Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Photochem Photobiol Sci ; 12(4): 678-83, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23080056

ABSTRACT

In this work, the photocatalytic degradation of selected estrogens (E2 and EE2) was evaluated, using bench-scale and continuous treatment systems assisted by artificial UV-A and solar radiation. Processes based on the use of TiO2 permit an efficient degradation of E2 and EE2 estrogens, usually at reaction times lower than 15 min. Especially remarkable is the high degradation efficiency shown by sunlight-assisted processes, which are extremely favored by the high efficiency of compound parabolic collectors.


Subject(s)
Estradiol/chemistry , Light , Lynestrenol/chemistry , Titanium/chemistry , Ultraviolet Rays , Zinc Oxide/chemistry , Catalysis , Hydrogen-Ion Concentration , Photolysis
2.
J Steroid Biochem Mol Biol ; 110(1-2): 56-66, 2008 May.
Article in English | MEDLINE | ID: mdl-18356043

ABSTRACT

This study examined the cytochrome P450 (CYP) enzyme selectivity of in vitro bioactivation of lynestrenol to norethindrone and the further metabolism of norethindrone. Screening with well-established chemical inhibitors showed that the formation of norethindrone was potently inhibited by CYP3A4 inhibitor ketoconazole (IC(50)=0.02 microM) and with CYP2C9 inhibitor sulphaphenazole (IC(50)=2.13 microM); the further biotransformation of norethindrone was strongly inhibited by ketoconazole (IC(50)=0.09 microM). Fluconazole modestly inhibited both lynestrenol bioactivation and norethindrone biotransformation. Lynestrenol bioactivation was mainly catalysed by recombinant human CYP2C9, CYP2C19 and CYP3A4; rCYP3A4 was responsible for the hydroxylation of norethindrone. A significant correlation was observed between norethindrone formation and tolbutamide hydroxylation, a CYP2C9-selective activity (r=0.63; p=0.01). Norethindrone hydroxylation correlated significantly with model reactions of CYP2C19 and CYP3A4. The greatest immunoinhibition of lynestrenol bioactivation was seen in incubations with CYP2C-Ab. The CYP3A4-Ab reduced norethindrone hydroxylation by 96%. Both lynestrenol and norethindrone were weak inhibitors of CYP2C9 (IC(50) of 32 microM and 46 microM for tolbutamide hydroxylation, respectively). In conclusion, CYP2C9, CYP2C19 and CYP3A4 are the primary cytochromes in the bioactivation of lynestrenol in vitro, while CYP3A4 catalyses the further metabolism of norethindrone.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Cytochrome P-450 Enzyme System/metabolism , Lynestrenol/pharmacokinetics , Norethindrone/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Biotransformation/drug effects , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors , Cytochrome P-450 Enzyme Inhibitors , Fluconazole/pharmacology , Humans , Hydroxylation/drug effects , Ketoconazole/pharmacology , Lynestrenol/chemistry , Lynestrenol/metabolism , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Norethindrone/chemistry , Norethindrone/metabolism , Sulfaphenazole/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...