PT - JOURNAL ARTICLE AU - Choon-myung Lee AU - Bang-sub Lee AU - Samuel L. Arnold AU - Nina Isoherranen AU - Edward T. Morgan TI - Nitric Oxide and Interleukin-1<em>β</em> Stimulate the Proteasome-Independent Degradation of the Retinoic Acid Hydroxylase CYP2C22 in Primary Rat Hepatocytes AID - 10.1124/jpet.113.209841 DP - 2014 Jan 01 TA - Journal of Pharmacology and Experimental Therapeutics PG - 141--152 VI - 348 IP - 1 4099 - http://jpet.aspetjournals.org/content/348/1/141.short 4100 - http://jpet.aspetjournals.org/content/348/1/141.full SO - J Pharmacol Exp Ther2014 Jan 01; 348 AB - CYP2C22 was recently described as a retinoic acid–metabolizing cytochrome P450 enzyme whose transcription is induced by all-trans-retinoic acid (atRA) in hepatoma cells (Qian L, Zolfaghari R, and Ross AC (2010) J Lipid Res 51:1781–1792). We identified CYP2C22 as a putative nitric oxide (NO)–regulated protein in a proteomic screen and raised specific polyclonal antibodies to CYP2C22 to study its protein expression. We found that CYP2C22 is a liver-specific protein that was not significantly induced by activators of the pregnane X receptor, constitutive androstane receptor, or peroxisome proliferator-activated receptor-α, but was downregulated to &lt;25% of control by the aryl hydrocarbon receptor agonist β-naphthoflavone in cultured rat hepatocytes. CYP2C22 protein and its mRNA both were induced by atRA in hepatocytes, with EC50 of 100–300 nM, whereas the maximal extent of mRNA induction was twice that of the protein. CYP2C22 protein, but not its mRNA, was rapidly downregulated in hepatocytes by interleukin-1 (IL-1) or NO-donating compounds, and the downregulation by IL-1 was blocked by inhibition of NO synthases. The NO donor (Z)-1-[N-(3-aminopropyl)-N-(3-ammoniopropyl)amino]diazen-1-ium-1,2-diolate reduced the half-life of CYP2C22 from 8.7 to 3.4 hours in the presence of cycloheximide, demonstrating that NO-dependent downregulation is due to stimulated proteolysis. No intermediate degradation products were detected. However, this degradation was insensitive to inhibitors of calpains or the canonical proteasomal or lysosomal pathways, indicating that NO-dependent degradation of CYP2C22 proceeds via a novel pathway.