Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 30
1.
Cells ; 13(8)2024 Apr 14.
Article En | MEDLINE | ID: mdl-38667296

This review explores the likely clinical impact of Pregnane X Receptor (PXR) activation by vitamin K on human health. PXR, initially recognized as a master regulator of xenobiotic metabolism in liver, emerges as a key regulator influencing intestinal homeostasis, inflammation, oxidative stress, and autophagy. The activation of PXR by vitamin K highlights its role as a potent endogenous and local agonist with diverse clinical implications. Recent research suggests that the vitamin K-mediated activation of PXR highlights this vitamin's potential in addressing pathophysiological conditions by promoting hepatic detoxification, fortifying gut barrier integrity, and controlling pro-inflammatory and apoptotic pathways. PXR activation by vitamin K provides an intricate association with cancer cell survival, particularly in colorectal and liver cancers, to provide new insights into potential novel therapeutic strategies. Understanding the clinical implications of PXR activation by vitamin K bridges molecular mechanisms with health outcomes, further offering personalized therapeutic approaches for complex diseases.


Pregnane X Receptor , Signal Transduction , Vitamin K , Humans , Clinical Relevance , Health , Pregnane X Receptor/metabolism , Vitamin K/metabolism
2.
Cells ; 10(11)2021 11 22.
Article En | MEDLINE | ID: mdl-34831484

Highly expressed in the enterohepatic system, pregnane X receptor (PXR, NR1I2) is a well-characterized nuclear receptor (NR) that regulates the expression of genes in the liver and intestines that encode key drug metabolizing enzymes and drug transporter proteins in mammals. The net effect of PXR activation is to increase metabolism and clear drugs and xenobiotics from the body, producing a protective effect and mediating clinically significant drug interaction in patients on combination therapy. The complete understanding of PXR biology is thus important for the development of safe and effective therapeutic strategies. Furthermore, PXR activation is now known to specifically transrepress the inflammatory- and nutrient-signaling pathways of gene expression, thereby providing a mechanism for linking these signaling pathways together with enzymatic drug biotransformation pathways in the liver and intestines. Recent research efforts highlight numerous post-translational modifications (PTMs) which significantly influence the biological function of PXR. However, this thrust of research is still in its infancy. In the context of gene-environment interactions, we present a review of the recent literature that implicates PXR PTMs in regulating its clinically relevant biology. We also provide a discussion of how these PTMs likely interface with each other to respond to extracellular cues to appropriately modify PXR activity.


Pregnane X Receptor/metabolism , Signal Transduction , Animals , Humans , Models, Biological , Protein Processing, Post-Translational
3.
Cells ; 9(10)2020 10 15.
Article En | MEDLINE | ID: mdl-33076284

Pregnane X receptor (PXR, NR1I2) is a member of the ligand-activated nuclear receptor superfamily. This receptor is promiscuous in its activation profile and is responsive to a broad array of both endobiotic and xenobiotic ligands. PXR is involved in pivotal cellular detoxification processes to include the regulation of genes that encode key drug-metabolizing cytochrome-P450 enzymes, oxidative stress response, as well as enzymes that drive steroid and bile acid metabolism. While PXR clearly has important regulatory roles in the liver and gastrointestinal tract, this nuclear receptor also has biological functions in breast tissue. In this review, we highlight current knowledge of PXR's role in mammary tumor carcinogenesis. The elevated level of PXR expression in cancerous breast tissue suggests a likely interface between aberrant cell division and xeno-protection in cancer cells. Moreover, PXR itself exerts positive effect on the cell cycle, thereby predisposing tumor cells to unchecked proliferation. Activation of PXR also plays a key role in regulating apoptosis, as well as in acquired resistance to chemotherapeutic agents. The repressive role of PXR in regulating inflammatory mediators along with the existence of genetic polymorphisms within the sequence of the PXR gene may predispose individuals to developing breast cancer. Further investigations into the role that PXR plays in driving tumorigenesis are needed.


Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinogenesis , Gene Expression Regulation, Neoplastic , Pregnane X Receptor/physiology , Cell Cycle , Cell Division , Drug Therapy , Female , Humans , Inflammation , Polymorphism, Genetic
4.
J Pharmacol Exp Ther ; 373(3): 370-380, 2020 06.
Article En | MEDLINE | ID: mdl-32205367

The pregnane X receptor (PXR), or nuclear receptor (NR) 1I2, is a ligand-activated NR superfamily member that is enriched in liver and intestine in mammals. Activation of PXR regulates the expression of genes encoding key proteins involved in drug metabolism, drug efflux, and drug transport. Recent mechanistic investigations reveal that post-translational modifications (PTMs), such as phosphorylation, play a critical role in modulating the bimodal function of PXR-mediated transrepression and transactivation of target gene transcription. Upon ligand binding, PXR undergoes a conformational change that promotes dissociation of histone deacetylase-containing multiprotein corepressor protein complexes while simultaneously favoring recruitment histone acetyl transferase-containing complexes. Here we describe a novel adenoviral vector used to deliver and recover recombinant human PXR protein from primary cultures of hepatocytes. Using liquid chromatography and tandem mass spectrometry we report here that PXR is phosphorylated at amino acid residues threonine 135 (T135) and serine 221 (S221). Biochemical analysis reveals that these two residues play an important regulatory role in the cycling of corepressor and coactivator multiprotein complexes. These data further our foundational knowledge regarding the specific role of PTMs, namely phosphorylation, in regulating the biology of PXR. Future efforts are focused on using the novel tools described here to identify additional PTMs and protein partners of PXR in primary cultures of hepatocytes, an important experimental model system. SIGNIFICANCE STATEMENT: Pregnane X receptor (PXR), or nuclear receptor 1I2, is a key master regulator of drug-inducible CYP gene expression in liver and intestine in mammals. The novel biochemical tools described in this study demonstrate for the first time that in cultures of primary hepatocytes, human PXR is phosphorylated at amino acid residues threonine 135 (T135) and serine 221 (S221). Moreover, phosphorylation of PXR promotes the transrepression of its prototypical target gene CYP3A4 through modulating its interactions with coregulatory proteins.


Phosphorylation/physiology , Pregnane X Receptor/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Hepatocytes/metabolism , Humans , Mice , Protein Processing, Post-Translational/physiology , Rats , Rats, Sprague-Dawley , Serine/metabolism , Threonine/metabolism
5.
Mol Cell Endocrinol ; 485: 61-71, 2019 04 05.
Article En | MEDLINE | ID: mdl-30726709

The canonical effect of Pregnane X Receptor (PXR, NR1I2) agonism includes enhanced hepatic uptake and a concomitant increase in the first-pass metabolism and efflux of drugs in mammalian liver and intestine. In patients undergoing combination therapy, PXR-mediated gene regulation represents the molecular basis of numerous food-drug, herb-drug, and drug-drug interactions. Moreover, PXR activation promotes chemotherapeutic resistance in certain malignancies. Additional research efforts suggest that sustained PXR activation exacerbates the development of fatty liver disease. Additional metabolic effects of PXR activation in liver are the inhibition of fatty acid oxidation and gluconeogenesis. The identification of non-toxic and selective PXR antagonists is therefore of current research interest. Inhibition of PXR should decrease adverse effects, improve therapeutic effectiveness, and advance clinical outcomes in patients with cancer, fatty liver, and diabetes. This review identifies small molecule PXR antagonists described to date, discusses possible molecular mechanisms of inhibition, and seeks to describe the likely biomedical consequences of the inhibition of this nuclear receptor superfamily member.


Intestinal Mucosa/metabolism , Liver/metabolism , Pregnane X Receptor/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Drug Synergism , Food-Drug Interactions , Gene Expression Regulation/drug effects , Herb-Drug Interactions , Humans , Intestinal Mucosa/drug effects , Liver/drug effects , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/metabolism , Small Molecule Libraries/therapeutic use
6.
Adv Exp Med Biol ; 963: 99-110, 2017.
Article En | MEDLINE | ID: mdl-28197908

The SUMO conjugation system regulates key cellular processes including cell growth, division, mitochondrial dynamics, and the maintenance of genome stability in eukaryotic cells. The ubiquitin conjugation system regulates the stability of a myriad of vital cellular proteins in a signal-dependent manner by targeting them for destruction via the proteasome-mediated degradation pathway. Recent research efforts have unveiled an evolutionarily conserved and fundamental molecular interface between the SUMO and ubiquitin systems. A coordinated and integrated interaction between these two pathways plays a key role in adapting the SUMO-related stress response to alterations in sub-cellular protein localization, specific protein recruitment strategies, and the regulation of stress-inducible protein stability. This chapter will describe the interconnected and interdependent role of the SUMO and ubiquitin systems in mediating DNA damage repair and the genesis and the resolution of inflammatory-related diseases such as cancer. New insights regarding the interdependence of these two important post-translational modifications with nuclear receptor superfamily members will also be highlighted.


Signal Transduction , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Animals , DNA Damage , DNA Repair , Genomic Instability , Humans , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Proteolysis , Stress, Physiological
7.
Biochim Biophys Acta ; 1859(9): 1170-1182, 2016 09.
Article En | MEDLINE | ID: mdl-26883953

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.


Hepatocytes/metabolism , Histone Deacetylases/metabolism , Lysine/metabolism , Nuclear Receptor Co-Repressor 2/metabolism , Protein Processing, Post-Translational , Receptors, Steroid/chemistry , Acetylation , Amino Acid Sequence , Animals , Cell Line , Genes, Reporter , Hepatocytes/cytology , Hepatocytes/drug effects , Histone Deacetylases/genetics , Hydroxamic Acids/pharmacology , Luciferases/genetics , Luciferases/metabolism , Lysine/chemistry , Male , Mice , Mice, Inbred C57BL , Nuclear Receptor Co-Repressor 2/genetics , Pregnane X Receptor , Primary Cell Culture , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Sumoylation , Transcriptional Activation/drug effects , Ubiquitination
8.
Drug Metab Dispos ; 43(9): 1316-25, 2015 Sep.
Article En | MEDLINE | ID: mdl-26063058

Several nuclear receptor (NR) superfamily members are known to be the molecular target of either the small ubiquitin-related modifier (SUMO) or ubiquitin-signaling pathways. However, little is currently known regarding how these two post-translational modifications interact to control NR biology. We show that SUMO and ubiquitin circuitry coordinately modifies the pregnane X receptor (PXR, NR1I2) to play a key role in regulating PXR protein stability, transactivation capacity, and transcriptional repression. The SUMOylation and ubiquitylation of PXR is increased in a ligand- and tumor necrosis factor alpha -: dependent manner in hepatocytes. The SUMO-E3 ligase enzymes protein inhibitor of activated signal transducer and activator of transcription-1 (STAT1) STAT-1 (PIAS1) and protein inhibitor of activated STAT Y (PIASy) drive high levels of PXR SUMOylation. Expression of protein inhibitor of activated stat 1 selectively increases SUMO(3)ylation as well as PXR-mediated induction of cytochrome P450, family 3, subfamily A and the xenobiotic response. The PIASy-mediated SUMO(1)ylation imparts a transcriptionally repressive function by ameliorating interaction of PXR with coactivator protein peroxisome proliferator-activated receptor gamma coactivator-1-alpha. The SUMO modification of PXR is effectively antagonized by the SUMO protease sentrin protease (SENP) 2, whereas SENP3 and SENP6 proteases are highly active in the removal of SUMO2/3 chains. The PIASy-mediated SUMO(1)ylation of PXR inhibits ubiquitin-mediated degradation of this important liver-enriched NR by the 26S proteasome. Our data reveal a working model that delineates the interactive role that these two post-translational modifications play in reconciling PXR-mediated gene activation of the xenobiotic response versus transcriptional repression of the proinflammatory response in hepatocytes. Taken together, our data reveal that the SUMOylation and ubiquitylation of the PXR interface in a fundamental manner directs its biologic function in the liver in response to xenobiotic or inflammatory stress.


Hepatocytes/metabolism , Receptors, Steroid/metabolism , Animals , Humans , Mice , Mice, Knockout , Pregnane X Receptor , Signal Transduction , Sumoylation , Ubiquitination
9.
Drug Metab Dispos ; 43(3): 335-43, 2015 Mar.
Article En | MEDLINE | ID: mdl-25527709

Bacterial sepsis is characterized by a rapid increase in the expression of inflammatory mediators to initiate the acute phase response in liver. Inflammatory mediator release is counterbalanced by the coordinated expression of anti-inflammatory molecules such as interleukin 1 receptor antagonist (IL1-Ra) through time. This study determined whether activation of pregnane X receptor (PXR, NR1I2) alters the lipopolysaccharide (LPS)-inducible gene expression program in primary cultures of hepatocytes (PCHs). Preactivation of PXR for 24 hours in PCHs isolated from wild-type mice suppressed the subsequent LPS-inducible expression of the key inflammatory mediators interleukin 1ß (IL-1ß), interleukin 6 (IL-6), and tumor necrosis factor α (TNFα) but not in PCHs isolated from Pxr-null (PXR-knockout [KO]) mice. Basal expression of key inflammatory cytokines was elevated in PCHs from PXR-KO mice. Stimulation of PCHs from PXR-KO mice with LPS alone produced enhanced levels of IL-1ß when compared with wild-type mice. Experiments performed using PCHs from both humanized-PXR transgenic mice as well as human donors indicate that prolonged activation of PXR produces an increased secretion of IL1-Ra from cells through time. Our data reveal a working model that describes a pivotal role for PXR in both inhibiting as well as in resolving the inflammatory response in hepatocytes. Understanding the molecular details of how PXR is converted from a positive regulator of drug-metabolizing enzymes into a transcriptional suppressor of inflammation in liver will provide new pharmacologic strategies for modulating inflammatory-related diseases in the liver and intestine.


Hepatocytes/metabolism , Inflammation/metabolism , Receptors, Steroid/metabolism , Animals , Cells, Cultured , Gene Expression/drug effects , Gene Expression/genetics , Humans , Inflammation/genetics , Interleukins/genetics , Interleukins/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic/genetics , Mice, Transgenic/metabolism , Pregnane X Receptor , Receptors, Steroid/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
10.
Drug Metab Rev ; 45(1): 48-59, 2013 Feb.
Article En | MEDLINE | ID: mdl-23330541

Adverse drug events (ADEs) are a common cause of patient morbidity and mortality and are classically thought to result, in part, from variation in expression and activity of hepatic enzymes of drug metabolism. It is now known that alterations in the expression of genes that encode drug- and bile-acid-transporter proteins in both the gut and liver play a previously unrecognized role in determining patient drug response and eventual clinical outcome. Four nuclear receptor (NR) superfamily members, including pregnane X receptor (PXR, NR1I2), constitutive androstane receptor (NR1I3), farnesoid X receptor (NR1H4), and vitamin D receptor (NR1I1), play pivotal roles in drug- and bile-acid-activated programs of gene expression to coordinately regulate drug- and bile-acid transport activity in the intestine and liver. This review focuses on the NR-mediated gene activation of drug and bile-acid transporters in these tissues as well as the possible underlying molecular mechanisms.


Bile Acids and Salts/metabolism , Carrier Proteins/metabolism , Gastrointestinal Tract/metabolism , Liver/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Biological Transport , Constitutive Androstane Receptor , Humans , Inactivation, Metabolic , Receptors, Cytoplasmic and Nuclear/genetics
11.
Chem Res Toxicol ; 25(9): 1955-63, 2012 Sep 17.
Article En | MEDLINE | ID: mdl-22867114

The hepatotoxicity of thioacetamide (TA) has been known since 1948. In rats, single doses cause centrolobular necrosis accompanied by increases in plasma transaminases and bilirubin. To elicit these effects, TA requires oxidative bioactivation, leading first to its S-oxide (TASO) and then to its chemically reactive S,S-dioxide (TASO(2)), which ultimately modifies amine-lipids and proteins. To generate a suite of liver proteins adducted by TA metabolites for proteomic analysis and to reduce the need for both animals and labeled compounds, we treated isolated hepatocytes directly with TA. Surprisingly, TA was not toxic at concentrations up to 50 mM for 40 h. On the other hand, TASO was highly toxic to isolated hepatocytes as indicated by LDH release, cellular morphology, and vital staining with Hoechst 33342/propidium iodide. TASO toxicity was partially blocked by the CYP2E1 inhibitors diallyl sulfide and 4-methylpyrazole and was strongly inhibited by TA. Significantly, we found that hepatocytes produce TA from TASO relatively efficiently by back-reduction. The covalent binding of [(14)C]-TASO is inhibited by unlabeled TA, which acts as a "cold-trap" for [(14)C]-TA and prevents its reoxidation to [(14)C]-TASO. This in turn increases the net consumption of [(14)C]-TASO despite the fact that its oxidation to TASO(2) is inhibited. The potent inhibition of TASO oxidation by TA, coupled with the back-reduction of TASO and its futile redox cycling with TA, may help explain phenomena previously interpreted as "saturation toxicokinetics" in the in vivo metabolism and toxicity of TA and TASO. The improved understanding of the metabolism and covalent binding of TA and TASO facilitates the use of hepatocytes to prepare protein adducts for target protein identification.


Hepatocytes/metabolism , Thioacetamide/analogs & derivatives , Thioacetamide/metabolism , Animals , Cells, Cultured , Cytochrome P-450 CYP2E1/metabolism , Cytochrome P-450 CYP2E1 Inhibitors , Hepatocytes/drug effects , L-Lactate Dehydrogenase/metabolism , Male , Rats , Rats, Sprague-Dawley , Thioacetamide/toxicity
12.
Pharmacol Res ; 64(1): 4-10, 2011 Jul.
Article En | MEDLINE | ID: mdl-21397695

Pregnane x receptor (PXR, NR1I2) was originally characterized as a broad spectrum entero-hepatic xenobiotic 'sensor' and master-regulator of drug inducible gene expression. A compelling description of ligand-mediated gene activation has been unveiled in the last decade that firmly establishes this receptor's central role in the metabolism and transport of xenobiotics in mammals. Interestingly, pharmacotherapy with potent PXR ligands produces several profound side effects including decreased capacities for gluconeogenesis, lipid metabolism, and inflammation; likely due to PXR-mediated repression of gene expression programs underlying these pivotal physiological functions. An integrated model is emerging that reveals a sophisticated interplay between ligand binding and the ubiquitylation, phosphorylation, SUMOylation, and acetylation status of this important nuclear receptor protein. These discoveries point to a key role for the post-translational modification of PXR in the selective suppression of gene expression, and open the door to the study of completely new modes of regulation of the biological activity of PXR.


Protein Processing, Post-Translational/physiology , Receptors, Steroid/physiology , Animals , Gene Expression Regulation/physiology , Humans , Pregnane X Receptor
13.
J Pharmacol Exp Ther ; 335(2): 342-50, 2010 Nov.
Article En | MEDLINE | ID: mdl-20719936

Long-term treatment of patients with the macrolide antibiotic and prototypical activator of pregnane X receptor (PXR) rifampicin (Rif) inhibits the inflammatory response in liver. We show here that activation of the inflammatory response in hepatocytes strongly modulates SUMOylation of ligand-bound PXR. We provide evidence that the SUMOylated PXR contains SUMO3 chains, and feedback represses the immune response in hepatocytes. This information represents the first step in developing novel pharmaceutical strategies to treat inflammatory liver disease and prevent adverse drug reactions in patients experiencing acute or systemic inflammation. These studies also provide a molecular rationale for constructing a novel paradigm that uniquely defines the molecular basis of the interface between PXR-mediated gene activation, drug metabolism, and inflammation.


Antibiotics, Antitubercular/pharmacology , Hepatitis/immunology , Hepatocytes/metabolism , Receptors, Steroid/metabolism , Rifampin/pharmacology , Small Ubiquitin-Related Modifier Proteins/metabolism , Animals , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/immunology , Electrophoresis, Polyacrylamide Gel , Gene Expression/drug effects , HeLa Cells , Hepatitis/genetics , Hepatitis/metabolism , Hepatocytes/drug effects , Hepatocytes/immunology , Humans , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnane X Receptor , Receptors, Steroid/genetics , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity
14.
Expert Opin Drug Metab Toxicol ; 6(3): 261-71, 2010 Mar.
Article En | MEDLINE | ID: mdl-20163318

IMPORTANCE OF THE FIELD: Emerging evidence demonstrates that several nuclear receptor (NR) family members regulate drug-inducible expression and activity of several important carboxylesterase (CES) enzymes in mammalian liver and intestine. Numerous clinically prescribed anticancer prodrugs, carbamate and pyrethroid insecticides, environmental toxicants and procarcinogens are substrates for CES enzymes. Moreover, a key strategy used in rational drug design frequently utilizes an ester linkage methodology to selectively target a prodrug, or to improve the water solubility of a novel compound. AREAS COVERED IN THIS REVIEW: This review summarizes the current state of knowledge regarding NR-mediated regulation of CES enzymes in mammals and highlights their importance in drug metabolism, drug-drug interactions and toxicology. WHAT THE READER WILL GAIN: New knowledge regarding the transcriptional regulation of CES enzymes by NR proteins pregnane x receptor (NR1I2) and constitutive androstane receptor (NR1I3) has recently come to light through the use of knockout and transgenic mouse models. Novel insights regarding the species-specific cross-regulation of glucocorticoid receptor (NR3C1) and PPAR-alpha (NR1C1) signaling and CES gene expression are discussed. TAKE HOME MESSAGE: Elucidation of the role of NR-mediated regulation of CES enzymes in liver and intestine will have a significant impact on rational drug design and the development of novel prodrugs, especially for patients on combination therapy.


Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Gene Expression Regulation, Enzymologic , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Carboxylic Ester Hydrolases/chemistry , Constitutive Androstane Receptor , Humans , Pharmaceutical Preparations/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry
15.
J Pharmacol Exp Ther ; 331(1): 65-76, 2009 Oct.
Article En | MEDLINE | ID: mdl-19617467

The pregnane X receptor (PXR, NR1I2) regulates the expression of genes that encode drug-metabolizing enzymes and drug transporter proteins in liver and intestine. Understanding the molecular mechanisms that modulate PXR activity is therefore critical for the development of effective therapeutic strategies. Several recent studies have implicated the activation of kinase signaling pathways in the regulation of PXR biological activity, although direct evidence and molecular mechanisms are currently lacking. We therefore sought to characterize potential phosphorylation sites within the PXR protein by use of a rational, comprehensive, and systematic site-directed mutagenesis approach to generate phosphomimetic mutations (Ser/Thr --> Asp) and phospho-deficient mutations (Ser/Thr --> Ala) at 18 predicted consensus kinase recognition sequences in the human PXR protein. Here, we identify amino acid residues Ser8, Thr57, Ser208, Ser305, Ser350, and Thr408 as being critical for biological activity of the PXR protein. Mutations at positions 57 and 408 abolish ligand-inducible PXR activity. Mutations in the extreme N terminus and in the PXR ligand-binding domain at positions Ser8, Ser305, Ser350, and Thr408 decrease the ability of PXR to form heterodimers with retinoid X receptor alpha. Mutations at positions Ser208, Ser305, Ser350, and Thr408 alter PXR-protein cofactor interactions. Finally, the subcellular localization of the PXR protein is profoundly affected by mutations at position Thr408. These data suggest that PXR activity can potentially be regulated by phosphorylation at specific amino acid residues within several predicted consensus kinase recognition sequences to differentially affect PXR biological activity.


Receptors, Steroid/metabolism , Amino Acid Substitution/genetics , Binding Sites/genetics , Consensus Sequence , Dimerization , Genes, Reporter , Humans , Molecular Mimicry/genetics , Mutagenesis, Site-Directed/methods , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphoproteins/physiology , Phosphorylation/genetics , Predictive Value of Tests , Pregnane X Receptor , Protein Structure, Tertiary/genetics , Receptors, Steroid/antagonists & inhibitors , Receptors, Steroid/genetics , Receptors, Steroid/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology
16.
Drug Metab Dispos ; 37(7): 1539-47, 2009 Jul.
Article En | MEDLINE | ID: mdl-19359405

The liver- and intestine-enriched carboxylesterase 2 (CES2) enzyme catalyzes the hydrolysis of several clinically important anticancer agents administered as prodrugs. For example, irinotecan, a carbamate prodrug used in the treatment of colorectal cancer, is biotransformed in vivo by CES2 in intestine and liver, thereby producing a potent topoisomerase I inhibitor. Pregnane X receptor (PXR) and constitutive androstane receptor (CAR), two members of the nuclear receptor superfamily of ligand-activated transcription factors, mediate gene activation in response to xenobiotic stress. Together, these receptors comprise a protective response in mammals that coordinately regulate hepatic transport, metabolism, and elimination of numerous xenobiotic compounds. In the present study, microarray analysis was used to identify PXR target genes in duodenum in mice. Here, we show that a gene encoding a member of the CES2 subtype of liver- and intestine-enriched CES enzymes, called Ces6, is induced after treatment with pregnenolone 16alpha-carbonitrile in a PXR-dependent manner in duodenum and liver in mice. Treatment of mice with the CAR activator 1,4-bis[2-(3,5-dichloropyridyloxy)] benzene also induced expression of Ces6 in duodenum and liver in a CAR-dependent manner, whereas treatment with phenobarbital produced induction of Ces6 exclusively in liver. These data identify a key role for PXR and CAR in regulating the drug-inducible expression and activity of an important CES enzyme in vivo. Future studies should focus on determining whether these signaling pathways governing drug-inducible CES expression in intestine and liver are conserved in humans.


Carboxylic Ester Hydrolases/metabolism , Gene Expression Regulation, Enzymologic/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Steroid/physiology , Xenobiotics/pharmacology , Animals , Carboxylesterase , Carboxylic Ester Hydrolases/genetics , Constitutive Androstane Receptor , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Duodenum/metabolism , Gene Expression/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Liver/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Oligonucleotide Array Sequence Analysis , Pregnane X Receptor , Pregnenolone Carbonitrile/pharmacology , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Recoverin/genetics , Recoverin/metabolism
17.
J Biol Chem ; 284(11): 6639-49, 2009 Mar 13.
Article En | MEDLINE | ID: mdl-19141612

Pregnane x receptor is a ligand-activated transcription factor that regulates drug-inducible expression of several key cytochrome P450 enzymes and drug transporter proteins in liver and intestine in a species-specific manner. Activation of this receptor modulates several key biochemical pathways, including gluconeogenesis, beta-oxidation of fatty acids, fatty acid uptake, cholesterol homeostasis, and lipogenesis. It is of current interest to determine whether the interaction between pregnane x receptor and these key biochemical pathways is evolutionarily conserved. We show here that activation of the cyclic AMP-dependent protein kinase signaling pathway synergizes with pregnane x receptor-mediated gene activation in mouse hepatocytes. Conversely, cyclic AMP-dependent protein kinase signaling has a repressive effect upon pregnane x receptor-mediated gene activation in rat and human hepatocytes. We show that the human pregnane x receptor protein can serve as an effective substrate for catalytically active cyclic AMP-dependent protein kinase in vitro. Metabolic labeling of the protein in vivo indicates that human pregnane x receptor exists as a phosphoprotein and that activation of cyclic AMP-dependent protein kinase signaling modulates the phosphorylation status of pregnane x receptor. Activation of cyclic AMP-dependent protein kinase signaling also modulates the interactions of pregnane x receptor with protein cofactors. Our results define the species-specific impact of cyclic AMP-dependent protein kinase signaling on pregnane x receptor and provide a molecular explanation of cyclic AMP-dependent protein kinase-mediated repression of human pregnane x receptor activity. Taken together, our results identify a novel mode of regulation of pregnane x receptor activity and highlight prominent functional differences in the process across species.


Cyclic AMP-Dependent Protein Kinases/metabolism , Phosphoproteins/metabolism , Receptors, Steroid/metabolism , Signal Transduction/physiology , Animals , Enzyme Activation/physiology , Humans , Male , Mice , Phosphorylation/physiology , Pregnane X Receptor , Rats , Species Specificity
18.
Int J Neuropsychopharmacol ; 12(5): 651-65, 2009 Jun.
Article En | MEDLINE | ID: mdl-18976543

We have previously demonstrated that olanzapine-induced desensitization of 5-HT2A receptor-stimulated phospholipase C (PLC) activity is associated with increases in RGS7 protein levels both in vivo and in cells in culture, and the increase in RGS7 is dependent on activation of the JAK-STAT pathway in cells in culture. In the present study, we found that desensitization of 5-HT2A receptor-stimulated PLC activity induced by olanzapine is dependent on activation of the JAK-STAT pathway. Similar to olanzapine, clozapine-induced desensitization of 5-HT2A receptor signalling is accompanied by increases in RGS7 and activation of JAK2. Treatment with the selective 5-HT2A receptor antagonist MDL 100907 also increased RGS7 protein levels and JAK2 activation. Using a JAK2 inhibitor AG490, we found that clozapine and MDL 100907-induced increases in RGS7 are dependent on activation of the JAK-STAT pathway. Olanzapine, clozapine, and MDL 100907 treatment increased mRNA levels of RGS7. Using a chromatin immunoprecipitation assay we found STAT3 binding to the putative RGS7 promoter region. Taken together, olanzapine-induced activation of the JAK-STAT pathway, and STAT3 binding to the RGS7 gene could underlie the increase in RGS7 mRNA which could subsequently increase protein expression. Furthermore, the increase in RGS7 protein could play a role in the desensitization of 5-HT2A receptor signalling by terminating the activated Galphaq/11 proteins more rapidly. Overall, our data suggest that the complete desensitization of 5-HT2A receptor-stimulated PLC activity by olanzapine, clozapine and MDL 100907 requires activation of the JAK-STAT pathway, which in turn increases RGS7 expression probably by direct transcriptional activity of STAT3.


Benzodiazepines/pharmacology , Clozapine/pharmacology , Fluorobenzenes/pharmacology , Janus Kinases/physiology , MAP Kinase Signaling System/drug effects , Piperidines/pharmacology , STAT3 Transcription Factor/physiology , Serotonin 5-HT2 Receptor Antagonists , Type C Phospholipases/physiology , Animals , Cell Line , Enzyme Activation/drug effects , Enzyme Activation/physiology , MAP Kinase Signaling System/physiology , Olanzapine , Receptor, Serotonin, 5-HT2A/metabolism
19.
Drug Metab Dispos ; 36(8): 1538-45, 2008 Aug.
Article En | MEDLINE | ID: mdl-18474680

The pregnane X receptor (PXR, NR1I2) is a member of the nuclear receptor superfamily that is activated by a myriad of clinically used compounds and natural products. Activation of PXR in liver regulates the expression genes encoding proteins that are intimately involved in the hepatic uptake, metabolism, and elimination of toxic compounds from our bodies. PXR-mediated herb-drug interactions can have undesirable effects in patients receiving combination therapy. This can be especially important in cancer patients who self-administer over-the-counter herbal remedies together with conventional anticancer chemotherapeutics. Tian xian is a traditional Chinese herbal anticancer remedy that activates human PXR in cell-based reporter gene assays. Moreover, tian xian alters the strength of interaction between the human PXR protein and transcriptional cofactor proteins. A novel line of humanized PXR mice are described and used here to show that tian xian increases expression of Cyp3a11 in primary cultures of rodent hepatocytes. Tian xian also induces expression of CYP3A4 in primary cultures of human hepatocytes. Taken together, these data indicate that coadministration of tian xian is probably contraindicated in patients undergoing anticancer therapy with conventional chemotherapeutic agents. These data are of particular importance due to the fact that this herbal remedy is currently marketed as an adjunct therapy that reduces the side effects of conventional chemotherapy and is available without a prescription. Future studies should be conducted to determine the extent to which coadministration of this Chinese herbal remedy alters the pharmacokinetic and pharmacodynamic properties of conventional anticancer therapy.


Cytochrome P-450 CYP3A/genetics , Drugs, Chinese Herbal , Gene Expression Regulation, Enzymologic/drug effects , Hepatocytes/drug effects , Receptors, Steroid/agonists , Animals , Cells, Cultured , Hepatocytes/enzymology , Mice , Mice, Knockout , Pregnane X Receptor , Receptors, Steroid/genetics
...