Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Immunogenetics ; 67(3): 163-78, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25597949

RESUMEN

Poultry are highly susceptible to the immunotoxic effects of the food-borne mycotoxin aflatoxin B1 (AFB1). Exposure impairs cell-mediated and humoral immunity, limits vaccine efficacy, and increases the incidence of costly secondary infections. We investigated the molecular mechanisms of AFB1 immunotoxicity and the ability of a Lactobacillus-based probiotic to protect against aflatoxicosis in the domestic turkey (Meleagris gallopavo). The spleen transcriptome was examined by RNA sequencing (RNA-seq) of 12 individuals representing four treatment groups. Sequences (6.9 Gb) were de novo assembled to produce over 270,000 predicted transcripts and transcript fragments. Differential expression analysis identified 982 transcripts with statistical significance in at least one comparison between treatment groups. Transcripts with known immune functions comprised 27.6 % of significant expression changes in the AFB1-exposed group. Short exposure to AFB1 suppressed innate immune transcripts, especially from antimicrobial genes, but increased the expression of transcripts from E3 ubiquitin-protein ligase CBL-B and multiple interleukin-2 response genes. Up-regulation of transcripts from lymphotactin, granzyme A, and perforin 1 could indicate either increased cytotoxic potential or activation-induced cell death in the spleen during aflatoxicosis. Supplementation with probiotics was found to ameliorate AFB1-induced expression changes for multiple transcripts from antimicrobial and IL-2-response genes. However, probiotics had an overall suppressive effect on immune-related transcripts.


Asunto(s)
Aflatoxina B1/toxicidad , Proteínas Aviares/genética , Enfermedades de las Aves/genética , Intoxicación por Setas/veterinaria , Probióticos/administración & dosificación , Transcriptoma/efectos de los fármacos , Animales , Proteínas Aviares/inmunología , Enfermedades de las Aves/inmunología , Perfilación de la Expresión Génica , Granzimas/genética , Granzimas/inmunología , Secuenciación de Nucleótidos de Alto Rendimiento , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/genética , Inmunomodulación/efectos de los fármacos , Interleucina-2/genética , Interleucina-2/inmunología , Linfocinas/genética , Linfocinas/inmunología , Anotación de Secuencia Molecular , Intoxicación por Setas/genética , Intoxicación por Setas/inmunología , Perforina/genética , Perforina/inmunología , Sialoglicoproteínas/genética , Sialoglicoproteínas/inmunología , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/metabolismo , Transcriptoma/inmunología , Pavos , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología
2.
PLoS Biol ; 8(9)2010 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-20838655

RESUMEN

A synergistic combination of two next-generation sequencing platforms with a detailed comparative BAC physical contig map provided a cost-effective assembly of the genome sequence of the domestic turkey (Meleagris gallopavo). Heterozygosity of the sequenced source genome allowed discovery of more than 600,000 high quality single nucleotide variants. Despite this heterozygosity, the current genome assembly (∼1.1 Gb) includes 917 Mb of sequence assigned to specific turkey chromosomes. Annotation identified nearly 16,000 genes, with 15,093 recognized as protein coding and 611 as non-coding RNA genes. Comparative analysis of the turkey, chicken, and zebra finch genomes, and comparing avian to mammalian species, supports the characteristic stability of avian genomes and identifies genes unique to the avian lineage. Clear differences are seen in number and variety of genes of the avian immune system where expansions and novel genes are less frequent than examples of gene loss. The turkey genome sequence provides resources to further understand the evolution of vertebrate genomes and genetic variation underlying economically important quantitative traits in poultry. This integrated approach may be a model for providing both gene and chromosome level assemblies of other species with agricultural, ecological, and evolutionary interest.


Asunto(s)
Genoma , Pavos/genética , Animales , Secuencia de Bases , Mapeo Cromosómico , ADN/genética , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN , Homología de Secuencia de Ácido Nucleico , Especificidad de la Especie
3.
Arch Toxicol ; 86(1): 121-35, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21818627

RESUMEN

The normally picturesque Cache Valley in northern Utah is frequently reported to have the worst particulate (PM) air pollution in the United States. Numerous epidemiological studies conducted elsewhere have associated PM exposure to a variety of cardiovascular diseases and early mortality. We have previously shown that Cache Valley PM (CVPM) is pro-inflammatory, through a variety of mechanisms involving the release of inflammatory cytokines, unfolded protein response, ER stress, and C-reactive protein (CRP). This study was undertaken to determine whether Cache Valley PM (CVPM) would activate Akt, an upstream mechanism common to these events. Human lung (BEAS-2B) cells were treated with either fine (PM(2.5)) or coarse (PM(10)) particles (12.5 and 25 µg/ml) for periods up to 24 h. PM-exposed cells exhibited Akt activation as evidenced by phosphorylation at Thr(308) and Ser(473). Events downstream of Akt activation such as NF-κB activation were observed at 1 and 24 h, but IκB phosphorylation occurred only at 24 h, indicating that mechanisms of PM-mediated NF-κB activation are time dependent. Akt and NF-κB related inflammatory cytokine IL-1α, and IL-6 and the chemokine IL-8 were upregulated in treated cells at 6 and 24 h. The calpain inhibitor leupeptin limited Akt phosphorylation to Ser(473) and reduced release of IL-1α, IL-6, and IL-8, indicating that calpain or similar protease(s) are involved in PM-induced activation of Akt and subsequent release of inflammatory cytokines. Our data indicate that PM activates Akt, which may play a role in the pro-inflammatory response to PM exposure.


Asunto(s)
Inflamación/inducido químicamente , Pulmón/efectos de los fármacos , Material Particulado/toxicidad , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Línea Celular , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Inflamación/patología , Interleucina-1alfa/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Pulmón/citología , Pulmón/metabolismo , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Tamaño de la Partícula , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos , Salud Urbana , Utah
4.
Toxicol Appl Pharmacol ; 254(3): 349-54, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21616088

RESUMEN

The extreme sensitivity of turkeys to aflatoxin B(1) (AFB(1)) is associated with efficient epoxidation by hepatic cytochromes P450 (P450) 1A5 and 3A37 to exo-aflatoxin B(1)-8,9-epoxide (exo-AFBO). The combined presence of 1A5 and 3A37, which obey different kinetic models, both of which metabolize AFB(1) to the exo-AFBO and to detoxification products aflatoxin M(1) (AFM(1)) and aflatoxin Q(1) (AFQ(1)), respectively, complicates the kinetic analysis of AFB(1) in turkey liver microsomes (TLMs). Antisera directed against 1A5 and 3A37, thereby individually removing the catalytic contribution of these enzymes, were used to identify the P450 responsible for epoxidating AFB(1) in TLMs. In control TLMs, AFB(1) was converted to exo-AFBO in addition to AFM(1) and AFQ(1) confirming the presence of functional 1A5 and 3A37. Pretreatment with anti-1A5 inhibited exo-AFBO formation, especially at low, submicromolar (~0.1µM), while anti-3A37, resulted in inhibition of exo-AFBO formation, but at higher (>50µM) AFB(1) concentrations. Metabolism in immunoinhibited TLMs resembled that of individual enzymes: 1A5 produced exo-AFBO and AFM(1), conforming to Michaelis-Menten, while 3A37 produced exo-AFBO and AFQ(1) following the kinetic Hill equation. At 0.1µM AFB(1), close to concentrations in livers of exposed animals, 1A5 contributed to 98% of the total exo-AFBO formation. At this concentration, 1A5 accounted for a higher activation:detoxification (50:1, exo-AFBO: AFM(1)) compared to 3A37 (0.15: 1, exo-AFBO: AFQ(1)), suggesting that 1A5 is high, while 3A4 is the low affinity enzyme in turkey liver. The data support the conclusion that P450 1A5 is the dominant enzyme responsible for AFB(1) bioactivation and metabolism at environmentally-relevant AFB(1) concentrations in turkey liver.


Asunto(s)
Aflatoxina B1/metabolismo , Hidrocarburo de Aril Hidroxilasas/fisiología , Microsomas Hepáticos/metabolismo , Animales , Familia 3 del Citocromo P450 , Microsomas Hepáticos/enzimología , Pavos
5.
Chem Res Toxicol ; 23(8): 1322-9, 2010 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-20707407

RESUMEN

Cytochrome P450s (P450) play an important role in the formation of carcinogenic and mutagenic electrophilic intermediates from a wide range of xenobiotics, including naturally occurring dietary compounds. The pathogenesis of hepatotoxic and hepatocarcinogenic action of the mycotoxin aflatoxin B(1) (AFB(1)) involves initial bioactivation by P450s to a reactive and electrophilic intermediate exo-aflatoxin B(1)-8,9-epoxide (exo-AFBO). Poultry, especially turkeys are extremely sensitive to AFB(1), a condition due, in part, to efficient epoxidation by P450 1A and 3A enzymes. We previously reported the discovery of P450 1A5 from turkey liver, which like its human homologue, 1A2, bioactivated AFB(1) to exo-AFBO and aflatoxin M(1) (AFM(1)). Here, we describe P450 3A37, the 3A4 homologue from turkey liver. This gene has an open reading frame (ORF) of 1512 bp, and the protein is predicted to be 504 amino acids with 97% identity to chicken P450 3A37. A truncated construct of the turkey P450 3A37 gene with 11 amino acids deleted from the hydrophobic N-terminal region was heterologously expressed in Escherichia coli, the protein from which exhibited a CO difference spectrum typical of P450s. Like human P450 3A4, 3A37 biotransformed AFB(1) to exo-AFBO and aflatoxin Q(1) (AFQ(1)) and possessed nifedipine oxidation activity, both of which were inhibited by the P450 3A4 inhibitor 17alpha-ethynylestradiol. Oxidation of AFB(1) to exo-AFBO and AFQ(1) by P450 3A37 followed sigmoidal Hill kinetics, suggestive of an allosteric interaction between the enzyme and AFB(1). The Hill coefficient (n) value was 1.9 for exo-AFBO and 1.6 for AFQ(1), indicative of positive cooperativity. The calculated K(m) and V(max) values for the formation of exo-AFBO were 287 +/- 21 muM and 1.45 +/- 0.07 nmol/min/nmol P450, respectively, whereas those of AFQ(1) formation were 302 +/- 51 muM and 7.86 +/- 0.75 nmol/min/nmol P450, respectively. These data strongly suggest that P450 3A37, along with P450 1A5, plays an important role in AFB(1) epoxidation in turkey liver.


Asunto(s)
Aflatoxina B1/metabolismo , Hidrocarburo de Aril Hidroxilasas/genética , Hidrocarburo de Aril Hidroxilasas/metabolismo , Hígado/metabolismo , Pavos , Secuencia de Aminoácidos , Animales , Hidrocarburo de Aril Hidroxilasas/antagonistas & inhibidores , Clonación Molecular , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Inhibidores del Citocromo P-450 CYP3A , Etinilestradiol/farmacología , Perfilación de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad de la Especie
6.
Toxins (Basel) ; 11(5)2019 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-31035349

RESUMEN

Susceptibility and/or resistance to aflatoxin B1 (AFB1) is a threshold trait governed principally by glutathione S transferase (GST)-mediated detoxification. In poultry, domesticated turkeys are highly sensitive to AFB1, most likely due to dysfunction in hepatic GSTs. In contrast, wild turkeys are comparatively resistant to aflatoxicosis due to the presence of functional hepatic GSTAs and other possible physiological and immunological interactions. The underlying genetic basis for the disparate GST function in turkeys is unknown as are the broader molecular interactions that control the systemic response. This study quantifies the effects of dietary AFB1 on gene expression in the turkey spleen, specifically contrasting genetically distinct domesticated (DT, susceptible) and Eastern wild (EW, resistant) birds. Male turkey poults were subjected to a short-term AFB1 treatment protocol with feed supplemented with 320 ppb AFB1 beginning on day 15 of age and continuing for 14 days. Spleen tissues were harvested and subjected to deep RNA sequencing and transcriptome analysis. Analysis of differential gene expression found the effects of AFB1 treatment on the spleen transcriptomes considerably more prominent in the DT birds compared to EW. However, expression of the differentially expressed genes (DEGs) was directionally biased, with the majority showing higher expression in EW (i.e., down-regulation in DT). Significantly altered pathways included FXR/RXR and LXR/RXR activation, coagulation system, prothrombin activation, acute phase response, and atherosclerosis signaling. Differential extra-hepatic expression of acute phase protein genes was confirmed by quantitative real time PCR (qRT-PCR) in the original experiment and additional turkey lines. Results demonstrate that wild turkeys possess a capacity to more effectively respond to AFB1 exposure.


Asunto(s)
Aflatoxina B1/toxicidad , Bazo/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Alimentación Animal , Animales , Animales Domésticos , Animales Salvajes , Resistencia a la Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Micotoxicosis/genética , Bazo/metabolismo , Pavos
7.
Toxins (Basel) ; 11(1)2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30669283

RESUMEN

The nearly-ubiquitous food and feed-borne mycotoxin aflatoxin B1 (AFB1) is carcinogenic and mutagenic, posing a food safety threat to humans and animals. One of the most susceptible animal species known and thus a good model for characterizing toxicological pathways, is the domesticated turkey (DT), a condition likely due, at least in part, to deficient hepatic AFB1-detoxifying alpha-class glutathione S-transferases (GSTAs). Conversely, wild turkeys (Eastern wild, EW) are relatively resistant to the hepatotoxic, hepatocarcinogenic and immunosuppressive effects of AFB1 owing to functional gene expression and presence of functional hepatic GSTAs. This study was designed to compare the responses in gene expression in the gastrointestinal tract between DT (susceptible phenotype) and EW (resistant phenotype) following dietary AFB1 challenge (320 ppb for 14 days); specifically in cecal tonsil which functions in both nutrient absorption and gut immunity. RNAseq and gene expression analysis revealed significant differential gene expression in AFB1-treated animals compared to control-fed domestic and wild birds and in within-treatment comparisons between bird types. Significantly upregulated expression of the primary hepatic AFB1-activating P450 (CYP1A5) as well as transcriptional changes in tight junction proteins were observed in AFB1-treated birds. Numerous pro-inflammatory cytokines, TGF-ß and EGF were significantly down regulated by AFB1 treatment in DT birds and pathway analysis suggested suppression of enteroendocrine cells. Conversely, AFB1 treatment modified significantly fewer unique genes in EW birds; among these were genes involved in lipid synthesis and metabolism and immune response. This is the first investigation of the effects of AFB1 on the turkey gastro-intestinal tract. Results suggest that in addition to the hepatic transcriptome, animal resistance to this mycotoxin occurs in organ systems outside the liver, specifically as a refractory gastrointestinal tract.


Asunto(s)
Aflatoxina B1/toxicidad , Animales Domésticos/genética , Tracto Gastrointestinal/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Pavos/genética , Animales , Tracto Gastrointestinal/metabolismo , Glutatión Transferasa/genética , Isoenzimas/genética , Masculino
8.
Toxins (Basel) ; 10(1)2018 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-29342849

RESUMEN

The food-borne mycotoxin aflatoxin B1 (AFB1) poses a significant risk to poultry, which are highly susceptible to its hepatotoxic effects. Domesticated turkeys (Meleagris gallopavo) are especially sensitive, whereas wild turkeys (M. g. silvestris) are more resistant. AFB1 toxicity entails bioactivation by hepatic cytochrome P450s to the electrophilic exo-AFB1-8,9-epoxide (AFBO). Domesticated turkeys lack functional hepatic GST-mediated detoxification of AFBO, and this is largely responsible for the differences in resistance between turkey types. This study was designed to characterize transcriptional changes induced in turkey livers by AFB1, and to contrast the response of domesticated (susceptible) and wild (more resistant) birds. Gene expression responses to AFB1 were examined using RNA-sequencing. Statistically significant differences in gene expression were observed among treatment groups and between turkey types. Expression analysis identified 4621 genes with significant differential expression (DE) in AFB1-treated birds compared to controls. Characterization of DE transcripts revealed genes dis-regulated in response to toxic insult with significant association of Phase I and Phase II genes and others important in cellular regulation, modulation of apoptosis, and inflammatory responses. Constitutive expression of GSTA3 was significantly higher in wild birds and was significantly higher in AFB1-treated birds when compared to controls for both genetic groups. This pattern was also observed by qRT-PCR in other wild and domesticated turkey strains. Results of this study emphasize the differential response of these genetically distinct birds, and identify genes and pathways that are differentially altered in aflatoxicosis.


Asunto(s)
Aflatoxina B1/toxicidad , Hígado/efectos de los fármacos , Pavos/genética , Animales , Hígado/metabolismo , Masculino , Transcriptoma
9.
J Toxicol Environ Health A ; 70(20): 1731-44, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17885930

RESUMEN

In January 2004, the normally picturesque Cache Valley in northern Utah made national headlines with the highest PM2.5 levels in the nation. Epidemiological studies linked exposure to particulate air pollution in other locations with stroke and Alzheimer's disease and to early mortality from all causes, cancer, and cardiopulmonary diseases. To determine potential effects of these particles on human health, human bronchial epithelial cells (BEAS-2B) were cultured with PM2.5 collected from various locations in the Cache Valley. These particles were slightly cytotoxic, but more potent than NH4NO3, the major chemical component of Cache Valley PM2.5. Gene expression analysis of PM2.5-exposed cells was performed using microarray and quantitative reverse-transcription polymerase chain reaction (RT-PCR). Among other genes, PM2.5 exposure induced genes and proteins involved in the inflammatory response. Most notably, PM2.5-exposed cells showed significant gene level upregulation of activating receptors to interleukins 1 and 6 (IL-1R1 and IL-6R), as well as concomitant increases in protein. Increases in IL-1 receptor associated kinase-1 (IRAK) protein were observed. PM2.5 exposure resulted in release of IL-6, as well phosphorylated STAT3 protein, providing evidence that PM activates the IL-6/gp130/STAT3 signaling pathway in BEAS-2B cells. IL-20 and major histocompatibility complex peptide class-1 (MICA) were upregulated and cleavage of caspase-12 was detected. In total, our results indicate that Cache Valley PM2.5 produces the upregulation of important cytokine receptors and is able to activate both IL-1R- and IL-6R-mediated signaling pathways in human lung cells. These observations are generally consistent with the adverse effects associated with inhalation of fine particulate matter like PM2.5.


Asunto(s)
Inflamación/genética , Interleucinas/genética , Pulmón/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Material Particulado/toxicidad , Caspasa 3/efectos de los fármacos , Células Cultivadas , Pruebas Inmunológicas de Citotoxicidad , Humanos , Nitratos/análisis , Nitratos/toxicidad , Material Particulado/análisis , Utah
10.
Toxicol Sci ; 89(2): 399-407, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16280384

RESUMEN

Aflatoxin B1 (AFB1) is a potent dietary hepatocarcinogen in animals and probably in humans. Mutations (and altered expression) of the tumor suppresser gene p53 have been observed in liver tumors from patients exposed to high dietary AFB1. Inhalation of AFB1-laden grain dusts has been associated with an increased incidence of lung cancer in humans as well. We examined the effects of low concentrations of AFB1 on the expression of p53 and MDM2 in human bronchial epithelial cells (BEAS-2B) transfected with cDNA for either cytochrome P450 (CYP) 1A2 (B-CMV1A2) or CYP 3A4 (B3A4), two isozymes that are responsible for AFB1 activation in human liver and possibly the lung. Untreated B-CMV1A2 and B3A4 cells constitutively expressed p53. Exposure to a range (0.015-15 microM for 30 min) of AFB1 concentrations caused a concentration-dependent decline in p53 expression in B-CMV1A2 cells, and to a lesser extent, in B3A4 cells. The AFB1-mediated decrease in p53 continued for at least 12 h after 30-min exposures to 1.5 muM AFB(1). Mirroring the decrease in p53 expression was a concentration-dependent increase in the expression of the 76-kDa MDM2 isoform in B-CMV1A2 and B-3A4 cells. Interestingly, AFB1 did not induce DNA laddering, an indicator of apoptotic cell death, but proteolytic activation of caspase-3 was detected in AFB1-treated B-CVM1A2 cells. In total, these data show that low, environmentally-relevant concentrations of AFB1 alter the expression of p53 and MDM2 in these human lung cells, and that cells that stably express CYP 1A2 were more susceptible to this effect than nontransfected, or 3A4-expressing cells.


Asunto(s)
Aflatoxina B1/toxicidad , Apoptosis/efectos de los fármacos , Carcinógenos Ambientales/toxicidad , Sistema Enzimático del Citocromo P-450/biosíntesis , Pulmón/efectos de los fármacos , Proteína p53 Supresora de Tumor/biosíntesis , Western Blotting , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Pulmón/citología , Pulmón/metabolismo , Factores de Tiempo
11.
Cancer Res ; 62(1): 105-12, 2002 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11782366

RESUMEN

Some epidemiological evidence suggests a link between the inhalation of aflatoxin B(1) (AFB(1))-contaminated grain dusts and increased lung cancer risk. However, the mechanisms of AFB(1) activation and action in human lung are not well understood. We compared AFB(1) action in SV40 immortalized human bronchial epithelial cells (BEAS-2B) with two transfected cell lines that stably express human cytochromes P450 (CYPs) 1A2 (B-CMV1A2) and 3A4 (B3A4), the principal CYPs thought to activate this mycotoxin in human liver. All three cell types retained catalytically active glutathione S-transferase, the key phase II enzyme that detoxifies metabolically activated AFB(1). B-CMV1A2 and B3A4 cells expressed methoxyresorufin-O-demethylase (MROD) and nifedipine oxidase activities, respectively, and were 3000- and 70-fold more susceptible, respectively, to the cytotoxic effects of AFB(1) than the control cell line (BEAS-2B). When cultured with a range of low, environmentally relevant AFB(1) concentrations (0.02-1.5 microM), control cells formed barely detectable AFB(1)-DNA adducts, whereas B-CMV1A2 cells formed significantly more adducts than B3A4 cells. In B-CMV1A2 cells, formation of AFB(1)-DNA adducts was inhibited by the CYP 1A2 inhibitor 7,8-benzoflavone, whereas formation of AFB(1)-DNA adducts in B3A4 cells was inhibited by the CYP 3A4 inhibitor 17alpha-ethynylestradiol. Competitive reverse transcription-PCR analysis showed that only the CYP-transfected cell lines expressed CYP mRNA. When adjusted for CYP mRNA expression, B-CMV1A2 cells were more efficient in the formation of cytotoxic and DNA-alkylating species at low AFB(1) concentrations, whereas B3A4 cells were more efficient at high concentrations. Our results affirm the hypothesis that, as in human liver microsomes, CYP 1A2 in human lung cells appears to have a more important role than CYP 3A4 in the bioactivation of low AFB(1) concentrations associated with many human exposures. Therefore, it is possible that under conditions in which appropriate CYPs are expressed in lung, inhalation of AFB(1) may result in increased risk of lung cancer in exposed persons.


Asunto(s)
Aflatoxina B1/farmacocinética , Aflatoxina B1/toxicidad , Bronquios/enzimología , Citocromo P-450 CYP1A2/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Oxigenasas de Función Mixta/metabolismo , Benzoflavonas/farmacología , Biotransformación , Bronquios/citología , Bronquios/metabolismo , Línea Celular , Supervivencia Celular/fisiología , Citocromo P-450 CYP1A2/biosíntesis , Citocromo P-450 CYP1A2/genética , Inhibidores del Citocromo P-450 CYP1A2 , Citocromo P-450 CYP3A , Sistema Enzimático del Citocromo P-450/biosíntesis , Sistema Enzimático del Citocromo P-450/genética , Inhibidores Enzimáticos/farmacología , Humanos , Oxigenasas de Función Mixta/biosíntesis , Oxigenasas de Función Mixta/genética , Oxidorreductasas/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
12.
Toxins (Basel) ; 8(1)2016 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-26751476

RESUMEN

The mycotoxin, aflatoxin B1 (AFB1) is a hepatotoxic, immunotoxic, and mutagenic contaminant of food and animal feeds. In poultry, AFB1 can be maternally transferred to embryonated eggs, affecting development, viability and performance after hatch. Domesticated turkeys (Meleagris gallopavo) are especially sensitive to aflatoxicosis, while Eastern wild turkeys (M. g. silvestris) are likely more resistant. In ovo exposure provided a controlled AFB1 challenge and comparison of domesticated and wild turkeys. Gene expression responses to AFB1 in the embryonic hepatic transcriptome were examined using RNA-sequencing (RNA-seq). Eggs were injected with AFB1 (1 µg) or sham control and dissected for liver tissue after 1 day or 5 days of exposure. Libraries from domesticated turkey (n = 24) and wild turkey (n = 15) produced 89.2 Gb of sequence. Approximately 670 M reads were mapped to a turkey gene set. Differential expression analysis identified 1535 significant genes with |log2 fold change| ≥ 1.0 in at least one pair-wise comparison. AFB1 effects were dependent on exposure time and turkey type, occurred more rapidly in domesticated turkeys, and led to notable up-regulation in cell cycle regulators, NRF2-mediated response genes and coagulation factors. Further investigation of NRF2-response genes may identify targets to improve poultry resistance.


Asunto(s)
Aflatoxina B1/toxicidad , Hígado/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Pavos/genética , Animales , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Femenino , Hígado/embriología , Hígado/metabolismo , Masculino , Análisis de Secuencia de ARN , Especificidad de la Especie
13.
Toxicol Sci ; 82(2): 497-503, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15329442

RESUMEN

Pyrrolizidine alkaloids (PAs) are ubiquitous plant toxins, many of which, upon oxidation by hepatic mixed-function oxidases, become reactive bifunctional pyrrolic electrophiles that form DNA-DNA and DNA-protein cross-links. The anti-mitotic, toxic, and carcinogenic action of PAs is thought to be caused, at least in part, by these cross-links. We wished to determine whether the activated PA pyrrole dehydromonocrotaline (DHMO) exhibits base sequence preferences when cross-linked to a set of model duplex poly A-T 14-mer oligonucleotides with varying internal and/or end 5'-d(CG), 5'-d(GC), 5'-d(TA), 5'-d(CGCG), or 5'-d(GCGC) sequences. DHMO-DNA cross-links were assessed by electrophoretic mobility shift assay (EMSA) of 32P endlabeled oligonucleotides and by HPLC analysis of cross-linked DNAs enzymatically digested to their constituent deoxynucleosides. The degree of DNA cross-links depended upon the concentration of the pyrrole, but not on the base sequence of the oligonucleotide target. Likewise, HPLC chromatograms of cross-linked and digested DNAs showed no discernible sequence preference for any nucleotide. Added glutathione, tyrosine, cysteine, and aspartic acid, but not phenylalanine, threonine, serine, lysine, or methionine competed with DNA as alternate nucleophiles for cross-linking by DHMO. From these data it appears that DHMO exhibits no strong base preference when forming cross-links with DNA, and that some cellular nucleophiles can inhibit DNA cross-link formation.


Asunto(s)
Reactivos de Enlaces Cruzados/farmacología , ADN/efectos de los fármacos , Monocrotalina/análogos & derivados , Monocrotalina/farmacología , Aminoácidos/química , Ácido Aspártico/química , Secuencia de Bases , Cromatografía Líquida de Alta Presión , Reactivos de Enlaces Cruzados/síntesis química , Cisteína/química , ADN/química , Ensayo de Cambio de Movilidad Electroforética , Glutatión/química , Indicadores y Reactivos , Monocrotalina/síntesis química , Desnaturalización de Ácido Nucleico , Oligonucleótidos/química , Pirroles/química , Tirosina/química
14.
J Toxicol Environ Health A ; 65(12): 853-67, 2002 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-12079611

RESUMEN

The mycotoxin aflatoxin B(1) (AFB(1)) is a hepatocarcinogen in many animal models and probably a human carcinogen. Besides being a dietary carcinogen, AFB(1) has been detected in dusts generated in the processing and transportation of AFB(1)-contaminated products. Inhalation of grain dusts contaminated with AFB(1) may be a risk factor in human lung cancer. Aflatoxin B(1) requires cytochrome P-450 (CYP)-mediated activation to form cytotoxic and DNA-reactive intermediates, and this activation in human liver is mediated by the CYP 1A2 and 3A4 isoforms. Which isoforms are important in AFB(1) activation in human lung is not well understood. To investigate whether these CYPs can activate AFB(1) at low, environmentally relevant concentrations in human lung cells, SV40 immortalized human bronchial epithelial cells (BEAS-2B) that were transfected with cDNA for CYPs 3A4 (B3A4) or 1A2 (B-CMV1A2) were used. B-CMV1A2 cultured in 15 nM AFB(1) produced the AFB(1)-glutathione conjugate (AFB(1)-GSH) and aflatoxin M(1) (AFM(1)), while B3A4 cells produced only aflatoxin Q(1) (AFQ(1)) at 0.15 microM AFB(1). Nontransfected BEAS-2B cells produced no metabolites, even at 1.5 mM AFB(1). Microsomes prepared from B-CMV1A2 and B3A4 cells activated AFB(1) to AFB(1) 8,9-epoxide (AFBO), while those from BEAS-2B cells did not produce AFBO. Cytosol from all three cell types was ineffective at glutathione S-transferase (GST)-mediated trapping of enzymatically generated AFB(1) 8,9-epoxide. B-CMV1A2 cells were 100-fold more sensitive to AFB(1) compared to B3A4 cells, and were 6000-fold more sensitive than control BEAS-2B cells. Western immunoblots confirmed that only B-CMV1A2 cells expressed CYP 1A2 protein, while CYP 3A4 was only in B3A4 cells. B-CMV1A2 cells were the most sensitive to AFB(1), followed by B3A4 cells. CYP 3A4, which has been predicted to activate AFB(1) primarily at higher AFB(1) concentrations, was also responsible for significant AFB(1) toxicity at low concentrations. These data indicate that human lung cells expressing these CYP isoforms are capable of activating AFB(1), even at environmentally relevant concentrations.


Asunto(s)
Aflatoxina B1/metabolismo , Aflatoxina B1/toxicidad , Citocromo P-450 CYP1A2/farmacología , Sistema Enzimático del Citocromo P-450/farmacología , Pulmón/citología , Oxigenasas de Función Mixta/farmacología , Western Blotting , Técnicas de Cultivo de Célula , Citocromo P-450 CYP1A2/biosíntesis , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP3A , Sistema Enzimático del Citocromo P-450/biosíntesis , Sistema Enzimático del Citocromo P-450/genética , ADN Complementario , Células Epiteliales , Humanos , Isoenzimas , Pulmón/efectos de los fármacos , Oxigenasas de Función Mixta/biosíntesis , Oxigenasas de Función Mixta/genética , Transfección
15.
PLoS One ; 9(6): e100930, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24979717

RESUMEN

Dietary exposure to aflatoxin B1 (AFB1) is detrimental to avian health and leads to major economic losses for the poultry industry. AFB1 is especially hepatotoxic in domestic turkeys (Meleagris gallopavo), since these birds are unable to detoxify AFB1 by glutathione-conjugation. The impacts of AFB1 on the turkey hepatic transcriptome and the potential protection from pretreatment with a Lactobacillus-based probiotic mixture were investigated through RNA-sequencing. Animals were divided into four treatment groups and RNA was subsequently recovered from liver samples. Four pooled RNA-seq libraries were sequenced to produce over 322 M reads totaling 13.8 Gb of sequence. Approximately 170,000 predicted transcripts were de novo assembled, of which 803 had significant differential expression in at least one pair-wise comparison between treatment groups. Functional analysis linked many of the transcripts significantly affected by AFB1 exposure to cancer, apoptosis, the cell cycle or lipid regulation. Most notable were transcripts from the genes encoding E3 ubiquitin-protein ligase Mdm2, osteopontin, S-adenosylmethionine synthase isoform type-2, and lipoprotein lipase. Expression was modulated by the probiotics, but treatment did not completely mitigate the effects of AFB1. Genes identified through transcriptome analysis provide candidates for further study of AFB1 toxicity and targets for efforts to improve the health of domestic turkeys exposed to AFB1.


Asunto(s)
Aflatoxina B1/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/veterinaria , Enfermedades de las Aves de Corral/genética , Probióticos/farmacología , Transcriptoma , Aflatoxina B1/aislamiento & purificación , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Aspergillus/química , Aspergillus/patogenicidad , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Perfilación de la Expresión Génica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Lactobacillus/fisiología , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Metionina Adenosiltransferasa/genética , Metionina Adenosiltransferasa/metabolismo , Enfermedades de las Aves de Corral/inducido químicamente , Enfermedades de las Aves de Corral/metabolismo , Enfermedades de las Aves de Corral/patología , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Pavos
16.
Res Vet Sci ; 97(2): 274-81, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24997556

RESUMEN

Turkeys are extremely sensitive to aflatoxin B1 (AFB1) which causes decreased growth, immunosuppression and liver necrosis. The purpose of this study was to determine whether probiotic Lactobacillus, shown to be protective in animal and clinical studies, would likewise confer protection in turkeys, which were treated for 11 days with either AFB1 (AFB; 1 ppm in diet), probiotic (PB; 1 × 10(11) CFU/ml; oral, daily), probiotic + AFB1 (PBAFB), or PBS control (CNTL). The AFB1 induced drop in body and liver weights were restored to normal in CNTL and PBAFB groups. Hepatotoxicity markers were not significantly reduced by probiotic treatment. Major histocompatibility complex (MHC) genes BG1 and BG4, which are differentially expressed in liver and spleens, were not significantly affected by treatments. These data indicate modest protection, but the relatively high dietary AFB1 treatment, and the extreme sensitivity of this species may reveal limits of probiotic-based protection strategies.


Asunto(s)
Aflatoxina B1/toxicidad , Enfermedades Transmitidas por los Alimentos/veterinaria , Lactobacillus , Complejo Mayor de Histocompatibilidad/efectos de los fármacos , Micotoxicosis/veterinaria , Enfermedades de las Aves de Corral/prevención & control , Probióticos/farmacología , Probióticos/uso terapéutico , Aflatoxina B1/administración & dosificación , Animales , Quimioprevención/métodos , Quimioprevención/veterinaria , Dieta/efectos adversos , Enfermedades Transmitidas por los Alimentos/patología , Enfermedades Transmitidas por los Alimentos/prevención & control , Hígado/efectos de los fármacos , Hígado/patología , Complejo Mayor de Histocompatibilidad/genética , Micotoxicosis/patología , Micotoxicosis/prevención & control , Tamaño de los Órganos/efectos de los fármacos , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/patología , Especificidad de la Especie , Bazo/efectos de los fármacos , Bazo/patología , Resultado del Tratamiento , Pavos
17.
PLoS One ; 8(4): e60662, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23613737

RESUMEN

Domestic turkeys (Meleagris gallopavo) are one of the most susceptible animals known to the toxic effects of the mycotoxin aflatoxin B1 (AFB1), a potent human hepatocarcinogen, and universal maize contaminant. We have demonstrated that such susceptibility is associated with the inability of hepatic glutathione S-transferases (GSTs) to detoxify the reactive electrophilic metabolite exo-AFB1-8,9-epoxide (AFBO). Unlike their domestic counterparts, wild turkeys, which are relatively AFB1-resistant, possess hepatic GST-mediated AFBO conjugating activity. Here, we characterized the molecular and functional properties of hepatic alpha-class GSTs (GSTAs) from wild and domestic turkeys to shed light on the differences in resistance between these closely related strains. Six alpha-class GST genes (GSTA) amplified from wild turkeys (Eastern and Rio Grande subspecies), heritage breed turkeys (Royal Palm) and modern domestic (Nicholas strain) turkeys were sequenced, and catalytic activities of heterologously-expressed recombinant enzymes determined. Alpha-class identity was affirmed by conserved GST domains and four signature motifs. All GSTAs contained single nucleotide polymorphisms (SNPs) in their coding regions: GSTA1.1 (5 SNPs), GSTA1.2 (7), GSTA1.3 (3), GSTA2 (3), GSTA3 (1) and GSTA4 (2). E. coli-expressed GSTAs possessed varying activities toward GST substrates 1-chloro-2,4-dinitrobenzene (CDNB), 1,2-dichloro-4-nitrobenzene (DCNB), ethacrynic acid (ECA), cumene hydroperoxide (CHP). As predicted by their relative resistance, livers from domestic turkeys lacked detectable GST-mediated AFBO detoxification activity, whereas those from wild and heritage birds possessed this critical activity, suggesting that intensive breeding and selection resulted in loss of AFB1-protective alleles during domestication. Our observation that recombinant tGSTAs detoxify AFBO, whereas their hepatic forms do not, implies that the hepatic forms of these enzymes are down-regulated, silenced, or otherwise modified by one or more mechanisms. These data may inform of possible molecular mechanisms of resistance to AFB1, and may also have the benefit of identifying genetic markers which could be used to enhance AFB1 resistance in modern domestic strains.


Asunto(s)
Aflatoxina B1/toxicidad , Carcinógenos/toxicidad , Glutatión Transferasa/genética , Isoenzimas/metabolismo , Animales , Derivados del Benceno/farmacología , Dinitroclorobenceno/farmacología , Ácido Etacrínico/farmacología , Glutatión Transferasa/clasificación , Glutatión Transferasa/metabolismo , Nitrobencenos/farmacología , Filogenia , Polimorfismo de Nucleótido Simple/genética , Pavos
18.
Comp Biochem Physiol C Toxicol Pharmacol ; 158(2): 109-16, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23712008

RESUMEN

Hepatic glutathione S-transferases (GSTs: EC2.5.1.1.8) catalyze the detoxification of reactive electrophilic compounds, many of which are toxic and carcinogenic intermediates, via conjugation with the endogenous tripeptide glutathione (GSH). Glutathione S-transferase (GST)-mediated detoxification is a critical determinant of species susceptibility to the toxic and carcinogenic mycotoxin aflatoxin B1 (AFB1), which in resistant animals efficiently detoxifies the toxic intermediate produced by hepatic cytochrome P450 bioactivation, the exo-AFB1-8,9-epoxide (AFBO). Domestic turkeys (Meleagris gallopavo) are one of the most sensitive animals known to AFB1, a condition associated with a deficiency of hepatic GST-mediated detoxification of AFBO. We have recently shown that unlike their domestic counterparts, wild turkeys (Meleagris gallopavo silvestris), which are relatively resistant, express hepatic GST-mediated detoxification activity toward AFBO. Because of the importance of GSTs in species susceptibility, and to explore possible GST classes involved in AFB1 detoxification, we amplified, cloned, expressed and functionally characterized the hepatic mu-class GSTs tGSTM3 (GenBank accession no. JF340152), tGSTM4 (JF340153) from domestic turkeys, and a GSTM4 variant (ewGSTM4, JF340154) from Eastern wild turkeys. Predicted molecular masses of tGSTM3 and two tGSTM4 variants were 25.6 and 25.8kDa, respectively. Multiple sequence comparisons revealed four GSTM motifs and the mu-loop in both proteins. tGSTM4 has 89% amino acid sequence identity to chicken GSTM2, while tGSTM3 has 73% sequence identity to human GSTM3 (hGSTM3). Specific activities of Escherichia coli-expressed tGSTM3 toward 1-chloro-2,4-dinitrobenzene (CDNB) and peroxidase activity toward cumene hydroperoxide were five-fold greater than tGSTM4 while tGSTM4 possessed more than three-fold greater activity toward 1,2-dichloro-4-nitrobenzene (DCNB). The two enzymes displayed equal activity toward ethacrynic acid (ECA). However, none of the GSTM proteins had AFBO detoxification capability, in contrast to recombinant alpha-class GSTs shown in our recent study to possess this important activity. In total, our data indicate that although turkey hepatic GSTMs may contribute to xenobiotic detoxification, they probably play no role in detoxification of AFBO in the liver.


Asunto(s)
Aflatoxina B1/metabolismo , Glutatión Transferasa/biosíntesis , Pavos/metabolismo , Aflatoxina B1/toxicidad , Secuencia de Aminoácidos , Animales , Derivados del Benceno/metabolismo , Dinitroclorobenceno/metabolismo , Ácido Etacrínico/metabolismo , Glutatión Transferasa/metabolismo , Inactivación Metabólica , Hígado/enzimología , Masculino , Datos de Secuencia Molecular , Nitrobencenos/metabolismo , Alineación de Secuencia , Especificidad por Sustrato
19.
Cell Biochem Biophys ; 67(3): 1147-56, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23712864

RESUMEN

Diesel exhaust particles (DEP) in urban air are associated with numerous respiratory diseases. The role of underlying biomechanics in cytotoxicity of individual lung cells relating to DEP exposure is unclear. In this study, atomic force microscopy (AFM), confocal Raman microspectroscopy (RM), and fluorescence (FL) microscopy were used to monitor alterations of single A549 cells exposed to DEP. Results revealed a significant decrease in membrane surface adhesion force and a significant change in cell elasticity as a function of DEP-cell interaction time, and the dynamic changes in cellular biocomponents which were reflected by changes of characteristic Raman bands: 726 cm(-1) (adenine), 782 cm(-1) (uracil, cytosine, thymine), 788 cm(-1) (O-P-O), 1006 cm(-1) (phenylalanine), and 1320 cm(-1) (guanine) after DEP exposure. These findings suggest that the combination of multi-instruments (e.g., AFM/FL) may offer an exciting platform for investigating the roles of biophysical and biochemical responses to particulate matter-induced cell toxicity.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Material Particulado/toxicidad , Emisiones de Vehículos/toxicidad , Línea Celular Tumoral , Membrana Celular/química , Membrana Celular/efectos de los fármacos , Módulo de Elasticidad , Células Epiteliales/química , Humanos , Microscopía de Fuerza Atómica , Microscopía Confocal , Espectrometría Raman , Factores de Tiempo
20.
Toxicol Sci ; 124(1): 45-53, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21876218

RESUMEN

Six Alpha-class glutathione S-transferase (GST) subunits were cloned from domestic turkey livers, which are one of the most susceptible animals known to the carcinogenic mycotoxin aflatoxin B1. In most animals, GST dysfunction is a risk factor for susceptibility toward AFB1, and we have shown that turkeys lack GSTs with affinity toward the carcinogenic intermediate exo-aflatoxin B(1)-8-9-epoxide (AFBO). Conversely, mice are resistant to AFB1 carcinogenesis, due to high constitutive expression of mGSTA3 that has high affinity toward AFBO. When expressed in Escherichia coli, all six tGSTA subunits possessed conjugating activities toward substrates 1-chloro-2,4-dinitrobenzene (CDNB), 1,2-dichloro-4-nitrobenzene (DCNB), ethacrynic acid (ECA), and cumene hydroperoxide (CHP) with tGSTA1.2 appearing most active. Interestingly, tGSTA1.1, which lacks one of the four Alpha-class signature motifs, possessed enzymatic activities toward all substrates. All had comparable activities toward AFBO conjugation, an activity absent in turkey liver cytosols. E. coli-expressed mGSTA3 conjugated AFBO with more than 3-fold greater activity than that of tGSTAs and had higher activity toward GST prototype substrates. Mouse hepatic cytosols had approximately 900-fold higher catalytic activity toward AFBO compared with those from turkey. There was no apparent amino acid profile in tGSTAs that might correspond to specificity toward AFBO, although tGSTA1.2, which had slightly higher AFBO-trapping ability, shared Tyr¹°8 with mGSTA3, a residue postulated to be critical for AFBO trapping activity in mammalian systems. The observation that recombinant tGSTAs detoxify AFBO, whereas their hepatic forms do not, implies that the hepatic forms of these enzymes are silenced by one or more regulatory mechanisms.


Asunto(s)
Aflatoxina B1/análogos & derivados , Glutatión Transferasa/fisiología , Isoenzimas/fisiología , Hígado/enzimología , Pavos , Aflatoxina B1/farmacocinética , Aflatoxina B1/toxicidad , Secuencia de Aminoácidos , Animales , Clonación Molecular , Citosol/enzimología , Electroforesis en Gel de Poliacrilamida , Escherichia coli/genética , Glutatión Transferasa/química , Glutatión Transferasa/genética , Humanos , Isoenzimas/química , Isoenzimas/genética , Hígado/efectos de los fármacos , Masculino , Ratones , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Subunidades de Proteína , Ratas , Alineación de Secuencia , Especificidad de la Especie , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA