Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Toxins (Basel) ; 11(5)2019 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-31035349

RESUMO

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.


Assuntos
Aflatoxina B1/toxicidade , Baço/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Ração Animal , Animais , Animais Domésticos , Animais Selvagens , Resistência à Doença , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Micotoxicose/genética , Baço/metabolismo , Perus
2.
Toxins (Basel) ; 11(1)2019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30669283

RESUMO

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.


Assuntos
Aflatoxina B1/toxicidade , Animais Domésticos/genética , Trato Gastrointestinal/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Perus/genética , Animais , Trato Gastrointestinal/metabolismo , Glutationa Transferase/genética , Isoenzimas/genética , Masculino
3.
Toxins (Basel) ; 10(1)2018 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-29342849

RESUMO

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.


Assuntos
Aflatoxina B1/toxicidade , Fígado/efeitos dos fármacos , Perus/genética , Animais , Fígado/metabolismo , Masculino , Transcriptoma
4.
Toxins (Basel) ; 8(1)2016 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-26751476

RESUMO

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.


Assuntos
Aflatoxina B1/toxicidade , Fígado/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Perus/genética , Animais , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Feminino , Fígado/embriologia , Fígado/metabolismo , Masculino , Análise de Sequência de RNA , Especificidade da Espécie
5.
Immunogenetics ; 67(3): 163-78, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25597949

RESUMO

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.


Assuntos
Aflatoxina B1/toxicidade , Proteínas Aviárias/genética , Doenças das Aves/genética , Intoxicação Alimentar por Cogumelos/veterinária , Probióticos/administração & dosagem , Transcriptoma/efeitos dos fármacos , Animais , Proteínas Aviárias/imunologia , Doenças das Aves/imunologia , Perfilação da Expressão Gênica , Granzimas/genética , Granzimas/imunologia , Sequenciamento de Nucleotídeos em Larga Escala , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/genética , Imunomodulação/efeitos dos fármacos , Interleucina-2/genética , Interleucina-2/imunologia , Linfocinas/genética , Linfocinas/imunologia , Anotação de Sequência Molecular , Intoxicação Alimentar por Cogumelos/genética , Intoxicação Alimentar por Cogumelos/imunologia , Perforina/genética , Perforina/imunologia , Sialoglicoproteínas/genética , Sialoglicoproteínas/imunologia , Baço/efeitos dos fármacos , Baço/imunologia , Baço/metabolismo , Transcriptoma/imunologia , Perus , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/imunologia
6.
PLoS One ; 9(6): e100930, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24979717

RESUMO

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.


Assuntos
Aflatoxina B1/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/veterinária , Doenças das Aves Domésticas/genética , Probióticos/farmacologia , Transcriptoma , Aflatoxina B1/isolamento & purificação , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Aspergillus/química , Aspergillus/patogenicidade , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Perfilação da Expressão Gênica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Lactobacillus/fisiologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Lipase Lipoproteica/genética , Lipase Lipoproteica/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Metionina Adenosiltransferase/genética , Metionina Adenosiltransferase/metabolismo , Doenças das Aves Domésticas/induzido quimicamente , Doenças das Aves Domésticas/metabolismo , Doenças das Aves Domésticas/patologia , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Perus
7.
Res Vet Sci ; 97(2): 274-81, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24997556

RESUMO

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.


Assuntos
Aflatoxina B1/toxicidade , Doenças Transmitidas por Alimentos/veterinária , Lactobacillus , Complexo Principal de Histocompatibilidade/efeitos dos fármacos , Micotoxicose/veterinária , Doenças das Aves Domésticas/prevenção & controle , Probióticos/farmacologia , Probióticos/uso terapêutico , Aflatoxina B1/administração & dosagem , Animais , Quimioprevenção/métodos , Quimioprevenção/veterinária , Dieta/efeitos adversos , Doenças Transmitidas por Alimentos/patologia , Doenças Transmitidas por Alimentos/prevenção & controle , Fígado/efeitos dos fármacos , Fígado/patologia , Complexo Principal de Histocompatibilidade/genética , Micotoxicose/patologia , Micotoxicose/prevenção & controle , Tamanho do Órgão/efeitos dos fármacos , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/patologia , Especificidade da Espécie , Baço/efeitos dos fármacos , Baço/patologia , Resultado do Tratamento , Perus
8.
Comp Biochem Physiol C Toxicol Pharmacol ; 158(2): 109-16, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23712008

RESUMO

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.


Assuntos
Aflatoxina B1/metabolismo , Glutationa Transferase/biossíntese , Perus/metabolismo , Aflatoxina B1/toxicidade , Sequência de Aminoácidos , Animais , Derivados de Benzeno/metabolismo , Dinitroclorobenzeno/metabolismo , Ácido Etacrínico/metabolismo , Glutationa Transferase/metabolismo , Inativação Metabólica , Fígado/enzimologia , Masculino , Dados de Sequência Molecular , Nitrobenzenos/metabolismo , Alinhamento de Sequência , Especificidade por Substrato
9.
Cell Biochem Biophys ; 67(3): 1147-56, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23712864

RESUMO

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.


Assuntos
Células Epiteliais/efeitos dos fármacos , Material Particulado/toxicidade , Emissões de Veículos/toxicidade , Linhagem Celular Tumoral , Membrana Celular/química , Membrana Celular/efeitos dos fármacos , Módulo de Elasticidade , Células Epiteliais/química , Humanos , Microscopia de Força Atômica , Microscopia Confocal , Análise Espectral Raman , Fatores de Tempo
10.
PLoS One ; 8(4): e60662, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23613737

RESUMO

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.


Assuntos
Aflatoxina B1/toxicidade , Carcinógenos/toxicidade , Glutationa Transferase/genética , Isoenzimas/metabolismo , Animais , Derivados de Benzeno/farmacologia , Dinitroclorobenzeno/farmacologia , Ácido Etacrínico/farmacologia , Glutationa Transferase/classificação , Glutationa Transferase/metabolismo , Nitrobenzenos/farmacologia , Filogenia , Polimorfismo de Nucleotídeo Único/genética , Perus
11.
Arch Toxicol ; 86(1): 121-35, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21818627

RESUMO

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.


Assuntos
Inflamação/induzido quimicamente , Pulmão/efeitos dos fármacos , Material Particulado/toxicidade , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Inflamação/patologia , Interleucina-1alfa/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Pulmão/citologia , Pulmão/metabolismo , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Tamanho da Partícula , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos , Saúde da População Urbana , Utah
12.
Toxicol Sci ; 124(1): 45-53, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21876218

RESUMO

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.


Assuntos
Aflatoxina B1/análogos & derivados , Glutationa Transferase/fisiologia , Isoenzimas/fisiologia , Fígado/enzimologia , Perus , Aflatoxina B1/farmacocinética , Aflatoxina B1/toxicidade , Sequência de Aminoácidos , Animais , Clonagem Molecular , Citosol/enzimologia , Eletroforese em Gel de Poliacrilamida , Escherichia coli/genética , Glutationa Transferase/química , Glutationa Transferase/genética , Humanos , Isoenzimas/química , Isoenzimas/genética , Fígado/efeitos dos fármacos , Masculino , Camundongos , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Subunidades Proteicas , Ratos , Alinhamento de Sequência , Especificidade da Espécie , Especificidade por Substrato
13.
Toxicol Appl Pharmacol ; 254(3): 349-54, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21616088

RESUMO

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.


Assuntos
Aflatoxina B1/metabolismo , Hidrocarboneto de Aril Hidroxilases/fisiologia , Microssomos Hepáticos/metabolismo , Animais , Família 3 do Citocromo P450 , Microssomos Hepáticos/enzimologia , Perus
14.
PLoS Biol ; 8(9)2010 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-20838655

RESUMO

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.


Assuntos
Genoma , Perus/genética , Animais , Sequência de Bases , Mapeamento Cromossômico , DNA/genética , Polimorfismo de Nucleotídeo Único , Análise de Sequência de DNA , Homologia de Sequência do Ácido Nucleico , Especificidade da Espécie
15.
Chem Res Toxicol ; 23(8): 1322-9, 2010 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-20707407

RESUMO

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.


Assuntos
Aflatoxina B1/metabolismo , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Fígado/metabolismo , Perus , Sequência de Aminoácidos , Animais , Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Clonagem Molecular , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Inibidores do Citocromo P-450 CYP3A , Etinilestradiol/farmacologia , Perfilação da Expressão Gênica , Humanos , Dados de Sequência Molecular , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade da Espécie
16.
Res Vet Sci ; 89(3): 325-31, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20462619

RESUMO

Aflatoxins (AF) are ubiquitous in corn-based animal feed and causes hepatotoxic and hepatocarcinogenic effects. The most important AF in terms of toxic potency and occurrence is aflatoxin B1 (AFB1). Poultry, especially turkeys, are extremely sensitive to the toxic and carcinogenic action of AFB1, resulting in millions of dollars in annual losses to producers due to reduced growth rate, increased susceptibility to disease, reduced egg production and other adverse effects. The extreme sensitivity of turkeys and other poultry to AFB1 is associated with efficient hepatic cytochrome P450-mediated bioactivation and deficient detoxification by glutathione S-transferases (GST). Discerning the biochemical and molecular mechanisms of this extreme sensitivity of poultry to AFB1, will contribute in the development of novel strategies to increase aflatoxin resistance. Since AFB1 is an unavoidable contaminant of corn-based poultry feed, chemoprevention strategies aimed at reducing AFB1 toxicity in poultry and in other animals have been the subject of numerous studies. This brief review summarizes many of the key recent findings regarding the action of aflatoxins in poultry.


Assuntos
Aflatoxina B1/toxicidade , Doenças das Aves Domésticas/induzido quimicamente , Aflatoxina B1/metabolismo , Animais , Galinhas/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Glutationa Transferase/metabolismo , Doenças das Aves Domésticas/prevenção & controle , Perus/metabolismo
17.
Gene ; 452(2): 45-53, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19913078

RESUMO

Glutathione S-transferases (GSTs: EC2.5.1.18) are a superfamily of multifunctional dimeric enzymes that catalyze the conjugation of glutathione (GSH) to electrophilic chemicals. In most animals and in humans, GSTs are the principal enzymes responsible for detoxifying the mycotoxin aflatoxin B(1) (AFB(1)) and GST dysfunction is a known risk factor for susceptibility towards AFB(1). Turkeys are one of the most susceptible animals known to AFB(1), which is a common contaminant of poultry feeds. The extreme susceptibility of turkeys is associated with hepatic GSTs unable to detoxify the highly reactive and electrophilic metabolite exo-AFB(1)-8,9-epoxide (AFBO). In this study, comparative genomic approaches were used to amplify and identify the alpha-class tGST genes (tGSTA1.1, tGSTA1.2, tGSTA1.3, tGSTA2, tGSTA3 and tGSTA4) from turkey liver. The conserved GST domains and four alpha-class signature motifs in turkey GSTs (with the exception of tGSTA1.1 which lacked one motif) confirm the presence of hepatic alpha-class GSTs in the turkey. Four signature motifs and conserved residues found in alpha-class tGSTs are (1) xMExxxWLLAAAGVE, (2) YGKDxKERAxIDMYVxG, (3) PVxEKVLKxHGxxxL and (4) PxIKKFLXPGSxxKPxxx. A BAC clone containing the alpha-class GST gene cluster was isolated and sequenced. The turkey alpha-class GTS genes genetically map to chromosome MGA2 with synteny between turkey and human alpha-class GSTs and flanking genes. This study identifies the alpha-class tGST gene cluster and genetic markers (SNPs, single nucleotide polymorphisms) that can be used to further examine AFB(1) susceptibility and resistance in turkeys. Functional characterization of heterologously expressed proteins from these genes is currently underway.


Assuntos
Glutationa Transferase/genética , Perus/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Sequência Conservada , Genômica , Glutationa Transferase/química , Humanos , Masculino , Dados de Sequência Molecular , Família Multigênica , Alinhamento de Sequência
18.
Toxicol Sci ; 112(1): 111-22, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19675143

RESUMO

Because of its presumed adverse health effects, particulate air pollution (PM) has received growing attention, but the cellular mechanisms by which PM exerts toxicity are not well elucidated. PM has been associated with early mortality from illnesses that share endoplasmic reticulum (ER) stress as a mechanism of pathogenesis. In this study, we examined whether PM would induce the unfolded protein response (UPR) which is a cellular response to ER stress. Coarse (PM(10)) and fine (PM(2.5)) PM was collected from a single location in Northern Utah's Cache Valley during atmospheric inversions occurring in January 2002 and January 2003. Extracts of PM samples were added (12.5 and 25 microg/ml) to cultured human bronchial epithelial (BEAS-2B) cells for 24 h. At these concentrations neither PM nor LPS exhibited demonstrable cytotoxicity by the neutral red assay. However, PM elicited significant increases of unfolded protein response (UPR)-related post-translational modifications, such as S6 ribosomal protein, heat-shock protein (Hsp)27, and protein kinase related protein phosphorylation and cleavage of activating transcription factor (ATF)-6. PM exposure also resulted in significant increases in the UPR-associated proteins ATF-4, Hsp70, Hsp90, and binding immunoglobulin protein. PM also interfered with the export of Hsp70 from the cells in a concentration-dependent manner and resulted in release of C-reactive protein. Calpain was upregulated and activated in PM-treated cultures, though these events were not proapoptotic. This study demonstrates that PM is capable of inducing ER stress and the UPR in vitro and may be a mechanism by which PM exerts toxicity.


Assuntos
Poluentes Atmosféricos/toxicidade , Retículo Endoplasmático/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Saúde da População Urbana , Western Blotting , Linhagem Celular , Retículo Endoplasmático/metabolismo , Proteínas de Choque Térmico/metabolismo , Humanos , Pulmão/citologia , Pulmão/metabolismo , Tamanho da Partícula , Fosforilação , Desnaturação Proteica , Processamento de Proteína Pós-Traducional
19.
Toxicol Appl Pharmacol ; 227(3): 339-46, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18177683

RESUMO

Chemoprevention of toxicoses and/or cancer through the use of nutrients or pharmacologic compounds is the subject of intense study. Among the many compounds examined, food additives such as antioxidants are being considered due to their ability to reduce disease formation by either induction or inhibition of key enzyme systems. One such compound, butylated hydroxytoluene (BHT), has been found to protect against cancer formation caused by exposure to aflatoxin B1 (AFB1) in rodents. We have shown that dietary BHT protects against clinical signs of aflatoxicosis in turkeys, a species that is very susceptible to this mycotoxin. In this study, the effect of BHT on AFB1 metabolism and other cytochrome P450 (CYP)-related enzyme activities in turkey liver microsomes was examined to discern possible mechanisms of BHT-mediated protection against aflatoxicosis. Ethoxyresorufin O-deethylase (EROD), methoxyresorufin O-demethylase (MROD), prototype activities for CYP1A1 and 1A2, respectively, were decreased in the BHT fed (4000 ppm) animals, while oxidation of nifedipine, a prototype activity for CYP3A4, was increased. However, BHT added to microsomal incubations inhibited these CYP activities in a concentration-related manner. Importantly, BHT inhibited conversion of AFB1 to the reactive intermediate AFB1-8,-9-epoxide (AFBO), exhibiting Michaelis-Menton competitive inhibition kinetics (Ki=0.81 microM). Likewise, microsomes prepared from turkeys fed BHT were significantly less active in AFBO formation compared to those from control birds. When turkeys were fed BHT for up to 40 days, residual BHT was present in liver, breast meat, thigh meat and abdominal fat in concentrations substantially below U.S. FDA guidelines for this antioxidant, but in concentrations greater than the Ki, likely sufficient to inhibit bioactivation of AFB1in vivo. BHT-induced hydropic degeneration in the livers of BHT fed animals was significantly greater in birds that remained on BHT treatment for up to 30 days, but this lesion diminished in animals fed for 40 days or when returned to a control diet. The data indicate that the observed chemopreventive properties of BHT in turkeys may be due, at least in part, to its ability to inhibit hepatic AFB1 epoxidation and also that the BHT-induced hydropic degeneration is reversible and does not appear to cause long-term effects.


Assuntos
Aflatoxina B1/antagonistas & inibidores , Aflatoxina B1/toxicidade , Antioxidantes/farmacologia , Hidroxitolueno Butilado/farmacologia , Fígado/efeitos dos fármacos , Fígado/enzimologia , Aflatoxina B1/metabolismo , Animais , Antioxidantes/administração & dosagem , Hidroxitolueno Butilado/administração & dosagem , Hidroxitolueno Butilado/metabolismo , Quimioprevenção , Citocromo P-450 CYP1A1/antagonistas & inibidores , Citocromo P-450 CYP1A1/metabolismo , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/metabolismo , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Nifedipino/metabolismo , Oxirredutases/antagonistas & inibidores , Oxirredutases/metabolismo , Distribuição Tecidual , Perus
20.
J Toxicol Environ Health A ; 70(20): 1731-44, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17885930

RESUMO

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.


Assuntos
Inflamação/genética , Interleucinas/genética , Pulmão/efeitos dos fármacos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Material Particulado/toxicidade , Caspase 3/efeitos dos fármacos , Células Cultivadas , Testes Imunológicos de Citotoxicidade , Humanos , Nitratos/análise , Nitratos/toxicidade , Material Particulado/análise , Utah
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA