Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 214
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(2)2021 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-33450918

RESUMEN

The multimycotoxin-degrading efficiency of the Rhodococcus erythropolis NI1 strain was investigated with a previously developed three-step method. NI1 bacterial metabolites, single and combined mycotoxins and their NI1 degradation products, were injected into one cell stage zebrafish embryos in the same doses. Toxic and interaction effects were supplemented with UHPLC-MS/MS measurement of toxin concentrations. Results showed that the NI1 strain was able to degrade mycotoxins and their mixtures in different proportions, where a higher ratio of mycotoxins were reduced in combination than single ones. The NI1 strain reduced the toxic effects of mycotoxins and mixtures, except for the AFB1+T-2 mixture. Degradation products of the AFB1+T-2 mixture by the NI1 strain were more toxic than the initial AFB1+T-2 mixture, while the analytical results showed very high degradation, which means that the NI1 strain degraded this mixture to toxic degradation products. The NI1 strain was able to detoxify the AFB1, ZEN, T-2 toxins and mixtures (except for AFB1+T-2 mixture) during the degradation experiments, which means that the NI1 strain degraded these to non-toxic degradation products. The results demonstrate that single exposures of mycotoxins were very toxic. The combined exposure of mycotoxins had synergistic effects, except for ZEN+T-2 and AFB1+ZEN +T-2, whose mixtures had very strong antagonistic effects.


Asunto(s)
Micotoxinas/metabolismo , Rhodococcus/metabolismo , Pruebas de Toxicidad , Pez Cebra , Aflatoxina B1/metabolismo , Aflatoxina B1/farmacología , Aflatoxina B1/toxicidad , Animales , Bacterias/metabolismo , Relación Dosis-Respuesta a Droga , Dosificación Letal Mediana , Microinyecciones , Micotoxinas/toxicidad , Pruebas de Toxicidad/métodos , Zearalenona/metabolismo
2.
Reproduction ; 160(5): 709-723, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33065550

RESUMEN

This study aims to evaluate the deleterious effect of the mycotoxin aflatoxin B1 (AFB1) on bull spermatozoa and the carryver effect on the developing embryo. Proteomic analysis of AFB1-treated spermatozoa revealed differential expression of proteins associated with biological processes and cellular pathways that involved in spermatozoon function, fertilization competence and embryonic development. Therefore, we assume that factors delivered by the spermatozoa, regardless of DNA fragmentation, are also involved. To confirm this hypothesis, we have used the annexin V (AV) kit to separate the spermatozoa into apoptotic (AV+) and non-apoptotic (AV-) subpopulations which were found to correlate with high- and low DNA fragmentation, respectively. Fertilization with AV+ AFB1-treated spermatozoa, resulted in no blastocyst formation, whereas fertilization with AV- spermatozoa resulted in reduced cleavage rate and formation of genetically altered blastocysts (POU5F1 and SOX2). Microarray analysis of blastocysts derived from 10 µM AFB1-treated spermatozoa revealed differential expression of 345 genes that involved in cellular pathways such as embryo and placenta development, cell cycle, DNA repair and histone modification, and in signaling pathways, especially calcium signaling pathway. This is the first report on deleterious carrying over effects of AFB1 from the bovine spermatozoa to the formed embryo. Our findings suggest that aside from the damage caused by AFB1 to spermatozoa's DNA integrity, additional damage mechanisms are involved.


Asunto(s)
Aflatoxina B1/farmacología , Blastocisto/metabolismo , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteoma/efectos de los fármacos , Espermatozoides/metabolismo , Transcriptoma/efectos de los fármacos , Animales , Blastocisto/efectos de los fármacos , Bovinos , Embrión de Mamíferos/efectos de los fármacos , Femenino , Masculino , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Venenos/farmacología , Embarazo , Espermatozoides/efectos de los fármacos
3.
Int J Mol Sci ; 21(18)2020 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-32899983

RESUMEN

Aflatoxin B1 (AFB1) is a mycotoxin widely distributed in a variety of food commodities and exhibits strong toxicity toward multiple tissues and organs. However, little is known about its neurotoxicity and the associated mechanism. In this study, we observed that brain integrity was markedly damaged in mice after intragastric administration of AFB1 (300 µg/kg/day for 30 days). The toxicity of AFB1 on neuronal cells and the underlying mechanisms were then investigated in the neuroblastoma cell line IMR-32. A cell viability assay showed that the IC50 values of AFB1 on IMR-32 cells were 6.18 µg/mL and 5.87 µg/mL after treatment for 24 h and 48 h, respectively. ROS levels in IMR-32 cells increased significantly in a time- and AFB1 concentration-dependent manner, which was associated with the upregulation of NOX2, and downregulation of OXR1, SOD1, and SOD2. Substantial DNA damage associated with the downregulation of PARP1, BRCA2, and RAD51 was also observed. Furthermore, AFB1 significantly induced S-phase arrest, which is associated with the upregulation of CDKN1A, CDKN2C, and CDKN2D. Finally, AFB1 induced apoptosis involving CASP3 and BAX. Taken together, AFB1 manifests a wide range of cytotoxicity on neuronal cells including ROS accumulation, DNA damage, S-phase arrest, and apoptosis-all of which are key factors for understanding the neurotoxicology of AFB1.


Asunto(s)
Aflatoxina B1/toxicidad , Apoptosis/efectos de los fármacos , Daño del ADN , Síndromes de Neurotoxicidad , Especies Reactivas de Oxígeno/metabolismo , Fase S/efectos de los fármacos , Aflatoxina B1/farmacología , Animales , Apoptosis/fisiología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Daño del ADN/fisiología , Masculino , Ratones , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Fase S/genética
4.
Anal Chem ; 91(10): 6600-6607, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31026147

RESUMEN

During apoptosis process, the release of cytochrome c (Cyt c) is considered to be a key factor in the intrinsic pathway and is often defined as no regression point. Quantitative detection of intracellular Cyt c remains a challenge. Herein, we have developed surface-enhanced Raman scattering (SERS)-fluorescence dual-mode nanosensors for the quantitative assay of Cyt c in living cells. Dual signal detection was achieved by constructing gold nanotriangles (AuNTs) nanosensors capable of specifically recognizing Cyt c. The nanosensors were prepared by modifying the aptamer of Cyt c on AuNTs and connecting the complementary strands modified with Cy5. The AuNTs provided both enhanced SERS signals and fluorescence quenching effects. Once cells were induced by external stimulus (such as toxins) to release Cyt c, Cyt c would specifically bind to its aptamer, and the complementary strands modified with Cy5 would detach which would result in weakened SERS signal and recovery of fluorescence signal. The experimental results showed that the nanosensors not only had excellent selectivity and sensitivity but also realized real-time monitoring of Cyt c translocation event from mitochondria to cytoplasm. The SERS and fluorescence intensity showed good linear relationship with Cyt c concentration ranging from 0.044 to 9.95 µM and achieved a minimum limit of detection (LOD) of 0.02 µM in living cells. The accuracy of intracellular Cyt c quantitative results was more than 90% compared with the ELISA results.


Asunto(s)
Aptámeros de Nucleótidos/química , Carbocianinas/química , Citocromos c/análisis , Colorantes Fluorescentes/química , Nanopartículas del Metal/química , Aflatoxina B1/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Citocromos c/química , ADN Complementario/química , Transferencia Resonante de Energía de Fluorescencia/métodos , Oro/química , Humanos , Límite de Detección , Espectrometría Raman/métodos
5.
J Pineal Res ; 66(4): e12543, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30584671

RESUMEN

Aflatoxin B1 (AFB1) is a major food and feed contaminant that threaten public health. Previous studies indicate that AFB1 exposure disrupted oocyte maturation. However, an effective and feasible method is unavailable for protecting oocytes against toxicity of AFB1. In the present study, using in vitro matured porcine oocytes and parthenogenetic embryos as model, we confirmed that AFB1 exposure during in vitro oocyte maturation (IVM) significantly impaired both nuclear and cytoplasmic maturation in a dose- and time-dependent manner. The different concentrations of melatonin were also tested for their protective effects on oocytes against the AFB1-induced toxicity. Our results showed that supplementation of a relative high concentration of melatonin (10-3 mol/L) during IVM efficiently reversed the impaired development rate and blastocyst quality, to the levels comparable to those of the control group. Further analysis indicated that melatonin application efficiently alleviated reactive oxygen species accumulation and initiation of apoptosis induced by AFB1 exposure. In addition, disrupted GSH/GPX system, as well as inhibited mitochondrial DNA (mtDNA) replication and mitochondrial biogenesis in AFB1-treated oocytes, can be notably reversed by melatonin application. Furthermore, cumulus cells may be important in mediating the toxicity of AFB1 to oocytes, and the metabolism of AFB1 in cumulus cells can be depressed by melatonin. To the best of our knowledge, this is the first report to confirm that melatonin application can efficiently protect oocytes from AFB1-induced toxicity. Our study provides a promising and practical strategy for alleviating or reversing AFB1-induced female reproductive toxicity in both clinical treatment and domestic reproductive management.


Asunto(s)
Aflatoxina B1/farmacología , Técnicas de Maduración In Vitro de los Oocitos , Melatonina/farmacología , Oocitos/citología , Oocitos/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Células del Cúmulo/citología , Células del Cúmulo/efectos de los fármacos , Células del Cúmulo/metabolismo , Variaciones en el Número de Copia de ADN/genética , Variaciones en el Número de Copia de ADN/fisiología , ADN Mitocondrial/efectos de los fármacos , Femenino , Glutatión/metabolismo , Etiquetado Corte-Fin in Situ , Oocitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Porcinos
6.
J Nat Prod ; 82(6): 1694-1703, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-31136174

RESUMEN

Aflatoxin B1 (AfB1) ranks among the most potent liver carcinogens known, and the accidental or intentional exposure of humans and livestock to this toxin remains a serious global threat. One protective measure that had been proposed is employing small-molecule therapeutics capable of mitigating the toxicity of AfB1; however, to date, these efforts have had little clinical success. To identify molecular scaffolds that reduce the toxicity of AfB1, we developed a cell-based high-throughput high-content imaging assay that enabled our team to test natural products (pure compounds, fractions, and extracts) for protection of monolayers and spheroids composed of HepG2 liver cells against AfB1. The spheroid assay showed notable potential for further development, as it afforded greater sensitivity of HepG2 cells to AfB1, which is believed to better mimic the in vivo response of hepatocytes to the toxin. One of the most bioactive compounds to arise from this investigation was alternariol-9-methyl ether (1, purified from an Alternaria sp. isolate), which inspired the synthesis and testing of several structurally related molecules. Based on these findings, it is proposed that several types of natural and synthetic polyarene molecules that have undergone oxidative functionalization (e.g., compounds containing 3-methoxyphenol moieties) are promising starting points for the development of new agents that protect against AfB1 toxicity.


Asunto(s)
Aflatoxina B1/farmacología , Aflatoxina B1/toxicidad , Antineoplásicos Fitogénicos/farmacología , Carcinógenos/toxicidad , Hepatocitos/efectos de los fármacos , Hígado/efectos de los fármacos , Sustancias Protectoras/farmacología , Aflatoxina B1/química , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/aislamiento & purificación , Productos Biológicos/farmacología , Carcinógenos/química , Hepatocitos/química , Humanos , Hígado/química , Estructura Molecular , Sustancias Protectoras/química
7.
Cell Physiol Biochem ; 49(3): 1110-1126, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30196302

RESUMEN

BACKGROUND/AIMS: Swine influenza virus (SIV) is a major pathogen of both animals and humans. Afatoxin B1 (AFB1) is one of the most common mycotoxins in feed and food. However, the central contribution of AFB1 to SIV infection remains unclear. METHODS: Here, TCID50 assays, fluorescence-based quantitative real-time PCR, western blotting, immunofluorescence staining, histopathological examination, flow cytometry and scanning electron microscopy were performed to investigate the involvement and underlying mechanism of AFB1 in SIV infection in vivo and in vitro using mouse models and porcine alveolar macrophage (PAM) models, respectively. RESULTS: The in vivo study showed that low levels of AFB1 promoted SIV infection and increased its severity, as demonstrated by the increased mRNA expression of viral matrix protein (M); by the increased protein expression of nucleoprotein (NP), matrix protein 1 and ion channel protein; and by animal weight loss, lung index and lung histologic damage. In addition, the increased occurrence of SIV infection accompanied by increases in the level of IL-10 in sera and lungs, in the spleen index and in the number of CD206-positive mouse alveolar macrophages but decreases in the level of TNF-α in sera and lungs, in the thymus index and in the number of CD80-positive mouse alveolar macrophages was observed in SIV-infected mice after low-level AFB1 exposure. The in vitro study showed that low concentrations of AFB1 promoted SIV infection, as demonstrated by the increases in viral titers and viral M mRNA and NP expression levels in SIV-infected PAMs as well as by the number of cells positive for NP protein expression. Furthermore, AFB1 promoted the polarization of SIV-infected PAMs to the M1 phenotype at 8 hpi and to the M2 phenotype at 24 hpi, as measured by the increases in IL-10 expression and in the number of CD206-positive PAMs as well as by the morphological changes observed by scanning electron microscopy. The administration of the immune stimulant lipopolysaccharide (LPS) reversed the switch in PAM polarization from M2 to M1 and thereby counteracted the promotion of influenza virus infection induced by AFB1. CONCLUSION: Our results are the first to confirm that low-level exposure to AFB1 promotes SIV infection and modulates a switch in macrophage polarization from M1 to M2. The work reported here provides important data that point to a role for AFB1 in SIV infection, and it opens a new field of study.


Asunto(s)
Aflatoxina B1/farmacología , Macrófagos Alveolares/efectos de los fármacos , Infecciones por Orthomyxoviridae/patología , Animales , Líquido del Lavado Bronquioalveolar/citología , Línea Celular , Proliferación Celular/efectos de los fármacos , Virus de la Influenza A/aislamiento & purificación , Virus de la Influenza A/metabolismo , Interleucina-10/análisis , Lectinas Tipo C/metabolismo , Lipopolisacáridos/farmacología , Pulmón/metabolismo , Pulmón/patología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Masculino , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteínas de la Nucleocápside , Infecciones por Orthomyxoviridae/veterinaria , Infecciones por Orthomyxoviridae/virología , Fenotipo , ARN Viral/análisis , Proteínas de Unión al ARN/metabolismo , Receptores de Superficie Celular/metabolismo , Porcinos , Factor de Necrosis Tumoral alfa/análisis , Proteínas del Núcleo Viral/metabolismo
8.
Fish Shellfish Immunol ; 80: 480-486, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29782917

RESUMEN

Aflatoxins, which was produced by Aspergillus flavus or Aspergillus parasiticus fungi during grain and feed processing or storage, could cause severe health problems and reduction of yield during shrimp cultures. To evaluate toxic effects of aflatoxin B1 (AFB1) in juvenile Pacific white shrimp (Litopenaeus vannamei) and potential protective effect of Zn(II)-curcumin (Zn-CM), four experimental diets (control, 500 µg/kg AFB1, 500 µg/kg AFB1+100 mg/kg Zn-CM, 500 µg/kg AFB1+200 mg/kg Zn-CM) were formulated in quadruplicate to feed the shrimp for 8 weeks. The results revealed that AFB1 could induce significant decrease in final body weight (FBW), weight gain (WG, %) and visible variations of the hepatopancreas structures in L.vannamei. Compared with AFB1 group, AFB1+100 mg/kg Zn-CM group significantly ameliorated the toxic effects of AFB1 on growth performance, while AFB1+100 mg/kg Zn-CM group had no effect on growth performance. Dietary AFB1+100 mg/kg Zn-CM enhanced phenoloxidase (PO) (P < 0.05) activity. Both dietary AFB1+100 mg/kg Zn-CM and AFB1+200 mg/kg Zn-CM reduced inducible nitric oxide synthase (iNOS) activity and glutathione (GSH) level, decreased the content of malondialdehyde (MDA) (P < 0.05) in hepatopancreas compared with AFB1 group. Transmission electron microscopy (TEM) analysis demonstrated that Zn-CM could relieve the microvilli transformation and mitochondria accumulation reduction caused by AFB1. Consequently, the results demonstrated that suitable Zn-CM could mitigate the AFB1-induced hepatotoxicity and immunotoxicity effects on L.vannamei.


Asunto(s)
Aflatoxina B1/farmacología , Curcumina/farmacología , Penaeidae/efectos de los fármacos , Sustancias Protectoras/farmacología , Zinc/farmacología , Aflatoxina B1/toxicidad , Alanina Transaminasa/metabolismo , Alimentación Animal , Animales , Glutatión/metabolismo , Hepatopáncreas/efectos de los fármacos , Hepatopáncreas/metabolismo , Hepatopáncreas/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Penaeidae/crecimiento & desarrollo , Penaeidae/inmunología , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
9.
J Dairy Sci ; 101(4): 3008-3020, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29428756

RESUMEN

The study was conducted to examine the effect of supplementing bentonite clay with or without a Saccharomyces cerevisiae fermentation product (SCFP; 19 g of NutriTek + 16 g of MetaShield, both from Diamond V, Cedar Rapids, IA) on the performance and health of dairy cows challenged with aflatoxin B1 (AFB1). Twenty-four lactating Holstein cows (64 ± 11 d in milk) were stratified by parity and milk production and randomly assigned to 1 of 4 treatment sequences. The experiment had a balanced 4 × 4 Latin square design with 6 replicate squares, four 33-d periods, and a 5-d washout interval between periods. Cows were fed a total mixed ration containing 36.1% corn silage, 8.3% alfalfa hay, and 55.6% concentrate (dry matter basis). Treatments were (1) control (no additives), (2) toxin (T; 1,725 µg of AFB1/head per day), (3) T + clay (CL; 200 g/head per day; top-dressed), and (4) CL+SCFP (CL+SCFP; 35 g/head per day; top-dressed). Cows were adapted to diets from d 1 to 25 (predosing period) and then orally dosed with AFB1 from d 26 to 30 (dosing period), and AFB1 was withdrawn from d 31 to 33 (withdrawal period). Milk samples were collected twice daily from d 21 to 33, and plasma was sampled on d 25 and 30 before the morning feeding. Transfer of ingested AFB1 into milk aflatoxin M1 (AFM1) was greater in T than in CL or CL+SCFP (1.65 vs. 1.01 and 0.94%, respectively) from d 26 to 30. The CL and CL+SCFP treatments reduced milk AFM1 concentration compared with T (0.45 and 0.40 vs. 0.75 µg/kg, respectively), and, unlike T, both CL and CL+SCFP lowered AFM1 concentrations below the US Food and Drug Administration action level (0.5 µg/kg). Milk yield tended to be greater during the dosing period in cows fed CL+SCFP compared with T (39.7 vs. 37.7 kg/d). Compared with that for T, plasma glutamic oxaloacetic transaminase concentration, indicative of aflatoxicosis and liver damage, was reduced by CL (85.9 vs. 95.2 U/L) and numerically reduced by CL+SCFP (87.9 vs. 95.2 U/L). Dietary CL and CL+SCFP reduced transfer of dietary AFB1 to milk and milk AFM1 concentration. Only CL prevented the increase in glutamic oxaloacetic transaminase concentration, and only CL+SCFP prevented the decrease in milk yield caused by AFB1 ingestion.


Asunto(s)
Aflatoxina B1/farmacología , Silicatos de Aluminio/metabolismo , Bentonita/metabolismo , Bovinos/metabolismo , Leche/química , Saccharomyces cerevisiae/química , Silicatos de Aluminio/administración & dosificación , Alimentación Animal/análisis , Animales , Bentonita/administración & dosificación , Bovinos/inmunología , Arcilla , Dieta/veterinaria , Suplementos Dietéticos/análisis , Femenino , Fermentación , Estado de Salud , Lactancia , Distribución Aleatoria
10.
J Anim Physiol Anim Nutr (Berl) ; 102(4): 947-952, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29604131

RESUMEN

The purpose of this study was to investigate the short-term (48 hr) effects of feeding aflatoxin contaminated diet (170.3 µg/kg AFB1) in 49-week-old laying hens. Liver samples were taken at 12-hr intervals. Feed intake, body weight, absolute and relative liver weight were the same in groups. However, there was no feed intake during both dark periods (between 12nd to 24th and 36th to 48th hours of the experiment); therefore, aflatoxin intake was also negligible. Markers of initial phase of lipid peroxidation, conjugated dienes and trienes did not change as effect of aflatoxin, but terminal marker, malondialdehyde content was significantly higher at 12 hr as effect of aflatoxin. No significant difference was found in reduced glutathione concentration and glutathione peroxidase activity between the groups. Expression of glutathione peroxidase 4 gene (GPX4) was significantly reduced due to aflatoxin treatment at 12 and 24 hr, but induced later, while glutathione reductase gene (GSR) expression was significantly lower at 24 hr and glutathione synthetase gene (GSS) in aflatoxin-treated group at 12 hr. The results suggest that aflatoxin induced oxygen-free radical formation, but it did not reach critical level during this short period of time to cause activation of the expression of glutathione system.


Asunto(s)
Aflatoxina B1/farmacología , Pollos/metabolismo , Glutatión/metabolismo , Peroxidación de Lípido , Hígado/efectos de los fármacos , Animales , Femenino , Hígado/metabolismo , Oxidación-Reducción
11.
J Anim Physiol Anim Nutr (Berl) ; 102(4): 953-961, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29638014

RESUMEN

This study aimed to investigate the effect of lactic acid bacteria (LAB) and smectite on the growth performance, nutrient digestibility and blood parameters of broilers that were fed diets contaminated with aflatoxin B1 (AFB1 ). A total of 480 newly hatched male Arbor Acres broilers were randomly allocated into four groups with six replicates of 20 chicks each. The broilers were fed diets with the AFB1 (40 µg/kg) challenge or without (control) it and supplemented with smectite (3.0 g/kg) or LAB (4.0 × 1010  CFU/kg) based on the AFB1 diet. The trial lasted for 42 days. The results showed that during days 1-42 of AFB1 challenge, the feed intake (FI) and body weight gain (BWG) were depressed (p < .05). The inclusion of LAB and smectite increased (p < .05) the BWG by 71.58 and 41.89 g/bird, respectively, which reached the level of the control diet (p ≥ .05), but there were no differences (p ≥ .05) in performance between LAB and smectite. LAB and smectite also increased (p < .05) the apparent total tract digestibility of the crude protein. Regarding the blood parameters, AFB1 decreased (p < .05) the levels of red blood cell count, haematocrit, mean corpuscular volume, haemoglobin, albumin and total protein. In the meantime, the AFB1 increased (p < .05) leucocyte counts, urea nitrogen, cholesterol, total bilirubin, creatinine, glutamic-pyruvic transaminase, glutamic oxaloacetic transaminase and alkaline phosphatase. By contrast, LAB and smectite affected (p < .05) these parameters in the opposite direction. It can be concluded that after the AFB1 challenge, LAB and smectite have similar effects on the growth and health of the broilers, suggesting that LAB could be an alternative against AFB1 in commercial animal feeds.


Asunto(s)
Pollos/crecimiento & desarrollo , Dieta/veterinaria , Lactobacillales/fisiología , Silicatos/administración & dosificación , Aflatoxina B1/farmacología , Alimentación Animal , Animales , Pollos/metabolismo , Digestión/efectos de los fármacos , Digestión/fisiología , Masculino , Aumento de Peso
12.
Neurochem Res ; 42(10): 2743-2754, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28462452

RESUMEN

This study aimed to investigate the potential neurotoxic effects of aflatoxin B1 (AFB1) and the preventive effects of saffron. Male Balb-c mice received AFB1 (0.6 mg/kg/day intraperitoneally for 4 days), saffron infusion (90 mg styles/200 mL, ad libitum access for 2 weeks) or saffron infusion plus AFB1 (saffron treatment as previously plus 0.6 mg AFB1/kg/day intraperitoneally for the last 4 days). Control mice were intraperitoneally injected with DMSO:saline (1:1, v/v) during AFB1 treatment. Learning/memory was assessed by passive avoidance task. The activity of acetylcholinesterase [AChE, salt-(SS)/detergent-soluble(DS) isoforms], butyrylcholinesterase (BuChE, SS/DS isoforms), monoamine oxidase (MAO-A, MAO-B), the levels of lipid peroxidation (malondialdehyde, MDA) and reduced glutathione (GSH), were determined in whole brain (minus cerebellum) and cerebellum. We demonstrate for the first time that AFB1 administration impaired the memory of adult mice and decreased significantly whole brain AChE and BuChE activity, cerebellar AChE activity and cerebral GSH content. Moreover, MAO isoforms activity in whole brain, MAO-B activity in cerebellum and MDA levels of both tissues were significantly higher after AFB1 treatment. Pre-treatment with saffron prevented memory decline, activation of MAO-A and MAO-B in whole brain and cerebellum, respectively, and lipid peroxidation triggered by AFB1. Interestingly, the activity of AChE isoforms in whole brain, DS-AChE in cerebellum and GSH levels of both tissues were further significantly decreased in saffron +AFB1-treated mice compared with AFB1 group. Our findings support the neuroprotective efficacy of saffron against AFB1 in adult mice.


Asunto(s)
Encéfalo/efectos de los fármacos , Cognición/efectos de los fármacos , Aprendizaje/efectos de los fármacos , Memoria/efectos de los fármacos , Extractos Vegetales/farmacología , Aflatoxina B1/farmacología , Animales , Antioxidantes/farmacología , Encéfalo/metabolismo , Aprendizaje/fisiología , Peroxidación de Lípido/efectos de los fármacos , Masculino , Ratones Endogámicos BALB C , Estrés Oxidativo/efectos de los fármacos ,
13.
Cell Biol Toxicol ; 32(5): 403-17, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27287938

RESUMEN

Stem cell-induced hepatocytes (SC-iHeps) have been suggested as a valuable model for evaluating drug toxicology. Here, human-induced pluripotent stem cells (QIA7) and embryonic stem cells (WA01) were differentiated into hepatocytes, and the hepatotoxic effects of acetaminophen (AAP) and aflatoxin B1 (AFB1) were compared with primary hepatocytes (p-Heps) and HepG2. In a cytotoxicity assay, the IC50 of SC-iHeps was similar to that in p-Heps and HepG2 in the AAP groups but different from that in p-Heps of the AFB1 groups. In a multi-parameter assay, phenotypic changes in mitochondrial membrane potential, calcium influx and oxidative stress were similar between QIA7-iHeps and p-Heps following AAP and AFB1 treatment but relatively low in WA01-iHeps and HepG2. Most hepatic functional markers (hepatocyte-specific genes, albumin/urea secretion, and the CYP450 enzyme activity) were decreased in a dose-dependent manner following AAP and AFB1 treatment in SC-iHeps and p-Heps but not in HepG2. Regarding CYP450 inhibition, the cell viability of SC-iHeps and p-Heps was increased by ketoconazole, a CYP3A4 inhibitor. Collectively, SC-iHeps and p-Heps showed similar cytotoxicity and hepatocyte functional effects for AAP and AFB1 compared with HepG2. Therefore, SC-iHeps have phenotypic characteristics and sensitivity to cytotoxic chemicals that are more similar to p-Heps than to HepG2 cells.


Asunto(s)
Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Acetaminofén/farmacología , Aflatoxina B1/farmacología , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Sistema Enzimático del Citocromo P-450/metabolismo , Citotoxinas/farmacología , Evaluación Preclínica de Medicamentos/métodos , Células Hep G2 , Humanos , Hígado/citología , Hígado/efectos de los fármacos , Cultivo Primario de Células , Pruebas de Toxicidad/métodos
14.
J Sci Food Agric ; 96(12): 4009-14, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26694215

RESUMEN

BACKGROUND: Aflatoxin B1 (AFB1 ) is often detected in peanut oil, which comes from contaminated peanuts. AFB1 in peanut oil seriously threatens the health of consumers. However, there are few methods to effectively remove AFB1 in peanut oil. This study aimed to use an alkali-refining method to degrade AFB1 in peanut oil efficiently without increasing the equipment of oil and fat refining. RESULTS: The optimum detoxifying conditions of AFB1 in peanut oil with alkali refining were established using response surface methodology (RSM), and the safety of peanut oil after being refined with alkali was evaluated based on the Ames tests and HepG2 cell viability. The results showed that AFB1 in peanut oil was decreased from 34.78 to 0.37 µg kg(-1) (98.94% reduction) under the optimum detoxifying conditions, i.e. when the initial temperature of alkali refining was 43.51 °C, the amount of excess alkali was 0.30%, the content of alkali solution was 23.42% and the end temperature of alkali refining was 77.07 °C. The acid value and color of peanut oil refined by alkali were improved significantly, while the peroxide value was increased within an acceptable level. The safety of peanut oil contaminated by AFB1 was improved significantly after being refined with alkali. CONCLUSION: These results indicate that alkali refining is an effective method for removing AFB1 in peanut oil. The optimum detoxifying conditions of AFB1 in peanut oil with alkali refining could be used to guide the production of oil companies for ensuring food safety. © 2015 Society of Chemical Industry.


Asunto(s)
Aflatoxina B1/química , Arachis/microbiología , Análisis de Peligros y Puntos de Control Críticos , Aflatoxina B1/farmacología , Aflatoxina B1/toxicidad , Supervivencia Celular/efectos de los fármacos , Descontaminación , Manipulación de Alimentos , Microbiología de Alimentos , Células Hep G2/efectos de los fármacos , Humanos
15.
Immunopharmacol Immunotoxicol ; 36(1): 1-10, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24168324

RESUMEN

CONTEXT: Aflatoxins (AFs) are highly hazardous mycotoxins with potent carcinogenic, mutagenic and immune disregulatory properties. Cytochrome P450 (CYP) isoforms are central for enhanced AFB1 toxicity in situ. It remains to be seen whether and how these AFB1 activators work in human leukocytes. OBJECTIVE: To investigate the involvement of CYP isoforms in AFB1 toxicity of circulating mononuclear cells, we examined the impact of environmentally relevant levels of AFB1 on lymphocytes and monocytes. MATERIALS AND METHODS: Very low and moderate doses of AFB1 with/without CYP inducers on transcription of key CYP isoforms and toll-like receptor 4 (TLR4) were examined in human lymphocytes, monocytes and HepG2 cells; cell cycle distribution and viability were also analyzed in AFB1-exposed lymphocytes and monocytes. RESULTS: Only CYP1A1, CYP1B1, CYP3A4, CYP3A5 and CYP3A7 expressed in lymphocytes and monocytes. TLR4 much more expressed in monocytes than in lymphocytes, but HepG2 showed little TLR4 transcription. While CYP1A1, CYP1B1 and CYP3A4 were highly induced by AFB1 in monocytes, in lymphocytes only CYP1A1 was induced. Among CYP1A1, CYP1B1 and CYP3A4 only CYP1A1 responded to low and moderate levels of AFB1. Enhanced transcripts of CYPs by AFB1 yielded little synergies on TLR4 transcription in lymphocytes and monocytes. Cell cycle arrest and necrosis were also detected in AFB1-exposed lymphocytes and monocytes. CONCLUSIONS: Our novel findings indicate that AFB1 more intensively stimulates CYP genes expression in monocytes than in lymphocytes. Mechanistically, this could explain a more pronounced immunotoxicity of AFB1 in myeloid than in lymphoid lineage cells in vitro/situ/vivo.


Asunto(s)
Aflatoxina B1/farmacología , Sistema Enzimático del Citocromo P-450/biosíntesis , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Linfocitos/enzimología , Monocitos/enzimología , Venenos/farmacología , Regulación hacia Arriba/efectos de los fármacos , Adolescente , Adulto , Células Hep G2 , Humanos , Isoenzimas/biosíntesis , Linfocitos/citología , Masculino , Monocitos/citología , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/metabolismo
16.
Acta Histochem ; 126(1): 152116, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38101290

RESUMEN

INTRODUCTION: The combined pathogenesis of Aflatoxin B1 (AFB1) and several viruses such as HBV, EBV and influenza virus have been investigated yet the molecular mechanism of their interaction and possible synergistic effects is not fully understood. OBJECTIVES: The aim of the current systematic review was to review in-vitro and in-vivo studies investigating the combined pathogenesis of aflatoxins and viruses. METHODS: This systematic review was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines. PECO (Population, Exposure, Comparator, and Outcome) criteria for invitro and invivo studies were used to evaluate the eligibility of the studies for systematic review. RESULTS: 21 studies were eligible for qualitative analysis based on the inclusion criteria. Of all the included studies, 9 (42.9 %) were invivo, 7 (33.3 %) were invitro-invivo and 5(23.8) articles conducted only invitro assay. Furthermore 14 (66.6 %) article explored hepatitis B virus (HBV) combination with AFB1, 4 (19 %) studied influenza A virus (SIV), 2 (9.7 %) were about Epstein-Barr virus (EBV) and only 1 (4.7 %) included hepatitis C virus (HCV). CONCLUSION: The limited collected evidence suggests that AFB1 enhanced EBV and influenza virus pathogenesis. AFB1 also operated as a cofactor for HBV and EBV-mediated carcinogenesis. On the other hand HBV and HCV also induced AFB-1 carcinogenesis. Due to the limited amount of included studies and the inconsistency of their results further studies especially on HBV and SIV are essential for better understanding of their combined mechanisms.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Hepatitis C , Humanos , Aflatoxina B1/farmacología , Herpesvirus Humano 4 , Virus de la Hepatitis B/genética , Carcinogénesis
17.
Biochemistry ; 52(42): 7486-99, 2013 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-24040835

RESUMEN

Cholinergic synaptic transmission often requires extremely rapid hydrolysis of acetylcholine by acetylcholinesterase (AChE). AChE is inactivated by organophosphates (OPs) in chemical warfare nerve agents. The resulting accumulation of acetylcholine disrupts cholinergic synaptic transmission and can lead to death. A potential long-term strategy for preventing AChE inactivation by OPs is based on evidence that OPs must pass through a peripheral site or P-site near the mouth of the AChE active site gorge before reacting with a catalytic serine in an acylation site or A-site at the base of the gorge. An ultimate goal of this strategy is to design compounds that bind tightly at or near the P-site and exclude OPs from the active site while interfering minimally with the passage of acetylcholine. However, to target the AChE P-site with ligands and potential drugs that selectively restrict access, much more information must be gathered about the structure-activity relationships of ligands that bind specifically to the P-site. We apply here an inhibitor competition assay that can correctly determine whether an AChE inhibitor binds to the P-site, the A-site, or both sites. We have used this assay to examine three uncharged, natural product inhibitors of AChE, including aflatoxin B1, dihydrotanshinone I, and territrem B. The first two of these inhibitors are predicted by the competition assay to bind selectively to the P-site, while territrem B is predicted to span both the P- and A-sites. These predictions have recently been confirmed by X-ray crystallography. Dihydrotanshinone I, with an observed binding constant (KI) of 750 nM, provides a good lead compound for the development of high-affinity, uncharged inhibitors with specificity for the P-site.


Asunto(s)
Acetilcolinesterasa/química , Acetilcolinesterasa/metabolismo , Inhibidores de la Colinesterasa/farmacología , Fenantrenos/farmacología , Acetilcolina/metabolismo , Acetilcolinesterasa/genética , Aflatoxina B1/farmacología , Sitios de Unión , Unión Competitiva , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Furanos , Humanos , Hidrólisis , Cinética , Modelos Químicos , Piranos/farmacología , Quinonas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Especificidad por Sustrato
18.
J Proteome Res ; 12(7): 3434-48, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23742107

RESUMEN

Glucose-6-phosphate dehydrogenase (G6PD) is pivotal to reduced nicotinamide adenine dinucleotide phosphate (NADPH) production and cellular redox balance. Cells with G6PD deficiency are susceptible to oxidant-induced death at high oxidative stress. However, it remains unclear what precise biological processes are affected by G6PD deficiency due to altered cellular redox homeostasis, particularly at low oxidative stress. To further explore the biological role of G6PD, we generated G6PD-knockdown cell clones using lung cancer line A549. We identified proteins differentially expressed in the knockdown clones without the addition of exogenous oxidant by means of isobaric tags for relative and absolute quantification (iTRAQ) labeling coupled with multidimensional liquid chromatography-mass spectrometry (LC-MS/MS). We validated a panel of proteins that showed altered expression in G6PD-knockdown clones and were involved in metabolism of xenobiotic and glutathione (GSH) as well as energy metabolism. To determine the physiological relevancy of our findings, we investigated the functional consequence of G6PD depletion in cells treated with a prevalent xenobiotic, aflatoxin B1(AFB1). We found a protective role of G6PD in AFB1-induced cytotoxicity, possibly via providing NADPH for NADPH oxidase to induce epoxide hydrolase 1 (EPHX1), a xenobiotic-metabolizing enzyme. Collectively, our findings reveal for the first time a proteome-wide dysregulation by G6PD depletion under the condition without exogenous oxidant challenge, and we suggest a novel association of G6PD activity with AFB1-related xenobiotic metabolism.


Asunto(s)
Aflatoxina B1/farmacología , Glucosafosfato Deshidrogenasa/metabolismo , Estrés Oxidativo/genética , Sustancias Protectoras/farmacología , Proteoma/metabolismo , Línea Celular Tumoral , Metabolismo Energético , Técnicas de Silenciamiento del Gen , Glucosafosfato Deshidrogenasa/genética , Glutatión/metabolismo , Homeostasis , Humanos , NADP/metabolismo , Oxidación-Reducción , Xenobióticos/metabolismo
19.
Nutr Cancer ; 65(5): 775-80, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23859045

RESUMEN

The development of multidrug resistance (MDR) causes problems in the chemotherapy of human cancer. The present study was designed to evaluate and establish the role of Eclipta alba as MDR reversal agent using multidrug resistant hepatocellular carcinoma cell line (DR-HepG2). To develop DR-HepG2, hepatocellular carcinoma cell line (HepG2) was transfected with 2-Acetylaminofluorene (AAF) and Aflatoxin B1 (AFB). Cytotoxic effects of the Eclipta alba hydroalcoholic extract (EAE) and standard anti-ancer drug Doxorubicin (DOX) were determined in DR-HepG2 and the parental cells HepG2 using MTT assay. The expression level of MDR1 gene and P-glycoprotein (P-gp) level was analyzed by RT-PCR and western blotting. From the present investigation, it was found that EAE (10 and 20 µg/ml) could significantly inhibit cell proliferation in DR-HepG2 whereas DOX (0.5 µg/ml) could not because of enhancement effect of MDR1/P-gp. This study demonstrated for the first time the antiproliferative activities of EAE in multidrug resistant DR-HepG2 cells. The findings revealed that Eclipta alba components are effective inhibitors of MDR1/P-gp.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Eclipta/química , Neoplasias/metabolismo , Extractos Vegetales/farmacología , 2-Acetilaminofluoreno/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Aflatoxina B1/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Doxorrubicina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Humanos , Extracción Líquido-Líquido , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias/genética , Transfección , Xenobióticos/farmacología
20.
J Nat Prod ; 76(10): 1958-65, 2013 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-24156291

RESUMEN

Bilirubin exhibits antioxidant and antimutagenic effects in vitro. Additional tetrapyrroles that are naturally abundant were tested for antigenotoxicity in Salmonella. Un-/conjugated bilirubin (1 and 2), biliverdin (4), bilirubin and biliverdin dimethyl esters (3 and 5), stercobilin (6), urobilin (7), and protoporphyrin (8) were evaluated at physiological concentrations (0.01-2 µmol/plate; 3.5-714 µM) against the metabolically activated food-borne mutagens aflatoxin B1 (9) and 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (10). Compound 8 most effectively inhibited the mutagenic effects of 9 in strain TA102 and 10 in TA98. Compound 7 inhibited 9-induced mutagenesis in strain TA98 most effectively, while 1 and 4 were promutagenic in this strain. This is likely due to their competition with mutagens for phase-II detoxification. Mechanistic investigations into antimutagenesis demonstrate that tetrapyrroles react efficiently with a model epoxide of 9, styrene epoxide (11), to form covalent adducts. This reaction is significantly faster than that of 11 with guanine. Hence, the evaluated tetrapyrroles inhibited genotoxicity induced by poly-/heterocyclic amines found in foods, and novel evidence obtained in the present investigation suggests this may occur via chemical scavenging of genotoxic metabolites of the mutagens investigated. This may have important ramifications for maintaining health, especially with regard to cancer prevention.


Asunto(s)
Antimutagênicos/farmacología , Antioxidantes/farmacología , Bilirrubina/farmacología , Biliverdina/farmacología , Mutagénesis/efectos de los fármacos , Mutágenos/farmacología , Tetrapirroles/farmacología , Aflatoxina B1/química , Aflatoxina B1/farmacología , Antimutagênicos/química , Antioxidantes/química , Bilirrubina/química , Biliverdina/química , Alimentos , Humanos , Modelos Biológicos , Estructura Molecular , Pruebas de Mutagenicidad , Mutágenos/química , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/genética , Tetrapirroles/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA