Abstract

Inflammation and infection cause the down-regulation of cytochrome P450 enzymes in rodent and human hepatocytes. Although many of these effects are achieved via transcriptional suppression, we have found that CYP2B1 protein is rapidly down-regulated in primary cultures of rat hepatocytes by a NO-dependent mechanism that is independent of any transcriptional effects (Ferrari et al., 2001, Mol. Pharmacol. 60, 209-216). Here, we show that CYP2B proteins in primary rat hepatocyte cultures were suppressed more than 70% after 8hr treatment with interleukin-1 (IL-1β), in an NO-dependent manner. The calpain inhibtor E64-d or the lysosomal protease inhibitors NH4Cl and chloroquine did not attenuate the down-regulation of CYP2B by IL-1, but treatment with the proteasome inhibitors MG132, clasto-Lactacystin β-lactone, or gliotoxin each abolished this suppression under conditions in which NO production was not affected. However, CYP2C11 was not down-regulated by IL-1 treatment and was not affected by the treatments of lysosomal, calpain, and proteasome inhibitors. Treatment of cells for 24 h with the NO-donors 3,3-Bis(aminoethyl)-1-hydroxy-2-oxo-1-triazene (NOC-18), S-nitrosoglutathione, and S-nitroso N-acetylpenicillamine (SNAP) also suppressed CYP2B proteins. Treatment of HeLa cells with 500 μM NOC-18 enhanced the ubiquitination of c-myc-tagged CYP2B at 6 hr. These data suggest that NO-dependent CYP2B down-regulation results from proteasomal degradation through the ubiquitination of CYP2B. (This research was supported by NIH Grant GM 069971)

Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call