Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
1.
Toxins (Basel) ; 16(6)2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38922153

RESUMEN

Physiologically based pharmacokinetic (PBPK) models were utilized to investigate potential interactions between aflatoxin B1 (AFB1) and efavirenz (EFV), a non-nucleoside reverse transcriptase inhibitor drug and inducer of several CYP enzymes, including CYP3A4. PBPK simulations were conducted in a North European Caucasian and Black South African population, considering different dosing scenarios. The simulations predicted the impact of EFV on AFB1 metabolism via CYP3A4 and CYP1A2. In vitro experiments using human liver microsomes (HLM) were performed to verify the PBPK predictions for both single- and multiple-dose exposures to EFV. Results showed no significant difference in the formation of AFB1 metabolites when combined with EFV (0.15 µM) compared to AFB1 alone. However, exposure to 5 µM of EFV, mimicking chronic exposure, resulted in increased CYP3A4 activity, affecting metabolite formation. While co-incubation with EFV reduced the formation of certain AFB1 metabolites, other outcomes varied and could not be fully attributed to CYP3A4 induction. Overall, this study provides evidence that EFV, and potentially other CYP1A2/CYP3A4 perpetrators, can impact AFB1 metabolism, leading to altered exposure to toxic metabolites. The results emphasize the importance of considering drug interactions when assessing the risks associated with mycotoxin exposure in individuals undergoing HIV therapy in a European and African context.


Asunto(s)
Aflatoxina B1 , Alquinos , Benzoxazinas , Ciclopropanos , Interacciones Farmacológicas , Microsomas Hepáticos , Modelos Biológicos , Inhibidores de la Transcriptasa Inversa , Aflatoxina B1/farmacocinética , Aflatoxina B1/toxicidad , Humanos , Benzoxazinas/farmacocinética , Benzoxazinas/metabolismo , Microsomas Hepáticos/metabolismo , Microsomas Hepáticos/efectos de los fármacos , Inhibidores de la Transcriptasa Inversa/farmacocinética , Masculino , Citocromo P-450 CYP3A/metabolismo , Adulto , Femenino , Citocromo P-450 CYP1A2/metabolismo , Persona de Mediana Edad , Adulto Joven , Población Blanca
2.
J Biochem Mol Toxicol ; 36(1): e22941, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34726330

RESUMEN

This study aimed to assess the protective effect of encapsulating humic acid-iron complexed nanoparticles (HA-Fe NPs) inside glucanmannan lipid particles (GMLPs) extracted from yeast cell wall against aflatoxin B (AFB1 ) toxicity in vivo. Four groups of male Sprague-Dawley rats were treated orally for 2 weeks included the control group, AFB1 treated group (80 µg/kg b.w); GMLP/HA-Fe NPs treated group (0.5 mg/kg b.w), and the group treated with AFB1 plus GMLP/HA-Fe NPs. GMLPs are empty 3-4 micron permeable microspheres that provide an efficient system for the synthesis and encapsulation of AFB1 -absorbing nanoparticles (NPs). Humic acid nanoparticles (HA-NPs) were incorporated inside the GMLP cavity by complexation with ferric chloride. In vivo study revealed that AFB1 significantly elevated serum alanine aminotransferase, aspartate aminotransferase, creatinine, uric acid, urea, cholesterol, triglycerides, LDL, malondialdehyde, and nitric oxide. It significantly decreased total protein, high-density lipoprotein, hepatic and renal CAT and glutathione peroxidase content and induced histological changes in the liver and kidney (p ≤ 0.05). The coadministration of the synthesized formulation GMLP/HA-Fe NPs with AFB1 has a protective effect against AFB1 -induced hepato-nephrotoxicity, oxidative stress and histological alterations in the liver and kidney.


Asunto(s)
Aflatoxina B1 , Polisacáridos Fúngicos , Sustancias Húmicas , Nanopartículas , Saccharomyces cerevisiae/química , beta-Glucanos , Aflatoxina B1/farmacocinética , Aflatoxina B1/toxicidad , Animales , Polisacáridos Fúngicos/química , Polisacáridos Fúngicos/farmacología , Masculino , Nanopartículas/química , Nanopartículas/uso terapéutico , Ratas , Ratas Sprague-Dawley , beta-Glucanos/química , beta-Glucanos/farmacología
3.
Arch Toxicol ; 95(6): 2163-2177, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34003344

RESUMEN

Local accumulation of xenobiotics in human and animal tissues may cause adverse effects. Large differences in their concentrations may exist between individual cell types, often due to the expression of specific uptake and export carriers. Here we established a two-photon microscopy-based technique for spatio-temporal detection of the distribution of mycotoxins in intact kidneys and livers of anesthetized mice with subcellular resolution. The mycotoxins ochratoxin A (OTA, 10 mg/kg b.w.) and aflatoxin B1 (AFB1, 1.5 mg/kg b.w.), which both show blue auto-fluorescence, were analyzed after intravenous bolus injections. Within seconds after administration, OTA was filtered by glomeruli, and enriched in distal tubular epithelial cells (dTEC). A striking feature of AFB1 toxicokinetics was its very rapid uptake from sinusoidal blood into hepatocytes (t1/2 ~ 4 min) and excretion into bile canaliculi. Interestingly, AFB1 was enriched in the nuclei of hepatocytes with zonal differences in clearance. In the cytoplasm of pericentral hepatocytes, the half-life (t1/2~ 63 min) was much longer compared to periportal hepatocytes of the same lobules (t1/2 ~ 9 min). In addition, nuclear AFB1 from periportal hepatocytes cleared faster compared to the pericentral region. These local differences in AFB1 clearance may be due to the pericentral expression of cytochrome P450 enzymes that activate AFB1 to protein- and DNA-binding metabolites. In conclusion, the present study shows that large spatio-temporal concentration differences exist within the same tissues and its analysis may provide valuable additional information to conventional toxicokinetic studies.


Asunto(s)
Aflatoxina B1/farmacocinética , Riñón/metabolismo , Hígado/metabolismo , Ocratoxinas/farmacocinética , Animales , Sistema Enzimático del Citocromo P-450/metabolismo , Semivida , Hepatocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía/métodos , Análisis Espacio-Temporal , Distribución Tisular
4.
Toxins (Basel) ; 13(3)2021 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-33805637

RESUMEN

Mycotoxins are naturally occurring toxins that can affect livestock health and performance upon consumption of contaminated feedstuffs. To mitigate the negative effects of mycotoxins, sequestering agents, adsorbents, or binders can be included to feed to interact with toxins, aiding their passage through the gastrointestinal tract (GI) and reducing their bioavailability. The parietal cell wall components of Saccharomyces cerevisiae have been found to interact in vitro with mycotoxins, such as, but not limited to, aflatoxin B1 (AFB1), and to improve animal performance when added to contaminated diets in vivo. The present study aimed to examine the pharmacokinetics of the absorption of radiolabeled AFB1 in rats in the presence of a yeast cell wall-based adsorbent (YCW) compared with that in the presence of the clay-based binder hydrated sodium calcium aluminosilicate (HSCAS). The results of the initial pharmacokinetic analysis showed that the absorption process across the GI tract was relatively slow, occurring over a matter of hours rather than minutes. The inclusion of mycotoxin binders increased the recovery of radiolabeled AFB1 in the small intestine, cecum, and colon at 5 and 10 h, revealing that they prevented AFB1 absorption compared with a control diet. Additionally, the accumulation of radiolabeled AFB1 was more significant in the blood plasma, kidney, and liver of animals fed the control diet, again showing the ability of the binders to reduce the assimilation of AFB1 into the body. The results showed the potential of YCW in reducing the absorption of AFB1 in vivo, and in protecting against the damaging effects of AFB1 contamination.


Asunto(s)
Aflatoxina B1/farmacocinética , Silicatos de Aluminio/farmacología , Pared Celular/metabolismo , Colon/efectos de los fármacos , Suplementos Dietéticos , Absorción Intestinal/efectos de los fármacos , Intestino Delgado/efectos de los fármacos , Saccharomyces cerevisiae/metabolismo , Administración Oral , Adsorción , Aflatoxina B1/administración & dosificación , Aflatoxina B1/toxicidad , Animales , Colon/metabolismo , Intestino Delgado/metabolismo , Ratas Sprague-Dawley , Distribución Tisular
5.
PLoS One ; 15(9): e0239540, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32966316

RESUMEN

Aflatoxin B1 (AFB1), a mycotoxin, is acutely hepatotoxic to many animals including humans. However, there are marked interspecies differences in sensitivity to AFB1-induced toxicity depending on bioactivation by cytochrome P450s (CYPs). In the present study, we examined the applicability of chimeric mice with humanized livers and derived fresh human hepatocytes for in vivo and vitro studies on AFB1 cytotoxicity to human hepatocytes. Chimeric mice with highly humanized livers and SCID mice received daily injections of vehicle (corn oil), AFB1 (3 mg/kg), and carbon tetrachloride (50 mg/kg) for 2 days. Histological analysis revealed that AFB1 promoted hepatocyte vacuolation and inflammatory cell infiltration in the area containing human hepatocytes. A novel human alanine aminotransferase 1 specific enzyme-linked immunosorbent assay demonstrated the acute toxicity of AFB1 to human hepatocytes in the chimeric mouse livers. The sensitivity of cultured fresh human hepatocytes isolated from the humanized liver mice for AFB1 cytotoxicity was comparable to that of primary human hepatocytes. Long-term exposure to AFB1 (6 or 14 days) produced a more severe cytotoxicity. The half-maximal lethal concentration was 10 times lower in the 2-week treatment than after 2 days of exposure. Lastly, the significant reduction of AFB1 cytotoxicity by a pan-CYP inhibitor or transfection with CYP3A4 specific siRNA clearly suggested that bioactivation of AFB1 catalyzed by CYPs was essential for AFB1 cytotoxicity to the human hepatocytes in our mouse model. Collectively, our results implicate the humanized liver mice and derived fresh human hepatocytes are useful models for studies of AFB1 cytotoxicity to human hepatocytes.


Asunto(s)
Aflatoxina B1/toxicidad , Hepatocitos/efectos de los fármacos , Activación Metabólica , Aflatoxina B1/administración & dosificación , Aflatoxina B1/farmacocinética , Animales , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Hepatocitos/patología , Hepatocitos/trasplante , Humanos , Técnicas In Vitro , Dosificación Letal Mediana , Trasplante de Hígado , Masculino , Ratones , Ratones SCID , ARN Interferente Pequeño/genética , Quimera por Trasplante , Vacuolas/efectos de los fármacos , Vacuolas/patología
6.
J Environ Sci Health B ; 55(11): 1002-1008, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32816607

RESUMEN

Adsorption of molecules to the cell walls of microorganisms plays an important role in helping to prevent animal exposure to the toxic and carcinogenic effects of aflatoxins (AFs). The aim of this study was to evaluate the ability of LAB strains, isolated from brewers' grains, to adsorb aflatoxin B1 (AFB1). All LAB were able to reduce the bioavailability of AFB1 from phosphate buffered-saline (PBS). In addition, the strains retained their effectiveness even after heat treatment. The AFB1-LAB complex stability was first evaluated through sequential washing steps. These assays demonstrated that a low percentage of AFB1 was released after consecutive washes. After subjecting the complex to different pH and bile salt treatments, the percentage of bound AF decreased, as compared to the control, but remained at high levels. Finally, to simulate the formation of the AFB1-LAB complex at conditions similar to those of the gastrointestinal tract, LAB and AFB1 were homogenized in PBS adjusted at acidic conditions or under different bile salt concentrations. In general, LAB strains showed the highest AFB1 adsorption at the lowest pH (2) and bile salt concentration (0.05%). In conclusion, the studied strains represent promising biocontrol agents for preventing and/or ameliorating the AFB1 contamination of feed.


Asunto(s)
Aflatoxina B1/metabolismo , Lactobacillales/metabolismo , Adsorción , Aflatoxina B1/química , Aflatoxina B1/farmacocinética , Ácidos y Sales Biliares , Disponibilidad Biológica , Tampones (Química) , Concentración de Iones de Hidrógeno
7.
Food Addit Contam Part B Surveill ; 13(4): 244-251, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32643592

RESUMEN

In this study Aflatoxin B1 (AFB1), ochratoxin A (OTA) and zearalenone (ZEN) occurrence in fish feed, regarding its chemical composition, were investigated. Besides, AFB1 bioaccessibility to fish was evaluated by in vitro digestion. Mycotoxins were extracted by QuEChERS and quantified by HPLC-FLD. Results showed that 93.3% of the samples were contaminated at maximum levels of 16.5, 31.6, and 322 µg/kg in the cases of AFB1, OTA, and ZEN, respectively. A positive correlation between OTA, ZEN contamination, and lipid content was observed. Risk estimation of feed consumption by fish at the highest levels of AFB1, OTA, and ZEN shows that the younger the fish, the higher the risk of exposure to mycotoxins. The AFB1 bioaccessibility assay showed that 85% of this mycotoxin may be absorbed by fish. Therefore, establishing maximum levels in the fishing sector is fundamental to contribute to feed quality and nutritional safety of fish species.


Asunto(s)
Alimentación Animal/análisis , Peces/metabolismo , Micotoxinas/análisis , Micotoxinas/farmacocinética , Aflatoxina B1/análisis , Aflatoxina B1/farmacocinética , Alimentación Animal/microbiología , Animales , Acuicultura , Carpas/metabolismo , Cíclidos/metabolismo , Contaminación de Alimentos/análisis , Ocratoxinas/análisis , Ocratoxinas/farmacocinética , Zearalenona/análisis , Zearalenona/farmacocinética
8.
Mol Nutr Food Res ; 64(13): e2000063, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32421213

RESUMEN

SCOPE: High-level exposure to aflatoxin B1 (AFB1) is known to cause acute liver damage and fatality in animals and humans. The intakes actually causing this acute toxicity have so far been estimated based on AFB1 levels in contaminated foods or biomarkers in serum. The aim of the present study is to predict the doses causing acute liver toxicity of AFB1 in rats and humans by an in vitro-in silico testing strategy. METHODS AND RESULTS: Physiologically based kinetic (PBK) models for AFB1 in rats and humans are developed. The models are used to translate in vitro concentration-response curves for cytotoxicity in primary rat and human hepatocytes to in vivo dose-response curves using reverse dosimetry. From these data, the dose levels at which toxicity would be expected are obtained and compared to toxic dose levels from available rat and human case studies on AFB1 toxicity. The results show that the in vitro-in silico testing strategy can predict dose levels causing acute toxicity of AFB1 in rats and human. CONCLUSIONS: Quantitative in vitro in vivo extrapolation (QIVIVE) using PBK modeling-based reverse dosimetry can predict AFB1 doses that cause acute liver toxicity in rats and human.


Asunto(s)
Aflatoxina B1/toxicidad , Relación Dosis-Respuesta a Droga , Hígado/efectos de los fármacos , Pruebas de Toxicidad Aguda/métodos , Aflatoxina B1/administración & dosificación , Aflatoxina B1/farmacocinética , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Hepatocitos/efectos de los fármacos , Humanos , Modelos Biológicos , Ratas , Sensibilidad y Especificidad
9.
Sci Rep ; 10(1): 5508, 2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-32218462

RESUMEN

A study was conducted to determine the cytosolic in vitro hepatic enzymatic kinetic parameters Vmax, KM, and intrinsic clearance (CLint) for aflatoxin B1 (AFB1) reductase [aflatoxicol (AFL) production] and AFL dehydrogenase (AFB1 production) in four commercial poultry species (chicken, quail, turkey and duck). Large differences were found in AFB1 reductase activity, being the chicken the most efficient producer of AFL (highest CLint value). Oxidation of AFL to AFB1 showed only slight differences among the different poultry species. On average all species produced AFB1 from AFL at a similar rate, except for the turkey which produced AFB1 from AFL at a significantly lower rate than chickens and quail, but not ducks. Although the turkey and duck showed differences in AFL oxidation Vmax and KM parameters, their CLint values did not differ significantly. The ratio AFB1 reductase/AFL dehydrogenase enzyme activity was inversely related to the known in vivo sensitivity to AFB1 being highest for the chicken, lowest for the duck and intermediate for turkeys and quail. Since there is no evidence that AFL is a toxic metabolite of AFB1, these results suggest that AFL production is a detoxication reaction in poultry. Conversion of AFB1 to AFL prevents the formation of the AFB1-8,9-exo-epoxide which, upon conversion to AFB1-dihydrodiol, is considered to be the metabolite responsible for the acute toxic effects of AFB1.


Asunto(s)
Aflatoxina B1/farmacocinética , Aflatoxina B1/toxicidad , Aflatoxinas/biosíntesis , Hígado/efectos de los fármacos , Hígado/metabolismo , Animales , Carcinógenos/farmacocinética , Carcinógenos/toxicidad , Pollos , Citosol/metabolismo , Resistencia a Medicamentos , Patos , Inactivación Metabólica , Aves de Corral , Codorniz , Pavos
10.
Toxins (Basel) ; 11(2)2019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30781814

RESUMEN

The aim of the study was to investigate the toxic effects of aflatoxin B1 (AFB1) and efficacy of a probiotic preparation containing L. reuteri, L. plantarum, L. pentosus, L. rhamnosus and L. paracasei and Saccharomyces cerevisiae yeasts to ameliorate their effects in broiler chickens. A total of 168 one-day-old female Ross 308 broilers were randomly allocated to six groups. Three wheat and soybean meal-based diets were prepared: Control diet and diets contaminated with 1 or 5 mg/kg AFB1 supplied in moldy wheat. All diets were unsupplemented or supplemented with probiotic, cold pelleted and fed from 1 to 35 day of life. Feeding diet with 1 mg AFB1/kg did not affect performance, but a diet with 5 mg AFB1 resulted in a significant reduction of feed intake and BWG, both diets induced liver and kidneys enlargement. The probiotic supplementation of the diets partially ameliorated those negative effects and resulted in a significant increase of AFB1 excretion. It was accompanied by the reduced level of AFB1 residues in the liver from 8.9 to 3.7 and from 11.8 to 5.9 µg/kg, in kidneys from 7.9 to 2.5 and from 13.7 to 4.1 µg/kg in birds fed the less and more contaminated diets, respectively. AFB1 exposure caused many severe histopathological changes in the liver and kidneys of broilers, probiotic supplementation significantly reduced the changes of these organs. It may be concluded that the probiotic supplementation can be used to alleviate the negative effects of contamination of broiler feed with AFB1 on bird health and product security.


Asunto(s)
Aflatoxina B1/toxicidad , Riñón/efectos de los fármacos , Lactobacillus , Hígado/efectos de los fármacos , Probióticos/farmacología , Sustancias Protectoras/farmacología , Saccharomyces cerevisiae , Aflatoxina B1/farmacocinética , Alimentación Animal , Animales , Pollos , Femenino , Riñón/metabolismo , Riñón/patología , Hígado/metabolismo , Hígado/patología , Masculino
11.
Toxins (Basel) ; 11(2)2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30717092

RESUMEN

Research on mycotoxins now requires a systematic study of post-exposure organisms. In this study, the effects of aflatoxin B1 (AFB1) on biofluids biomarkers were examined with metabolomics and biochemical tests. The results showed that milk concentration of aflatoxin M1 changed with the addition or removal of AFB1. AFB1 significantly affected serum concentrations of superoxide dismutase (SOD) and malon dialdehyde (MDA), SOD/MDA, and the total antioxidant capacity. Significant differences of volatile fatty acids and NH3-N were detected in the rumen fluid. Eighteen rumen fluid metabolites, 11 plasma metabolites, and 9 milk metabolites were significantly affected by the AFB1. These metabolites are mainly involved in the pathway of amino acids metabolism. Our results suggest that not only is the study of macro-indicators (milk composition and production) important, but that more attention should be paid to micro-indicators (biomarkers) when assessing the risks posed by mycotoxins to dairy cows.


Asunto(s)
Aflatoxina B1/toxicidad , Aflatoxina B1/farmacocinética , Aminoácidos/metabolismo , Alimentación Animal , Animales , Bovinos , Dieta/veterinaria , Exposición Dietética/efectos adversos , Femenino , Contaminación de Alimentos , Lactancia/efectos de los fármacos , Metabolómica , Leche/química , Plasma/metabolismo , Rumen/metabolismo
12.
J Dairy Sci ; 102(2): 1330-1340, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30594375

RESUMEN

The aim of the present study was to evaluate the effects of Lactobacillus rhamnosus GG (LGG; ATCC 53013) on growth performance and hepatotoxicity in calves fed a single dose of aflatoxin B1 (AFB1) and to investigate the absorption, distribution, and elimination of AFB1 and the hydroxylated metabolite aflatoxin M1 (AFM1) in rumen fluid, blood, and excretions. Twenty-four male Holstein calves were blocked for body weight and age and were randomly assigned to 1 of 3 treatment groups: (1) untreated control, (2) treated with 4.80 mg of AFB1 (AFB1 only), or (3) treated with 1 × 1010 cfu of LGG suspension and 4.80 mg of AFB1 (AFB1 plus LGG). The calves received LGG suspension in 50 mL of phosphate-buffered saline daily via oral administration for 14 d before and on the day they received a single oral dose of AFB1. Body weight was recorded at the beginning of the study (before LGG administration), at the day of AFB1 administration, and at the end of the trial. Rumen fluid, blood, urine, and feces samples were collected continuously for 96 h after AFB1 administration. Average daily gain (ADG) and plasma biochemical parameters were analyzed, and concentrations of AFB1 and AFM1 in the samples were determined for monitoring excretion pattern and toxicokinetics. The results showed that ADG was lower in AFB1-treated animals; LGG administration partially mitigated the decrease in ADG (0.85 ± 0.08 vs. 0.76 ± 0.18 kg of gain/d). The AFB1 treatment increased plasma aspartate aminotransferase, alkaline phosphatase, and lactate dehydrogenase levels. Administration of LGG alleviated the AFB1-induced increase in plasma enzymes activity. The excretion patterns of AFB1 and AFM1 were surprisingly regular; toxins were rapidly detected in all samples after a single oral dose of AFB1, and the peak of toxins concentrations was sequentially reached in rumen fluid, plasma, urine, and feces (except AFM1 in rumen fluid), followed by an exponential decrease. The excretion curves showed that AFB1 and AFM1 concentrations were the highest in feces and urine, respectively. Administration of LGG decreased the concentrations of free AFB1 and AFM1 in rumen fluid and reduced the release of toxins into plasma and urine. Toxicokinetic parameters (except for the time of maximum concentration and the terminal half-life) were reduced by LGG administration. In conclusion, the absorption, distribution, and excretion of AFB1 and AFM1 were rapid in calves fed a single dose of AFB1. Urine was the main route for the excretion of AFM1, and the clearance pattern from the peak of concentration was well fitted by exponential decreasing function. Administration of LGG reduced the absorption of AFB1 in the gastrointestinal tract by increasing the excretion via the feces, thus alleviating the hepatotoxic effect of AFB1.


Asunto(s)
Aflatoxina B1/toxicidad , Bovinos/microbiología , Absorción Gastrointestinal/fisiología , Lacticaseibacillus rhamnosus/fisiología , Hígado/efectos de los fármacos , Aflatoxina B1/administración & dosificación , Aflatoxina B1/farmacocinética , Aflatoxina M1/análisis , Aflatoxina M1/farmacocinética , Animales , Peso Corporal , Bovinos/crecimiento & desarrollo , Heces/química , Masculino , Leche/química , Rumen/química
13.
Artículo en Inglés | MEDLINE | ID: mdl-29132020

RESUMEN

The objective of the study was to examine adsorption of the aflatoxin B1 from synthetic gastric fluid and synthetic intestinal fluid by talc, raw and calcined diatomite. The kinetic and equilibrium adsorption processes were studied in the batch adsorption experiments applying high performance liquid chromatography for the aflatoxin B1 determination. The kinetic study showed a very fast adsorption of the aflatoxin B1 onto the selected adsorbents from the both physiological fluids with reaching equilibrium within 1-15min. The aflatoxin B1 was almost completely adsorbed in initial linear step of the kinetic process that can be described well by the zero-order kinetics model. The experimental data of the equilibrium adsorption were characterized using the Langmuir and Freundlich isotherm models. The high adsorption effectiveness was found in a range of 90%-100% and 60%-100% for the diatomite samples and the talc respectively at the initial concentrations of the aflatoxin B1 as 31-300ng/mL. The possible mechanisms of the aflatoxin adsorption onto the used mineral adsorbents are also discussed in the work.


Asunto(s)
Aflatoxina B1/aislamiento & purificación , Cromatografía Liquida/métodos , Tierra de Diatomeas/química , Talco/química , Adsorción , Aflatoxina B1/análisis , Aflatoxina B1/farmacocinética , Jugo Gástrico/química , Secreciones Intestinales/química , Cinética , Límite de Detección , Modelos Lineales , Modelos Biológicos , Reproducibilidad de los Resultados
14.
Toxins (Basel) ; 9(6)2017 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-28574433

RESUMEN

Crops contaminated with fungal mycotoxins such as aflatoxin B1 (AFB1) are often downgraded or removed from the food chain. This study aimed to evaluate the tolerance and accumulation of AFB1 in two insect species to determine whether they could be used to retain condemned mycotoxin contaminated crops in the food chain. First, instar black soldier fly larvae (Hermetia illucens, BSF) and yellow mealworm (Tenebrio molitor, YMW) were fed poultry feed spiked with AFB1 and formulated to contain levels of 0.01, 0.025, 0.05, 0.10, 0.25, and up to 0.5 mg/kg dry feed. Poultry feed without any additions and feed with only the solvent added served as controls. The AFB1 in the feed did not affect survival and body weight in the BSF and YMW larvae (p > 0.10), indicating a high tolerance to aflatoxin B1 in both species. Furthermore, AFB1 and aflatoxin M1 (AFM1) were below the detection limit (0.10 µg/kg) in BSF larvae, whereas the YMW had AFB1 levels that were approximately 10% of the European Union's legal limit for feed materials and excreted AFM1. It is concluded that both BSF larvae and YMW have a high AFB1 tolerance and do not accumulate AFB1.


Asunto(s)
Aflatoxina B1/toxicidad , Dípteros/efectos de los fármacos , Tenebrio/efectos de los fármacos , Aflatoxina B1/farmacocinética , Aflatoxina M1/metabolismo , Alimentación Animal , Animales , Dípteros/metabolismo , Larva/efectos de los fármacos , Larva/metabolismo , Tenebrio/metabolismo
15.
Nat Rev Dis Primers ; 2: 16018, 2016 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-27158749

RESUMEN

Liver cancer is the second leading cause of cancer-related deaths globally and has an incidence of approximately 850,000 new cases per year. Hepatocellular carcinoma (HCC) represents approximately 90% of all cases of primary liver cancer. The main risk factors for developing HCC are well known and include hepatitis B and C virus infection, alcohol intake and ingestion of the fungal metabolite aflatoxin B1. Additional risk factors such as non-alcoholic steatohepatitis are also emerging. Advances in the understanding of the molecular pathogenesis of HCC have led to identification of critical driver mutations; however, the most prevalent of these are not yet druggable targets. The molecular classification of HCC is not established, and the Barcelona Clinic Liver Cancer staging classification is the main clinical algorithm for the stratification of patients according to prognosis and treatment allocation. Surveillance programmes enable the detection of early-stage tumours that are amenable to curative therapies - resection, liver transplantation or local ablation. At more developed stages, only chemoembolization (for intermediate HCC) and sorafenib (for advanced HCC) have shown survival benefits. There are major unmet needs in HCC management that might be addressed through the discovery of new therapies and their combinations for use in the adjuvant setting and for intermediate- and advanced-stage disease. Moreover, biomarkers for therapy stratification, patient-tailored strategies targeting driver mutations and/or activating signalling cascades, and validated measurements of quality of life are needed. Recent failures in the testing of systemic drugs for intermediate and advanced stages have indicated a need to refine trial designs and to define novel approaches.


Asunto(s)
Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/fisiopatología , Aflatoxina B1/administración & dosificación , Aflatoxina B1/farmacocinética , Consumo de Bebidas Alcohólicas/efectos adversos , Consumo de Bebidas Alcohólicas/epidemiología , Carcinoma Hepatocelular/epidemiología , Fibrosis/etiología , Hepatitis B/complicaciones , Vacunas contra Hepatitis B/farmacología , Vacunas contra Hepatitis B/uso terapéutico , Hepatitis C/complicaciones , Humanos , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Estilo de Vida , Metformina/farmacología , Metformina/uso terapéutico , Factores de Riesgo
16.
Transgenic Res ; 24(3): 489-96, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25603989

RESUMEN

The aflatoxin-detoxifizyme (ADTZ) gene derived from Armillariella tabescens was cloned into parotid gland-specific expression vector (pPSPBGPneo) to construct the parotid gland-specific vector expressing ADTZ (pPSPBGPneo-ADTZ). Transgenic mice were generated by microinjection and identified by using PCR and Southern blotting analysis. PCR and Southern blotting analysis showed that total six transgenic mice carried the ADTZ gene were generated. RT-PCR analysis indicated that the expression of ADTZ mRNA could be detected only in parotid glands of the transgenic mice. The ADTZ activity in the saliva was found to be 3.72 ± 1.64 U/mL. After feeding a diet containing aflatoxin B1 (AFB1) for 14 days, the effect of ADTZ on serum biochemical indexes and AFB1 residues in serum and liver of mice were evaluated. The results showed that total protein and globulin contents in the test treatment (transgenic mice) produced ADTZ were significantly higher than that of the positive control, while alanine aminotransferase and aspartate aminotransferase activity in serum of the test treatment (transgenic mice) were remarkably lower compared to that of the positive control (P < 0.05). Moreover, AFB1 residues in serum and liver of the test treatment (transgenic mice) were significantly lower compared with that of the positive control (P < 0.05). These results in the study confirmed that ADTZ produced in transgenic mice could reduce, even eliminate the negative effects of AFB1 on mice.


Asunto(s)
Inactivación Metabólica/genética , Complejos Multienzimáticos/genética , Glándula Parótida/fisiología , Aflatoxina B1/sangre , Aflatoxina B1/metabolismo , Aflatoxina B1/farmacocinética , Animales , Femenino , Hígado/metabolismo , Masculino , Ratones Transgénicos , Complejos Multienzimáticos/metabolismo , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Saliva/fisiología , Porcinos/genética
17.
Toxicol Sci ; 139(2): 293-300, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24675090

RESUMEN

Mice are resistant to aflatoxin hepatotoxicity, primarily due to high expression of glutathione S-transferases (GSTs), and in particular the GSTA3 subunit. Nuclear factor erythroid 2 related factor 2 (Nrf2) signaling, which controls a broad-based cytoprotective response, was activated either genetically or pharmacologically in an attempt to rescue GSTA3 knockout mice from aflatoxin genotoxicity. Genetic activation of Nrf2 signaling was attained in a GSTA3: hepatocyte-specific Keap1 double knockout (DKO) mouse whereas pharmacologic activation of Nrf2 was achieved through pretreatment of mice with the triterpenoid 1-[2-cyano-3-,12-dioxoleana-1,9(11)-dien-28-oyl] imidazole (CDDO-Im) prior to aflatoxin B1 exposure. Following oral treatment with aflatoxin, urine was collected from mice for 24 h and hepatic and urinary aflatoxin metabolites then quantified using isotope dilution-mass spectrometry. Although Nrf2 was successfully activated genetically and pharmacologically, neither means affected the response of GSTA3 knockout mice to chemical insult with aflatoxin. Hepatic aflatoxin B1-N(7)-guanine levels were elevated 120-fold in GSTA3 knockout mice compared with wild-type and levels were not attenuated by the interventions. This lack of effect was mirrored in the urinary excretion of aflatoxin B1-N(7)-guanine. By contrast, urinary excretion of aflatoxin B1-N-acetylcysteine was >200-fold higher in wild-type mice compared with the single GSTA3 knockout or DKO mouse. The inability to rescue GSTA3 knockout mice from aflatoxin genotoxicity through the Nrf2 transcriptional program indicates that Gsta3 is unilaterally responsible for the detoxication of aflatoxin in mice.


Asunto(s)
Aflatoxina B1/toxicidad , Glutatión Transferasa/genética , Hipertensión/tratamiento farmacológico , Imidazoles/farmacología , Mutágenos/toxicidad , Factor 2 Relacionado con NF-E2 , Ácido Oleanólico/análogos & derivados , Transducción de Señal/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Aflatoxina B1/farmacocinética , Aflatoxina B1/orina , Animales , Proteínas del Citoesqueleto/genética , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Hepatocitos/metabolismo , Hipertensión/genética , Hipertensión/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Mutágenos/farmacocinética , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Ácido Oleanólico/farmacología , Transducción de Señal/genética
18.
Crit Rev Toxicol ; 44(4): 348-91, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24494825

RESUMEN

The framework analysis previously presented for using DNA adduct information in the risk assessment of chemical carcinogens was applied in a series of case studies which place the adduct information into context with the key events in carcinogenesis to determine whether they could be used to support a mutagenic mode of action (MOA) for the examined chemicals. Three data-rich chemicals, aflatoxin B1 (AFB1), tamoxifen (Tam) and vinyl chloride (VCl) were selected for this exercise. These chemicals were selected because they are known human carcinogens and have different characteristics: AFB1 forms a unique adduct and human exposure is through contaminated foods; Tam is a pharmaceutical given to women so that the dose and duration of exposure are known, forms unique adducts in rodents, and has both estrogenic and genotoxic properties; and VCl, to which there is industrial exposure, forms a number of adducts that are identical to endogenous adducts found in unexposed people. All three chemicals produce liver tumors in rats. AFB1 and VCl also produce liver tumors in humans, but Tam induces human uterine tumors, only. To support a mutagenic MOA, the chemical-induced adducts must be characterized, shown to be pro-mutagenic, be present in the tumor target tissue, and produce mutations of the class found in the tumor. The adducts formed by AFB1 and VCl support a mutagenic MOA for their carcinogenicity. However, the data available for Tam shows a mutagenic MOA for liver tumors in rats, but its carcinogenicity in humans is most likely via a different MOA.


Asunto(s)
Aflatoxina B1/toxicidad , Aductos de ADN , Mutágenos/toxicidad , Medición de Riesgo/métodos , Tamoxifeno/toxicidad , Cloruro de Vinilo/toxicidad , Aflatoxina B1/farmacocinética , Animales , Carcinógenos/toxicidad , Aductos de ADN/análisis , Aductos de ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Neoplasias Hepáticas Experimentales/inducido químicamente , Mutación , Ratas , Tamoxifeno/farmacocinética , Distribución Tisular , Cloruro de Vinilo/farmacocinética
19.
Int J Cancer ; 134(7): 1539-48, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24114584

RESUMEN

Cytochrome P450 2A13 (CYP2A13), mainly expressed in human respiratory tract, is highly efficient in the metabolic activation of aflatoxin (AF) B1 (AFB1) and is assumed to play a role in human lung tumorigenesis in airborne AFB1 exposure. To validate the assumption, we exposed human bronchial epithelial (BEAS-2B) cells stably expressing CYP2A13 (B-2A13), CYP1A2 (B-1A2) and CYP2A6 (B-2A6) to 0.1-10 nM AFB1 for 30-50 passages. B-2A13 cells showed increased sensitivity to 0.1 nM AFB1-induced neoplastic transformation and the formation of tumors in nude mice were observed at passage 30 (P30) while it occurred at P50 B-1A2 cells. B-2A6, similar to vector control, showed no neoplastic transformation in this condition. Additionally, AFB1-DNA adducts and 8-OHdG significantly increased in transformed P40 B-2A13, in parallel with the upregulation of p-ATR, p-BRCA1, Mre11, Rad50 and Rad51. However, the apoptosis of P40 cells was near normal, while the expression of Bax, C-Caspase 3 and C-PARP increased passage-dependently. Inhibition of ATR (ATR siRNA or NU6027) reversely increased the apoptosis of P40 B-2A13 cells in parallel with the upregulation of Bax, C-Caspase 3 and C-PARP, suggesting that ATR plays an important role in maintaining cell survival via antiapoptosis. Additionally, activation of ATR was necessary to neoplastic transformation since blockage of ATR in P40 cells inhibited DNA damage repair response and anchorage-independent growth. Our data demonstrated that CYP2A13 played a critical role in AFB1-induced neoplastic transformation. ATR-mediated the dysfunction of apoptosis and DNA damage repair might be involved. These results help establish a linkage between airborne AFB1 and human respiratory carcinoma.


Asunto(s)
Aflatoxina B1/toxicidad , Hidrocarburo de Aril Hidroxilasas/genética , Bronquios/efectos de los fármacos , Bronquios/enzimología , Transformación Celular Neoplásica/efectos de los fármacos , Aflatoxina B1/farmacocinética , Animales , Apoptosis/efectos de los fármacos , Hidrocarburo de Aril Hidroxilasas/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/genética , Bronquios/patología , Caspasa 3/genética , Línea Celular , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Sistema Enzimático del Citocromo P-450/genética , Aductos de ADN/efectos de los fármacos , Reparación del ADN , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Células Epiteliales/patología , Humanos , Ratones , Ratones Desnudos , Poli(ADP-Ribosa) Polimerasas/genética , Regulación hacia Arriba/efectos de los fármacos , Proteína X Asociada a bcl-2/genética
20.
J Appl Toxicol ; 34(1): 40-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23047854

RESUMEN

Food shortages and a lack of food supply regulation in developing countries often leads to chronic exposure of vulnerable populations to hazardous mixtures of mycotoxins, including aflatoxin B(1) (AFB(1)) and fumonisin B(1) (FB(1)). A refined calcium montmorillonite clay [i.e. uniform particle size NovaSil (UPSN)] has been reported to tightly bind these toxins, thereby decreasing bioavailability in humans and animals. Hence, our objectives in the present study were to examine the ability of UPSN to bind mixtures of AFB(1) and FB(1) at gastrointestinally relevant pH in vitro, and to utilize a rapid in vivo bioassay to evaluate AFB(1) and FB(1) toxicity and UPSN efficacy. Isothermal sorption data indicated tight AFB(1) binding to UPSN surfaces at both pH 2.0 and 6.5, but substantially more FB(1) bound at pH 2.0 than 6.5. Site-specific competition occurred between the toxins when exposed to UPSN in combination. Importantly, treatment with UPSN resulted in significant protection to mycotoxin-exposed hydra maintained at pH 6.9-7.0. Hydra were exposed to FB(1), AFB(1) and FB(1) /AFB(1) combinations with and without UPSN. A toxic response over 92 h was rated based on morphology and mortality. Hydra assay results indicated a minimum effective concentration (MEC) of 20 µg ml(-1) for AFB(1), whereas the MEC for FB(1) was not reached. The MEC for co-exposure was 400 µg ml(-1) FB(1) + 10 µg ml(-1) AFB(1). This study demonstrates that UPSN sorbs both mycotoxins tightly at physiologically relevant pH levels, resulting in decreased bioavailability, and that a modified hydra bioassay can be used as an initial screen in vivo to predict efficacy of toxin-binding agents.


Asunto(s)
Aflatoxina B1/toxicidad , Silicatos de Aluminio/química , Fumonisinas/toxicidad , Hydra/efectos de los fármacos , Pruebas de Toxicidad/métodos , Aflatoxina B1/farmacocinética , Animales , Arcilla , Fumonisinas/farmacocinética , Hydra/crecimiento & desarrollo , Concentración de Iones de Hidrógeno
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...