Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
1.
Biochem Pharmacol ; 192: 114678, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34265279

RESUMEN

Fibroblast growth factor 21 (FGF21) is mainly regulated by peroxisome proliferator-activated receptor α (PPARα) in liver. The PPARα-FGF21 axis protects against alcohol-related liver disease (ALD). FGF21 exerts its effect via FGF receptor 1 (FGFR1). However, liver specific FGFR1 abrogation had no effect on ALD. Adipose tissues highly express FGFR1. When adipocyte specific FGFR1 knockout (fgfr1adipoQ-cre) mice and corresponding normal control (fgfr1fl/fl) mice were fed with Lieber-DeCarli ethanol liquid diet for 3 weeks, liver triglyceride (TG) accumulation was increased in the fgfr1fl/fl mice to a greater extent than in the fgfr1adipoQ-cre mice. When PPARα agonist WY-14,643 was added in the liquid ethanol diet at 10 mg/L, the ethanol-induced liver TG accumulation was blunted in the fgfr1fl/fl mice but not in the fgfr1adipoQ-cre mice. There was no significant difference in WY-14,643-induced fatty acid oxidation, ethanol metabolism, and oxidative stress between the fgfr1fl/fl and fgfr1adipoQ-cre mice. Interestingly, adipose atrophy was induced by WY-14,643 in the fgfr1adipoQ-cre mice but not in the fgfr1fl/fl mice. Serum free fatty acid was also decreased by WY-14,643 in the fgfr1adipoQ-cre mice but not in the fgfr1fl/fl mice. These results suggest that WY-14,643 inhibits alcoholic fatty liver and regulates adipose tissue mass and fat mobilization from adipose tissues to liver in an adipocyte FGFR1-dependent manner.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Etanol/toxicidad , Hígado Graso Alcohólico/prevención & control , PPAR alfa/agonistas , Pirimidinas/uso terapéutico , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/deficiencia , Tejido Adiposo/metabolismo , Animales , Atrofia/inducido químicamente , Atrofia/metabolismo , Etanol/administración & dosificación , Hígado Graso Alcohólico/metabolismo , Femenino , Ratones , Ratones Noqueados , PPAR alfa/metabolismo , Proliferadores de Peroxisomas/uso terapéutico , Proliferadores de Peroxisomas/toxicidad , Pirimidinas/toxicidad , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética
2.
Aquat Toxicol ; 187: 153-162, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28415051

RESUMEN

Peroxisome proliferators cause species-specific effects, which seem to be primarily transduced by peroxisome proliferator-activated receptor alpha (PPARα). Interestingly, PPARα has a close interrelationship with estrogenic signaling, and this latter has already been promptly activated in brown trout primary hepatocytes. Thus, and further exploring this model, we assess here the reactivity of two PPARα agonists in direct peroxisomal routes and, in parallel the cross-interferences in estrogen receptor (ER) mediated paths. To achieve these goals, three independent in vitro studies were performed using single exposures to clofibrate - CLF (50, 500 and 1000µM), Wy-14,643 - Wy (50 and 150µM), GW6471 - GW (1 and 10µM), and mixtures, including PPARα agonist or antagonist plus an ER agonist or antagonist. Endpoints included gene expression analysis of peroxisome/lipidic related genes (encoding apolipoprotein AI - ApoAI, fatty acid binding protein 1 - Fabp1, catalase - Cat, 17 beta-hydroxysteroid dehydrogenase 4 - 17ß-HSD4, peroxin 11 alpha - Pex11α, PPARαBb, PPARαBa and urate oxidase - Uox) and those encoding estrogenic targets (ERα, ERß-1 and vitellogenin A - VtgA). A quantitative morphological approach by using a pre-validated catalase immunofluorescence technique allowed checking possible changes in peroxisomes. Our results show a low responsiveness of trout hepatocytes to model PPARα agonists in direct target receptor pathways. Additionally, we unveiled interferences in estrogenic signaling caused by Wy, leading to an up-regulation VtgA and ERα at 150µM; these effects seem counteracted with a co-exposure to an ER antagonist. The present data stress the potential of this in vitro model for further exploring the physiological/toxicological implications related with this nuclear receptor cross-regulation.


Asunto(s)
Hepatocitos/efectos de los fármacos , PPAR alfa/metabolismo , Proliferadores de Peroxisomas/toxicidad , Peroxisomas/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Trucha/metabolismo , Contaminantes Químicos del Agua/toxicidad , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/antagonistas & inhibidores , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Hepatocitos/metabolismo , Ratones , PPAR alfa/agonistas , PPAR alfa/genética , Peroxisomas/genética , Cultivo Primario de Células , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Estrógenos/genética , Transducción de Señal , Regulación hacia Arriba , Vitelogeninas/genética , Vitelogeninas/metabolismo
3.
Arch Toxicol ; 90(12): 3061-3071, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26838045

RESUMEN

It was reported that 2,4-dichlorophenoxyacetic acid (2,4-D), a commonly used herbicide and a possible endocrine disruptor, can disturb spermatogenesis, but the precise mechanism is not understood. Since 2,4-D is a weak peroxisome proliferator in hepatocytes and peroxisome proliferator-activated receptor α (PPARα) is also expressed in Leydig cells, this study aimed to investigate the link between PPARα and 2,4-D-mediated testicular dysfunction. 2,4-D (130 mg/kg/day) was administered to wild-type and Ppara-null mice for 2 weeks, and the alterations in testis and testosterone/cholesterol metabolism in Leydig cells were examined. Treatment with 2,4-D markedly decreased testicular testosterone in wild-type mice, leading to degeneration of spermatocytes and Sertoli cells. The 2,4-D decreased cholesterol levels in Leydig cells of wild-type mice through down-regulating the expression of 3-hydroxy-3-methylglutaryl coenzyme A synthase 1 and reductase, involved in de novo cholesterogenesis. However, the mRNAs encoding the important proteins involved in testosterone synthesis were unchanged by 2,4-D except for CYP17A1, indicating that exhausted cholesterol levels in the cells is a main reason for reduced testicular testosterone. Additionally, pregnancy rate and the number of pups between 2,4-D-treated wild-type male mice and untreated female mice were significantly lower compared with those between untreated couples. These phenomena were not observed in 2,4-D-treated Ppara-null males. Collectively, these results suggest a critical role for PPARα in 2,4-D-induced testicular toxicity due to disruption of cholesterol/testosterone homeostasis in Leydig cells. This study yields novel insights into the possible mechanism of testicular dysfunction and male infertility caused by 2,4-D.


Asunto(s)
Ácido 2,4-Diclorofenoxiacético/toxicidad , Disruptores Endocrinos/toxicidad , Herbicidas/toxicidad , Infertilidad Masculina/inducido químicamente , Células Intersticiales del Testículo/efectos de los fármacos , PPAR alfa/metabolismo , Testosterona/metabolismo , Ácido 2,4-Diclorofenoxiacético/administración & dosificación , Animales , Colesterol/química , Relación Dosis-Respuesta a Droga , Disruptores Endocrinos/administración & dosificación , Represión Enzimática/efectos de los fármacos , Herbicidas/administración & dosificación , Hidroximetilglutaril-CoA Reductasas/química , Hidroximetilglutaril-CoA Reductasas/genética , Hidroximetilglutaril-CoA Reductasas/metabolismo , Hidroximetilglutaril-CoA Sintasa/antagonistas & inhibidores , Hidroximetilglutaril-CoA Sintasa/genética , Hidroximetilglutaril-CoA Sintasa/metabolismo , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Infertilidad Masculina/fisiopatología , Células Intersticiales del Testículo/metabolismo , Células Intersticiales del Testículo/patología , Gotas Lipídicas/efectos de los fármacos , Gotas Lipídicas/metabolismo , Gotas Lipídicas/patología , Masculino , Ratones de la Cepa 129 , Ratones Noqueados , PPAR alfa/genética , Proliferadores de Peroxisomas/administración & dosificación , Proliferadores de Peroxisomas/toxicidad , Distribución Aleatoria , Epitelio Seminífero/efectos de los fármacos , Epitelio Seminífero/metabolismo , Epitelio Seminífero/patología , Epitelio Seminífero/fisiopatología , Espermatogénesis/efectos de los fármacos
4.
Arch Environ Contam Toxicol ; 68(1): 148-58, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25015730

RESUMEN

Perfluorooctanoic acid (PFOA) is a persistent environmental contaminant. Activation of the peroxisome proliferator activated receptor alpha (PPARα) resulting from exposure to PFOA has been extensively studied in rodents. However, marked differences in response to peroxisome proliferators prevent extrapolation of rodent PPARα activation to human health risks and additional molecular mechanisms may also be involved in the biological response to PFOA exposure. To further explore the potential involvement of such additional pathways, the effects of PFOA exposure on urinary metabolites were directly compared with those of other well-known PPARα agonists. Male rats were administered PFOA (10, 33, or 100 mg/kg/d), fenofibrate (100 mg/kg/d), or di(2-ethylhexyl) phthalate (100 mg/kg/d) by gavage for 3 consecutive days and allowed to recover for 4 days, and overnight urine was collected. Greater urinary output was observed exclusively in PFOA-treated rats as the total fraction of PFOA excreted in urine increased with the dose administered. Assessment of urinary metabolites (ascorbic acid, quinolinic acid, 8-hydroxy-2'-deoxyguanosine, and malondialdehyde) provided additional information on PFOA's effects on hepatic glucuronic acid and tryptophan-nicotinamide adenine dinucleotide (NAD) pathways and on oxidative stress, whereas increased liver weight and palmitoyl-CoA oxidase activity indicative of PPARα activation and peroxisomal proliferation persisted up to day five after the last exposure.


Asunto(s)
Caprilatos/toxicidad , Desoxiguanosina/análogos & derivados , Fluorocarburos/toxicidad , Proliferadores de Peroxisomas/toxicidad , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Caprilatos/orina , Desoxiguanosina/orina , Fluorocarburos/orina , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Estrés Oxidativo , Proliferadores de Peroxisomas/orina , Ratas
5.
J Toxicol Environ Health A ; 76(11): 635-50, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23941634

RESUMEN

Perfluorooctanoic acid (PFOA) is an environmental contaminant known to induce developmental toxicity in animal models through activation of the peroxisome proliferator-activated receptor α (PPARα). Previously, it was demonstrated that in ovo exposure to PFOA induced cardiotoxicity in chicken embryos and hatchlings. To investigate potential PPARα-mediated mechanisms, fertile chicken eggs were injected prior to incubation with WY 14,643, a PPARα agonist. Cardiac morphology and function were evaluated in late-stage embryos and hatchlings. Histologically, unlike PFOA, WY 14,643 did not induce thinning of the right ventricular wall. Via echocardiography, however, WY 14,643 induced effects similar to those of PFOA, including increased left ventricular wall thickness and mass, elevated heart rate, ejection fraction, fractional shortening, and decreased stroke volume. Additionally, to investigate mechanisms associated with early heart development, a separate group of fertile chicken eggs was injected prior to incubation with PFOA or WY 14,643 and in early-stage embryos, gene expression and protein concentration associated with the bone morphogenic protein (BMP2) pathway were determined. Although changes were not statistically consistent among doses, expression of BMP2, Nkx2.5, and GATA4 mRNA in early embryos was altered by PFOA exposure; however, protein concentrations of these targets were not markedly altered by either PFOA or WY 14,643. Protein levels of pSMAD1/5, a transcriptional regulator stimulated by BMPs, were altered by both PFOA and WY 14,643, but in different directions; PFOA reduced cytoplasmic pSMAD1/5, whereas WY 14,643 decreased nuclear pSMAD1/5. Taken together, these data suggest that developmental cardiotoxicity induced by PFOA likely involves both PPARα and BMP2 pathways.


Asunto(s)
Caprilatos/toxicidad , Embrión no Mamífero/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Corazón Fetal/efectos de los fármacos , Fluorocarburos/toxicidad , Cardiopatías Congénitas/inducido químicamente , PPAR alfa/metabolismo , Animales , Biomarcadores/metabolismo , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Embrión de Pollo , Modelos Animales de Enfermedad , Embrión no Mamífero/metabolismo , Corazón Fetal/anomalías , Corazón Fetal/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/patología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/fisiopatología , Miocardio/metabolismo , PPAR alfa/agonistas , PPAR alfa/genética , Proliferadores de Peroxisomas/toxicidad , Pirimidinas/toxicidad , Proteínas Smad/metabolismo
6.
Environ Toxicol Chem ; 31(12): 2856-60, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23001933

RESUMEN

The peroxisome proliferator (PP) di(2-ethylhexyl)phthalate (DEHP) is widely used as a plasticizer and can contaminate air, water, and soil. As yet, no data have been published on its potential to induce changes in cell growth of nonmammalian hepatocytes. In the present study, the effects of DEHP on cell turnover and induction of peroxisome proliferation were evaluated in primary hepatocyte cultures from chick embryos. Cells were treated after attachment with 0, 25, 50, 75, and 100 µM DEHP for up to 96 h. S-phase increased significantly (p < 0.01) from a background level of 5.5 ± 0.1% in solvent-control hepatocytes to a maximum level of 7.1 ± 0.1% in cells exposed for 48 h to 100 µM DEHP and decreased to near 6% by 96 h. Lower (p < 0.05) levels of induction were seen at 50 and 75 µM DEHP. Spontaneous apoptosis showed a slight (p < 0.05) decrease in hepatocytes treated with ≥75 µM dosages, as measured at 72 to 96 h. Induction of peroxisome proliferation was observed for cultures treated with ≥75 µM dosages at 48 h onwards. The results of the present study indicate that avian species may be responsive to the effects of PPs and may thus be affected by the presence of DEHP in the environment, but that this species is less sensitive than rodents.


Asunto(s)
Dietilhexil Ftalato/toxicidad , Hepatocitos/fisiología , Proliferadores de Peroxisomas/toxicidad , Peroxisomas/metabolismo , Plastificantes/toxicidad , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Embrión de Pollo , Hepatocitos/efectos de los fármacos
7.
Mutat Res ; 750(2): 141-158, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22198209

RESUMEN

Di(2-ethylhexyl) phthalate (DEHP) is a peroxisome proliferator agent that is widely used as a plasticizer to soften polyvinylchloride plastics and non-polymers. Both occupational (e.g., by inhalation during its manufacture and use as a plasticizer of polyvinylchloride) and environmental (medical devices, contamination of food, or intake from air, water and soil) routes of exposure to DEHP are of concern for human health. There is sufficient evidence for carcinogenicity of DEHP in the liver in both rats and mice; however, there is little epidemiological evidence on possible associations between exposure to DEHP and liver cancer in humans. Data are available to suggest that liver is not the only target tissue for DEHP-associated toxicity and carcinogenicity in both humans and rodents. The debate regarding human relevance of the findings in rats or mice has been informed by studies on the mechanisms of carcinogenesis of the peroxisome proliferator class of chemicals, including DEHP. Important additional mechanistic information became available in the past decade, including, but not limited to, sub-acute, sub-chronic and chronic studies with DEHP in peroxisome proliferator-activated receptor (PPAR) α-null mice, as well as experiments utilizing several transgenic mouse lines. Activation of PPARα and the subsequent downstream events mediated by this transcription factor represent an important mechanism of action for DEHP in rats and mice. However, additional data from animal models and studies in humans exposed to DEHP from the environment suggest that multiple molecular signals and pathways in several cell types in the liver, rather than a single molecular event, contribute to the cancer in rats and mice. In addition, the toxic and carcinogenic effects of DEHP are not limited to liver. The International Agency for Research on Cancer working group concluded that the human relevance of the molecular events leading to cancer elicited by DEHP in several target tissues (e.g., liver and testis) in rats and mice can not be ruled out and DEHP was classified as possibly carcinogenic to humans (Group 2B).


Asunto(s)
Carcinógenos/toxicidad , Dietilhexil Ftalato/toxicidad , Hígado/efectos de los fármacos , Plastificantes/toxicidad , Animales , Humanos , Sistema Inmunológico/efectos de los fármacos , Riñón/efectos de los fármacos , Neoplasias Hepáticas/inducido químicamente , Pulmón/efectos de los fármacos , Masculino , Ratones , Proliferadores de Peroxisomas/toxicidad , Ratas , Neoplasias Testiculares/inducido químicamente , Testículo/efectos de los fármacos
8.
Int J Toxicol ; 29(5): 496-504, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20884859

RESUMEN

Peroxisome proliferator-activated receptor α (PPARα) agonism in ocular inflammation has not been thoroughly investigated. The objective of this investigation was to determine the effect of WY-14 643, a selective PPARα agonist, on inflammatory cytokine release in human ocular cells. Stimulation of primary human corneal epithelial cells, keratocytes, and retinal endothelial cells with 1 to 10 ng/mL interleukin 1ß (IL-1ß) resulted in a significant increase in numerous inflammatory cytokines, including IL-6, IL-8, and tumor necrosis factor α (TNF-α); and dexamethasone was able to significantly inhibit these effects. However, WY-14 643 did not effectively block IL-1ß-induced cytokine release in ocular cells; rather, significant increases in IL-1ß-induced inflammatory cytokines were observed in these cells but not in aortic smooth muscle cells. WY-14 643 also significantly upregulated vascular endothelial growth factor (VEGF) expression in corneal epithelial cells and keratocytes. These studies demonstrate for the first time that PPARα agonism may be proinflammatory and proangiogenic in a variety of ocular cells and suggest that therapeutic applications of such agents in ophthalmology may be limited.


Asunto(s)
Ojo/efectos de los fármacos , Ojo/metabolismo , Mediadores de Inflamación/metabolismo , PPAR alfa/agonistas , Proliferadores de Peroxisomas/toxicidad , Pirimidinas/toxicidad , Inductores de la Angiogénesis/toxicidad , Células Cultivadas , Factores Estimulantes de Colonias/metabolismo , Citocinas/metabolismo , Epitelio Corneal/citología , Epitelio Corneal/efectos de los fármacos , Epitelio Corneal/metabolismo , Ojo/citología , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Interleucinas/metabolismo , Especificidad de Órganos , PPAR alfa/genética , PPAR alfa/metabolismo , ARN Mensajero/metabolismo , Retina/citología , Retina/efectos de los fármacos , Retina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/metabolismo , Factores de Crecimiento Endotelial Vascular/metabolismo
9.
Toxicol Ind Health ; 26(7): 407-16, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20504828

RESUMEN

Peroxisome proliferators (PPs)-induced DNA hypomethylation has been proposed as a mechanism of their toxicity, including carcinogenic action. The effect of di-butyl phthalate (DBP), a known peroxisome proliferators, on the methylation level of the c-myc promoter region in rat liver was studied. Changes in the methylation status of the c-myc gene were correlated with changes in DNA synthesis, DNA methyltransferase (DNMTs) activity and liver weight. Male Wistar rats received DBP in one, three or fourteen daily oral doses of 1800 mg/kg body weight (b.w.) x day(-1) (this dose is close to the dose that increases the numbers of peroxisomes in male Wistar rats). We have demonstrated that DBP decreased the methylation of the c-myc gene. Cytosine hypomethylation in the analyzed CpG sites of the c-myc gene promoter occurred during the whole period of study, although after 14 doses of DBP the difference from control was only on the borderline of significance (p = 0.066). An increase in DNA synthesis was only observed after 24 hours of treatment with DBP, and it preceded liver growth. We hypothesize that DBP-induced demethylation of the c-myc gene was an active mechanism, not associated with DNMTs activity and DNA replication.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Dibutil Ftalato/toxicidad , Genes myc/efectos de los fármacos , Hepatomegalia/inducido químicamente , Hígado/efectos de los fármacos , Animales , Citosina/metabolismo , ADN/biosíntesis , ADN/efectos de los fármacos , ADN/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Hepatomegalia/genética , Hígado/fisiología , Masculino , Proliferadores de Peroxisomas/toxicidad , Regiones Promotoras Genéticas , Ratas , Ratas Wistar
10.
Toxicol Sci ; 113(1): 45-59, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19850644

RESUMEN

Peroxisome proliferator chemicals (PPC) are thought to mediate their effects in rodents on hepatocyte growth and liver cancer through the nuclear receptor peroxisome proliferator-activated receptor (PPAR) alpha. Recent studies indicate that the plasticizer di-(2-ethylhexyl) phthalate (DEHP) increased the incidence of liver tumors in PPARalpha-null mice. We hypothesized that some PPC, including DEHP, induce transcriptional changes independent of PPARalpha but dependent on other nuclear receptors, including the constitutive-activated receptor (CAR) that mediates phenobarbital (PB) effects on hepatocyte growth and liver tumor induction. To determine the potential role of CAR in mediating effects of PPC, a meta-analysis was performed on transcript profiles from published studies in which rats and mice were exposed to PPC and compared the profiles to those produced by exposure to PB. Valproic acid, clofibrate, and DEHP in rat liver and DEHP in mouse liver induced genes, including Cyp2b family members that are known to be regulated by CAR. Examination of transcript changes by Affymetrix ST 1.0 arrays and reverse transcription-PCR in the livers of DEHP-treated wild-type, PPARalpha-null, and CAR-null mice demonstrated that (1) most (approximately 94%) of the transcriptional changes induced by DEHP were PPARalpha-dependent, (2) many PPARalpha-independent genes overlapped with those regulated by PB, (3) induction of genes Cyp2b10, Cyp3a11, and metallothionine-1 by DEHP was CAR dependent but PPARalpha-independent, and (4) induction of a number of genes (Cyp8b1, Gstm4, and Gstm7) was independent of both CAR and PPARalpha. Our results indicate that exposure to PPARalpha activators including DEHP leads to activation of multiple nuclear receptors in the rodent liver.


Asunto(s)
Dietilhexil Ftalato/toxicidad , Hígado/efectos de los fármacos , PPAR alfa/agonistas , Proliferadores de Peroxisomas/toxicidad , Plastificantes/toxicidad , Receptores Citoplasmáticos y Nucleares/agonistas , Animales , Clofibrato/toxicidad , Receptor de Androstano Constitutivo , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , PPAR alfa/genética , PPAR alfa/metabolismo , Fenobarbital/toxicidad , Receptor X de Pregnano , ARN Mensajero/metabolismo , Ratas , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Esteroides/agonistas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética/efectos de los fármacos , Ácido Valproico/toxicidad
11.
Environ Health Perspect ; 117(4): 545-51, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19440492

RESUMEN

BACKGROUND: Perfluoroalkanoates, [e.g., perfluorooctanoate (PFOA)], are known peroxisome proliferators that induce hepatomegaly and hepatocarcinogenesis in rodents, and are classic non-genotoxic carcinogens that inhibit in vitro gap-junctional intercellular communication (GJIC). This inhibition of GJIC is known to be a function of perfluorinated carbon lengths ranging from 7 to 10. OBJECTIVES: The aim of this study was to determine if the inhibition of GJIC by PFOA but not perfluoropentanoate (PFPeA) observed in F344 rat liver cells in vitro also occurs in F344 rats in vivo and to determine mechanisms of PFOA dysregulation of GJIC using in vitro assay systems. METHODS: We used an incision load/dye transfer technique to assess GJIC in livers of rats exposed to PFOA and PFPeA. We used in vitro assays with inhibitors of cell signaling enzymes and antioxidants known to regulate GJIC to identify which enzymes regulated PFOA-induced inhibition of GJIC. RESULTS: PFOA inhibited GJIC and induced hepatomegaly in rat livers, whereas PFPeA had no effect on either end point. Serum biochemistry of liver enzymes indicated no cytotoxic response to these compounds. In vitro analysis of mitogen-activated protein kinase (MAPK) indicated that PFOA, but not PFPeA, can activate the extracellular receptor kinase (ERK). Inhibition of GJIC, in vitro, by PFOA depended on the activation of both ERK and phosphatidylcholine-specific phospholipase C (PC-PLC) in the dysregulation of GJIC in an oxidative-dependent mechanism. CONCLUSIONS: The in vitro analysis of GJIC, an epigenetic marker of tumor promoters, can also predict the in vivo activity of PFOA, which dysregulated GJIC via ERK and PC-PLC.


Asunto(s)
Caprilatos/toxicidad , Comunicación Celular/efectos de los fármacos , Fluorocarburos/toxicidad , Proliferadores de Peroxisomas/toxicidad , Animales , Carcinógenos/toxicidad , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ácidos Grasos Volátiles/toxicidad , Uniones Comunicantes/metabolismo , Hepatocitos/metabolismo , Masculino , Proliferadores de Peroxisomas/química , Ratas , Ratas Endogámicas F344 , Relación Estructura-Actividad , Valeratos/toxicidad
12.
Environ Health Perspect ; 117(11): 1664-72, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20049115

RESUMEN

BACKGROUND: Diverse environmental contaminants, including the plasticizer di(2-ethylhexyl)phthalate (DEHP), are hepatocarcinogenic peroxisome proliferators in rodents. Peroxisome proliferator-activated receptor-alpha (PPAR-alpha) activation and its sequelae have been proposed to constitute a mode of action (MOA) for hepatocarcinogenesis by such agents as a sole causative factor. Further, based on a hypothesized lower sensitivity of humans to this MOA, prior reviews have concluded that rodent hepatocarcinogenesis by PPAR-alpha agonists is irrelevant to human carcinogenic risk. DATA SYNTHESIS: Herein, we review recent studies that experimentally challenge the PPAR-alpha activation MOA hypothesis, providing evidence that DEHP is hepatocarcinogenic in PPAR-alpha-null mice and that the MOA but not hepatocarcinogenesis is evoked by PPAR-alpha activation in a transgenic mouse model. We further examine whether relative potency for PPAR-alpha activation or other steps in the MOA correlates with tumorigenic potency. In addition, for most PPAR-alpha agonists of environmental concern, available data are insufficient to characterize relative human sensitivity to this rodent MOA or to induction of hepatocarcinogenesis. CONCLUSIONS: Our review and analyses raise questions about the hypothesized PPAR-alpha activation MOA as a sole explanation for rodent hepatocarcinogenesis by PPAR-alpha agonists and therefore its utility as a primary basis for assessing human carcinogenic risk from the diverse compounds that activate PPAR-alpha. These findings have broad implications for how MOA hypotheses are developed, tested, and applied in human health risk assessment. We discuss alternatives to the current approaches to these key aspects of mechanistic data evaluation.


Asunto(s)
Contaminantes Ambientales/toxicidad , Neoplasias Hepáticas/inducido químicamente , PPAR alfa/agonistas , Animales , Dietilhexil Ftalato/toxicidad , Humanos , Neoplasias Hepáticas Experimentales/inducido químicamente , Ratones , Ratones Noqueados , PPAR alfa/metabolismo , Proliferadores de Peroxisomas/toxicidad , Medición de Riesgo/métodos , Especificidad de la Especie
13.
Comp Biochem Physiol C Toxicol Pharmacol ; 149(4): 524-30, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19101656

RESUMEN

Perfluorooctane sulfonate (PFOS) is a widely distributed industrial compound that has been detected in the eggs of various wild avian species. Laboratory studies have indicated that PFOS is embryotoxic to domestic chickens (Gallus gallus domesticus), but the mechanisms of toxicity in the developing avian embryo remain unknown. We recently demonstrated that PFOS acts as a peroxisome proliferator by causing increased expression of peroxisome proliferator activated receptor alpha (PPARalpha)-regulated genes in cultured primary chicken embryo hepatocytes. The present study examined whether PPARalpha-regulated genes were dose-dependently affected in chicken embryos exposed in ovo to PFOS. White leghorn chicken eggs were injected with 0.1, 5.0 or 100.0 microg PFOS/g egg into the air cell prior to incubation. Embryos were incubated until pipping, after which the expression of PPARalpha-regulated genes was measured in the liver tissue of surviving embryos using real-time reverse transcription polymerase chain reaction. A dose-dependent decrease in embryo pippability was observed with an LD50 of 93 microg/g (3.54 microg/g-672,910 microg/g, 95% confidence interval). Hepatic PFOS concentrations increased concomitantly with dose. The PPARalpha-regulated genes measured were peroxisomal acyl CoA oxidase, bifunctional enzyme, liver fatty acid binding protein and peroxisomal 3-ketoacyl thiolase. PFOS exposure via egg injection prior to incubation did not affect the transcriptional activity of any of the assayed PPARalpha-regulated genes at any of the doses examined in day 21 chicken embryos.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Embrión de Pollo/efectos de los fármacos , Fluorocarburos/toxicidad , PPAR alfa/metabolismo , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Acetil-CoA C-Aciltransferasa/metabolismo , Acil-CoA Oxidasa , Animales , Enoil-CoA Hidratasa/metabolismo , Proteínas de Unión a Ácidos Grasos , Isomerasas/metabolismo , Complejos Multienzimáticos/metabolismo , Oxidorreductasas/metabolismo , Enzima Bifuncional Peroxisomal , Proliferadores de Peroxisomas/toxicidad , Peroxisomas/enzimología
14.
Methods Mol Biol ; 410: 123-35, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18642598

RESUMEN

Pollution in aquatic environment is of increasing concern for its impact on both human and natural populations. Applying proteomics to monitor marine pollution is a new approach to evaluate the effects of environmental pollutants on the biota. Aquatic organisms living in coastal and estuarine areas are particularly prone to exposures to a variety of pollutants, some of which can act as peroxisome proliferators. However, peroxisomal responses in particular and biomarker responses in general can be influenced by several biotic and abiotic factors. Utilizing proteomics-based techniques that permit the evaluation of hundreds to thousands of proteins in a single experiment can circumvent those drawbacks. Applying this method, the peroxisomal proteome from digestive glands of mussels Mytilus sp. can be analyzed by two-dimensional electrophoresis (2-DE) and the 2-DE maps from control samples and samples obtained in a polluted area can be compared. The up- and down-regulated proteins compose the protein expression signature (PES) associated with exposure to peroxisome proliferating pollutants. This method generates highly reproducible patterns that can be applied to laboratory or field experiments.


Asunto(s)
Sustancias Peligrosas/toxicidad , Mytilus/efectos de los fármacos , Proliferadores de Peroxisomas/toxicidad , Peroxisomas/metabolismo , Proteómica/métodos , Animales , Electroforesis en Gel Bidimensional , Contaminación Ambiental/análisis , Contaminación Ambiental/prevención & control , Biología Marina , Mytilus/metabolismo , Medición de Riesgo/métodos
15.
Mutat Res ; 644(1-2): 17-23, 2008 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-18639561

RESUMEN

Genomic hypomethylation is a consistent finding in both human and animal tumors and mounting experimental evidence suggests a key role for epigenetic events in tumorigenesis. Furthermore, it has been suggested that early changes in DNA methylation and histone modifications may serve as sensitive predictive markers in animal testing for carcinogenic potency of environmental agents. Alterations in metabolism of methyl donors, disturbances in activity and/or expression of DNA methyltransferases, and presence of DNA single-strand breaks could contribute to the loss of cytosine methylation during carcinogenesis; however, the precise mechanisms of genomic hypomethylation induced by chemical carcinogens remain largely unknown. This study examined the mechanism of DNA hypomethylation during hepatocarcinogenesis induced by peroxisome proliferators WY-14,643 (4-chloro-6-(2,3-xylidino)-pyrimidynylthioacetic acid) and DEHP (di-(2-ethylhexyl)phthalate), agents acting through non-genotoxic mode of action. In the liver of male Fisher 344 rats exposed to WY-14,643 (0.1% (w/w), 5 months), the level of genomic hypomethylation increased by approximately 2-fold, as compared to age-matched controls, while in the DEHP group (1.2% (w/w), 5 months) DNA methylation did not change. Global DNA hypomethylation in livers from WY-14,643 group was accompanied by the accumulation of DNA single-strand breaks, increased cell proliferation, and diminished expression of DNA methyltransferase 1, while the metabolism of methyl donors was not affected. In contrast, none of these parameters changed significantly in rats fed DEHP. Since WY-14,643 is much more potent carcinogen than DEHP, we conclude that the extent of loss of DNA methylation may be related to the carcinogenic potential of the chemical agent, and that accumulation of DNA single-strand breaks coupled to the increase in cell proliferation and altered DNA methyltransferase expression may explain genomic hypomethylation during peroxisome proliferator-induced carcinogenesis.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Proliferadores de Peroxisomas/toxicidad , Animales , Proliferación Celular/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Roturas del ADN de Cadena Simple , Dietilhexil Ftalato/toxicidad , Histonas/química , Histonas/metabolismo , Masculino , Metilación , Mutágenos/toxicidad , Antígeno Nuclear de Célula en Proliferación/metabolismo , Pirimidinas/toxicidad , Ratas , Ratas Endogámicas F344
16.
Toxicol Lett ; 178(2): 110-6, 2008 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-18407439

RESUMEN

Di-isodecyl phthalate (DIDP), a peroxisome proliferator-activated receptor-alpha activator, is widely used as a plasticizer in the manufacture of polyvinyl chloride (PVC), and ultimately in typical vinyl applications, particularly wire, cable and toys, etc. To examine its carcinogenic potential, DIDP was fed to Fischer 344 rats in the diet at doses of 0, 400, 2000 and 8000 ppm for 2 years. Briefly, significant decreases in the overall survival and body weights, and increases in the relative weights of kidneys and liver were noted in both sexes of the highest dose groups. However, no treatment-related neoplastic lesions were observed in the internal organs, including the liver. Unlike di(2-ethylhexyl) phthalate (DEHP), DIDP failed to maintain the catalase-inducing potential between early and late expressions of catalase protein from western blotting, immunohistochemistry and enzyme activity measurements. These results suggest that the non-carcinogenicity of DIDP in F344 rats was due to its limited potential for peroxisomal proliferating activity.


Asunto(s)
Carcinógenos , Proliferadores de Peroxisomas/toxicidad , Ácidos Ftálicos/toxicidad , Animales , Western Blotting , Peso Corporal/efectos de los fármacos , Pruebas de Carcinogenicidad , Catalasa/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Femenino , Inmunohistoquímica , Masculino , Neoplasias/inducido químicamente , Neoplasias/epidemiología , Tamaño de los Órganos/efectos de los fármacos , Ratas , Ratas Endogámicas F344 , Caracteres Sexuales , Análisis de Supervivencia
17.
Toxicology ; 246(1): 2-8, 2008 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-18006136

RESUMEN

Peroxisome proliferator chemicals are classic non-genotoxic carcinogens. These agents cause liver cancers when chronically administered to rats and mice. Peroxisome proliferators include the widely prescribed lipid and cholesterol lowering fibrate drugs. In contrast to the results in rodents, there is no evidence that fibrates are associated with elevated risk of liver cancer or any other neoplasms in humans thus indicating a species difference in the hepatocarcinogenic response. The biological effects of peroxisome proliferators are mediated by the peroxisome proliferator-activated receptor (PPAR)alpha. Pparalpha-null mice are resistant to all of the pleiotropic effects of peroxisome proliferators, including cell proliferation and hepatocarcinogenesis. The mechanism of hepatocellular proliferation involves downregulation of the microRNA let-7c gene by PPARalpha. Let-7c controls levels of proliferative c-myc by destabilizing its mRNA. Thus, upon suppression of let-7c, c-myc mRNA and protein are elevated resulting in enhanced hepatocellular proliferation. In contrast, PPARalpha-humanized mice, that respond to Wy-14,643 by lower serum triglycerides and induction of genes encoding fatty acid metabolizing enzymes, are resistant to peroxisome proliferator-induced cell proliferation and cancer. These mice do not exhibit downregulation of let-7c gene expression thus forming the basis for the resistance to hepatocellular carcinogenesis.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Proteínas de Unión al ADN/metabolismo , PPAR alfa/toxicidad , Proliferadores de Peroxisomas/toxicidad , Pirimidinas/toxicidad , Factores de Transcripción/metabolismo , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Regulación Neoplásica de la Expresión Génica , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , PPAR alfa/genética , PPAR alfa/fisiología , Proliferadores de Peroxisomas/metabolismo , Pirimidinas/metabolismo , alfa-Amilasas Salivales , Especificidad de la Especie
18.
Pharmacogenomics ; 8(11): 1521-34, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18034617

RESUMEN

INTRODUCTION: To reduce continuously increasing costs in drug development, adverse effects of drugs need to be detected as early as possible in the process. In recent years, compound-induced gene expression profiling methodologies have been developed to assess compound toxicity, including Gene Ontology term and pathway over-representation analyses. The objective of this study was to introduce an additional approach, in which literature information is used for compound profiling to evaluate compound toxicity and mode of toxicity. METHODS: Gene annotations were built by text mining in Medline abstracts for retrieval of co-publications between genes, pathology terms, biological processes and pathways. This literature information was used to generate compound-specific keyword fingerprints, representing over-represented keywords calculated in a set of regulated genes after compound administration. To see whether keyword fingerprints can be used for assessment of compound toxicity, we analyzed microarray data sets of rat liver treated with 11 hepatotoxicants. RESULTS: Analysis of keyword fingerprints of two genotoxic carcinogens, two nongenotoxic carcinogens, two peroxisome proliferators and two randomly generated gene sets, showed that each compound produced a specific keyword fingerprint that correlated with the experimentally observed histopathological events induced by the individual compounds. By contrast, the random sets produced a flat aspecific keyword profile, indicating that the fingerprints induced by the compounds reflect biological events rather than random noise. A more detailed analysis of the keyword profiles of diethylhexylphthalate, dimethylnitrosamine and methapyrilene (MPy) showed that the differences in the keyword fingerprints of these three compounds are based upon known distinct modes of action. Visualization of MPy-linked keywords and MPy-induced genes in a literature network enabled us to construct a mode of toxicity proposal for MPy, which is in agreement with known effects of MPy in literature. CONCLUSION: Compound keyword fingerprinting based on information retrieved from literature is a powerful approach for compound profiling, allowing evaluation of compound toxicity and analysis of the mode of action.


Asunto(s)
Carcinógenos/toxicidad , Bases de Datos Bibliográficas , Perfilación de la Expresión Génica , Mutágenos/toxicidad , Proliferadores de Peroxisomas/toxicidad , Toxicogenética/métodos , Algoritmos , Animales , Bases de Datos Genéticas , Hígado/efectos de los fármacos , MEDLINE , Procesamiento de Lenguaje Natural , Ratas , Vocabulario Controlado
19.
Toxicol Appl Pharmacol ; 225(3): 267-77, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17950772

RESUMEN

Administration of peroxisome proliferators to rodents causes proliferation of peroxisomes, induction of beta-oxidation enzymes, hepatocellular hypertrophy and hyperplasia, with chronic exposure ultimately leading to hepatocellular carcinomas. Many responses associated with peroxisome proliferators are nuclear receptor-mediated events involving peroxisome proliferators-activated receptor alpha (PPARalpha). A role for nuclear receptor-independent events has also been shown, with evidence of Kupffer cell-mediated free radical production, presumably through NAPDH oxidase, induction of redox-sensitive transcription factors involved in cytokine production and cytokine-mediated cell replication following acute treatment with peroxisome proliferators in rodents. Recent studies have demonstrated, by using p47(phox)-null mice which are deficient in NADPH oxidase, that this enzyme is not related to the phenotypic events caused by prolonged administration of peroxisome proliferators. In an effort to determine the timing of the transition from Kupffer cell-to PPARalpha-dependent modulation of peroxisome proliferator effects, gene expression was assessed in liver from Pparalpha-null, p47(phox)-null and corresponding wild-type mice following treatment with 4-chloro-6-(2,3-xylidino)-pyrimidynylthioacetic acid (WY-14,643) for 8 h, 24 h, 72 h, 1 week or 4 weeks. WY-14,643-induced gene expression in p47(phox)-null mouse liver differed substantially from wild-type mice at acute doses and striking differences in baseline expression of immune related genes were evident. Pathway mapping of genes that respond to WY-14,643 in a time- and dose-dependent manner demonstrates suppression of immune response, cell death and signal transduction and promotion of lipid metabolism, cell cycle and DNA repair. Furthermore, these pathways were largely dependent on PPARalpha, not NADPH oxidase demonstrating a temporal shift in response to peroxisome proliferators. Overall, this study shows that NADPH oxidase-dependent events, while detectable following acute treatment, are transient. To the contrary, a strong PPARalpha-specific gene signature was evident in mice that were continually exposed to WY-14,643.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Macrófagos del Hígado/efectos de los fármacos , Hígado/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , PPAR alfa/efectos de los fármacos , Proliferadores de Peroxisomas/toxicidad , Pirimidinas/toxicidad , Animales , Perfilación de la Expresión Génica , Macrófagos del Hígado/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , NADPH Oxidasas/metabolismo , PPAR alfa/metabolismo , Factores de Tiempo
20.
Mutat Res ; 625(1-2): 62-71, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17586532

RESUMEN

Peroxisome proliferators are potent rodent liver carcinogens that act via a non-genotoxic mechanism. The mode of action of these agents in rodent liver includes increased cell proliferation, decreased apoptosis, secondary oxidative stress and other events; however, it is not well understood how peroxisome proliferators are triggering the plethora of the molecular signals leading to cancer. Epigenetic changes have been implicated in the mechanism of liver carcinogenesis by a number of environmental agents. Short-term treatment with peroxisome proliferators and other non-genotoxic carcinogens leads to global and locus-specific DNA hypomethylation in mouse liver, events that were suggested to correlate with a burst of cell proliferation. In the current study, we investigated the effects of long-term exposure to a model peroxisome proliferator WY-14,643 on DNA and histone methylation. Male SV129mice were fed a control or WY-14,643-containing (1000ppm) diet for one week, five weeks or five months. Treatment with WY-14,643 led to progressive global hypomethylation of liver DNA as determined by an HpaII-based cytosine extension assay with the maximum effect reaching over 200% at five months. Likewise, trimethylation of histone H4 lysine 20 and H3 lysine 9 was significantly decreased at all time points. The majority of cytosine methylation in mammals resides in repetitive DNA sequences. In view of this, we measured the effect of WY-14,643 on the methylation status of major and minor satellites, as well as in IAP, LINE1 and LINE2 elements in liver DNA. Exposure to WY-14,643 resulted in a gradual loss of cytosine methylation in major and minor satellites, IAP, LINE1 and LINE2 elements. The epigenetic changes correlated with the temporal effects of WY-14,643 on cell proliferation rates in liver, but no sustained effect on c-Myc promoter methylation was observed. Finally, WY-14,643 had no effect on DNA and histone methylation status in Pparalpha-null mice at any of the time points considered in this study. These data indicate the importance of epigenetic alterations in the mechanism of action of peroxisome proliferators and the key role of Pparalpha.


Asunto(s)
Epigénesis Genética/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , PPAR alfa/metabolismo , Proliferadores de Peroxisomas/toxicidad , Pirimidinas/toxicidad , Animales , Secuencia de Bases , Carcinógenos/toxicidad , Islas de CpG/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Cartilla de ADN/genética , Epigénesis Genética/fisiología , Histonas/metabolismo , Elementos de Nucleótido Esparcido Largo/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , PPAR alfa/deficiencia , PPAR alfa/genética , Retroelementos/efectos de los fármacos , Secuencias Repetidas Terminales/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA