Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Environ Toxicol Chem ; 37(11): 2912-2918, 2018 11.
Article in English | MEDLINE | ID: mdl-30125981

ABSTRACT

Microplastics are an abundant pollutant in aquatic systems, but little is known regarding their effects on larval fish. We conducted foraging and growth experiments to observe how increasing densities of microplastics (polyethylene microspheres) impact the foraging and growth of Pimephales promelas larvae. We found minimal impacts on larval consumption of Artemia nauplii in the consumption study, as well as little impact on total length after 30 d of the growth experiment. Environ Toxicol Chem 2018;37:2912-2918. © 2018 SETAC.


Subject(s)
Cyprinidae/growth & development , Feeding Behavior/drug effects , Plastics/toxicity , Animals , Artemia/drug effects , Larva/drug effects , Larva/growth & development , Water Pollutants, Chemical/analysis
2.
Toxicol Sci ; 92(1): 33-41, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16611625

ABSTRACT

Gender is an important factor in pharmacokinetics and pharmacodynamics. In the current study, gender difference in acetaminophen (APAP)-induced hepatotoxicity has been examined. Male and female mice were injected with a toxic dose of APAP (500 mg/kg, ip). Female mice were resistant to the hepatotoxic effects of APAP, depicted by serum alanine aminotransferase and sorbital dehydrogenase activities and histological analysis. Basal hepatic reduced glutathione (GSH) levels were lower in females than in males, suggesting that basal GSH level may not be a factor in determining the gender difference of APAP hepatotoxicity. APAP metabolism was slower in females than males, revealed by lower levels of glucuronidation and sulfation and higher amounts of free APAP in the livers of female mice. Lower basal Cyp1a2 mRNA levels and lower expression of Cyp1a2 and Cyp3a11 mRNAs after APAP dosing were also observed in females compared with males. However, there was no gender difference in N-acetyl-p-benzoquinone imine covalent binding 2 h after APAP administration, suggesting similar APAP bioactivation between genders. Moreover, liver Gst pi mRNA levels were significantly lower in females than males. This finding is consistent with a previous report, which showed that Gst pi knockout mice are protected from APAP-induced liver toxicity. In conclusion, gender difference of APAP-induced hepatotoxicity is not likely due to APAP metabolism. Perhaps, it is in part due to gender-dependent Gst pi expression. However, the mechanism underlying the association between reduction in Gst pi expression and hepatoprotective effect against APAP toxicity remains to be further explored.


Subject(s)
Acetaminophen/pharmacokinetics , Acetaminophen/toxicity , Liver/drug effects , Animals , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP3A/genetics , Female , Glutathione/metabolism , Heme Oxygenase (Decyclizing)/genetics , Liver/enzymology , Liver/metabolism , Male , Membrane Proteins/genetics , Mice , Peroxidases/genetics , Peroxiredoxins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sex Factors
3.
Mol Pharmacol ; 68(6): 1590-6, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16157696

ABSTRACT

Nuclear receptors, including constitutive androstane receptor, pregnane X receptor, and retinoid X receptor (RXR), modulate acetaminophen (APAP)-induced hepatotoxicity by regulating the expression of phase I cytochrome P450 (P450) genes. It has not been fully resolved, however, whether they regulate APAP detoxification at the phase II level. The aim of the current study was to evaluate the role of RXRalpha in phase II enzyme-mediated detoxification of APAP. Wild-type and hepatocyte-specific RXRalpha knockout mice were treated with a toxic dose of APAP (500 mg/kg i.p.). Mutant mice were protected from APAP-induced hepatotoxicity, even though basal liver glutathione (GSH) levels were significantly lower in mutant mice compared with those of wild-type mice. High-performance liquid chromatography analysis of APAP metabolites revealed significantly greater levels of APAP-GSH conjugates in livers and bile of mutant mice compared with those of wild-type mice. Furthermore, hepatocyte RXRalpha deficiency altered the gene expression profile of the glutathione S-transferase (Gst) family. Basal expression of 13 of 15 Gst genes studied was altered in hepatocyte-specific RXRalpha-deficient mice. This probably led to enhanced APAP-GSH conjugation and reduced accumulation of N-acetyl-p-benzoquinone imine, a toxic electrophile that is produced by biotransformation of APAP by phase I P450 enzymes. In conclusion, the data presented in this study define an RXRalpha-Gst regulatory network that controls APAP-GSH conjugation. This report reveals a potential novel strategy to enhance the detoxification of APAP or other xenobiotics by manipulating Gst activity through RXRalpha-mediated pathways.


Subject(s)
Acetaminophen/pharmacokinetics , Gene Expression Regulation , Glutathione Transferase/genetics , Glutathione/metabolism , Liver/metabolism , Retinoid X Receptor alpha/physiology , Animals , Chromatography, High Pressure Liquid , Liver/chemistry , Metabolic Detoxication, Phase II , Mice , Mice, Knockout , Retinoid X Receptor alpha/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...