rose in thymus with increasing dietary PFOS. There was a significant dose-related increase in total peripheral blood lymphocyte numbers in female but not male rats. In both genders the percentages of cells within lymphocyte subclasses were altered. There was a significant trend toward increasing T and T-helper (Th) cells and decreasing B cells with higher PFOS dose. Serum total immunoglobulin (Ig) G1 levels were significantly reduced in males exposed to 2 and 20 mg PFOS/kg diet. The ability of male and female rats to mount delayed-type hypersensitivity (DTH) responses to the T-cell-dependent antigen keyhole limpet hemocyanin (KLH) was not altered by PFOS. There was a significant trend toward elevated KLH-specific IgG in serum from male rats exposed to increasing levels of PFOS in diet. Splenic T- and B-cell proliferation in response to ex vivo mitogen exposure was unaffected by exposure to dietary PFOS. In conclusion, changes in immune parameters in rat did not manifest as functional alterations in response to immune challenge with KLH and may be secondary to hepatic-mediated effects of PFOS in this model.


Subject(s)
Alkanesulfonic Acids/adverse effects , Environmental Pollutants/adverse effects , Fluorocarbons/adverse effects , Food Contamination , Immunosuppressive Agents/adverse effects , Animals , Dose-Response Relationship, Drug , Humans , Immunoglobulins/drug effects , Lymphocyte Subsets/drug effects , Male , Rats , Rats, Sprague-Dawley , Sex Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...