A SUMO-acetyl switch in PXR biology

W Cui, M Sun, S Zhang, X Shen, N Galeva… - … et Biophysica Acta (BBA …, 2016 - Elsevier
W Cui, M Sun, S Zhang, X Shen, N Galeva, TD Williams, JL Staudinger
Biochimica et Biophysica Acta (BBA)-Gene Regulatory Mechanisms, 2016Elsevier
Post-translational modification (PTM) of nuclear receptor superfamily members regulates
various aspects of their biology to include sub-cellular localization, the repertoire of protein-
binding partners, as well as their stability and mode of degradation. The nuclear receptor
pregnane X receptor (PXR, NR1I2) is a master-regulator of the drug-inducible gene
expression in liver and intestine. The PXR-mediated gene activation program is primarily
recognized to increase drug metabolism, drug transport, and drug efflux pathways in these …
Abstract
Post-translational modification (PTM) of nuclear receptor superfamily members regulates various aspects of their biology to include sub-cellular localization, the repertoire of protein-binding partners, as well as their stability and mode of degradation. The nuclear receptor pregnane X receptor (PXR, NR1I2) is a master-regulator of the drug-inducible gene expression in liver and intestine. The PXR-mediated gene activation program is primarily recognized to increase drug metabolism, drug transport, and drug efflux pathways in these tissues. The activation of PXR also has important implications in significant human diseases including inflammatory bowel disease and cancer. Our recent investigations reveal that PXR is modified by multiple PTMs to include phosphorylation, SUMOylation, and ubiquitination. Using both primary cultures of hepatocytes and cell-based assays, we show here that PXR is modified through acetylation on lysine residues. Further, we show that increased acetylation of PXR stimulates its increased SUMO-modification to support active transcriptional suppression. Pharmacologic inhibition of lysine de-acetylation using trichostatin A (TSA) alters the sub-cellular localization of PXR in cultured hepatocytes, and also has a profound impact upon PXR transactivation capacity. Both the acetylation and SUMOylation status of the PXR protein is affected by its ability to associate with the lysine de-acetylating enzyme histone de-acetylase (HDAC)3 in a complex with silencing mediator of retinoic acid and thyroid hormone receptor (SMRT). Taken together, our data support a model in which a SUMO-acetyl ‘switch’ occurs such that acetylation of PXR likely stimulates SUMO-modification of PXR to promote the active repression of PXR-target gene expression. This article is part of a Special Issue entitled: Xenobiotic nuclear receptors: New Tricks for An Old Dog, edited by Dr. Wen Xie.
Elsevier