Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Pharmacol ; 87(4): 674-82, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25628225

ABSTRACT

The solute carrier family 13 member 5 (SLC13A5) is a sodium-coupled transporter that mediates cellular uptake of citrate, which plays important roles in the synthesis of fatty acids and cholesterol. Recently, the pregnane X receptor (PXR, NR1I2), initially characterized as a xenobiotic sensor, has been functionally linked to the regulation of various physiologic processes that are associated with lipid metabolism and energy homeostasis. Here, we show that the SLC13A5 gene is a novel transcriptional target of PXR, and altered expression of SLC13A5 affects lipid accumulation in human liver cells. The prototypical PXR activator rifampicin markedly induced the mRNA and protein expression of SLC13A5 in human primary hepatocytes. Utilizing cell-based luciferase reporter assays, electrophoretic mobility shift assays, and chromatin immunoprecipitation assays, we identified and functionally characterized two enhancer modules located upstream of the SLC13A5 gene transcription start site that are associated with regulation of PXR-mediated SLC13A5 induction. Functional analysis further revealed that rifampicin can enhance lipid accumulation in human primary hepatocytes, and knockdown of SLC13A5 expression alone leads to a significant decrease of the lipid content in HepG2 cells. Overall, our results uncover SLC13A5 as a novel target gene of PXR and may contribute to drug-induced steatosis and metabolic disorders in humans.


Subject(s)
Fatty Liver/metabolism , Liver/metabolism , Receptors, Steroid/metabolism , Symporters/metabolism , Animals , Enhancer Elements, Genetic , Fatty Liver/chemically induced , Gene Knockdown Techniques , Hep G2 Cells , Hepatocytes/metabolism , Humans , Lipid Metabolism , Mice, Transgenic , Pregnane X Receptor , Receptors, Steroid/antagonists & inhibitors , Receptors, Steroid/genetics , Response Elements , Rifampin/toxicity , Symporters/genetics , Transcription, Genetic , Transcriptional Activation
2.
Toxicol Appl Pharmacol ; 279(1): 33-42, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24878338

ABSTRACT

OBJECTIVE: Accumulating evidence suggests that activation of mouse constitutive androstane receptor (mCAR) alleviates type 2 diabetes and obesity by inhibiting hepatic gluconeogenesis, lipogenesis, and fatty acid synthesis. However, the role of human (h) CAR in energy metabolism is largely unknown. The present study aims to investigate the effects of selective hCAR activators on hepatic energy metabolism in human primary hepatocytes (HPH). METHODS: Ligand-based structure-activity models were used for virtual screening of the Specs database (www.specs.net) followed by biological validation in cell-based luciferase assays. The effects of two novel hCAR activators (UM104 and UM145) on hepatic energy metabolism were evaluated in HPH. RESULTS: Real-time PCR and Western blotting analyses reveal that activation of hCAR by UM104 and UM145 significantly repressed the expression of glucose-6-phosphatase and phosphoenolpyruvate carboxykinase, two pivotal gluconeogenic enzymes, while exerting negligible effects on the expression of genes associated with lipogenesis and fatty acid synthesis. Functional experiments show that UM104 and UM145 markedly inhibit hepatic synthesis of glucose but not triglycerides in HPH. In contrast, activation of mCAR by 1,4-bis[2-(3,5-dichloropyridyloxy)]benzene, a selective mCAR activator, repressed the expression of genes associated with gluconeogenesis, lipogenesis, and fatty acid synthesis in mouse primary hepatocytes, which were consistent with previous observations in mouse model in vivo. CONCLUSION: Our findings uncover an important species difference between hCAR and mCAR in hepatic energy metabolism, where hCAR selectively inhibits gluconeogenesis without suppressing fatty acid synthesis. IMPLICATIONS: Such species selectivity should be considered when exploring CAR as a potential therapeutic target for metabolic disorders.


Subject(s)
Fatty Acids/biosynthesis , Gluconeogenesis/drug effects , Lipogenesis/drug effects , Receptors, Cytoplasmic and Nuclear/agonists , Animals , Blotting, Western , Cell Line , Computer Simulation , Constitutive Androstane Receptor , Energy Metabolism/drug effects , Glucose/metabolism , Glucose-6-Phosphatase/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Primary Cell Culture , Protein Transport/drug effects , Real-Time Polymerase Chain Reaction , Triglycerides/metabolism
3.
Drug Metab Dispos ; 42(6): 1090-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24713130

ABSTRACT

Studies were conducted to evaluate the impact of time and cryopreservation on aldehyde oxidase (AO) activity in human hepatocytes isolated from 10 donor livers, using O(6)-benzylguanine as a probe substrate. In addition, variability in activity was assessed using cryopreserved hepatocytes from 75 donors. Substantial donor-dependent loss in AO activity within 24 hours after isolation of hepatocytes was observed (average loss of 42%, range 15%-81%). Meanwhile, AO activity in cryopreserved hepatocytes more closely represented the activity observed in fresh hepatocytes that were incubated immediately after isolation for the same donors (within 81% of fresh, range 48%-100%). Activity of AO in cryopreserved hepatocytes from 75 donors varied by at least 17-fold (≤ 5.4 to 90 ml/minute per kilogram of body weight), with 63% of the donors having higher activity than a pooled 19-donor lot (34.2 ml/minute per kilogram). Comparison of demographics such as gender, body mass index, age, and ethnicity showed no statistically significant correlations with activity. Evaluation of medical histories revealed that three of the five donors with no measurable activity had immediate histories of extensive alcohol abuse. Meanwhile, two single nucleotide polymorphisms (SNPs) for AOX1 (rs3731772 and rs55754655) were detected in our donor pool and showed allelic frequencies similar to those reported from other cohort studies. However, these SNPs did not correlate with a statistically significant difference in intrinsic clearance compared with wild-type donors. With a general lack of clarity about what causes highly variable AO activity, prescreening donors for AO activity and creating a custom high-activity pooled lot of cryopreserved hepatocytes are advised to minimize underpredictions of clearance.


Subject(s)
Aldehyde Oxidase/genetics , Aldehyde Oxidase/metabolism , Cryopreservation , Hepatocytes/enzymology , Polymorphism, Single Nucleotide/genetics , Tissue Donors , Adult , Aged , Child , Child, Preschool , Cryopreservation/methods , Enzyme Activation/physiology , Female , Humans , Male , Metabolic Clearance Rate/physiology , Middle Aged
4.
Mol Pharmacol ; 85(2): 249-60, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24252946

ABSTRACT

Metformin is currently the most widely used drug for the treatment of type 2 diabetes. Mechanistically, metformin interacts with many protein kinases and transcription factors that alter the expression of numerous downstream target genes governing lipid metabolism, cell proliferation, and drug metabolism. The constitutive androstane receptor (CAR, NR1i3), a known xenobiotic sensor, has recently been recognized as a novel signaling molecule, in that its activation could be regulated by protein kinases in addition to the traditional ligand binding. We show that metformin could suppress drug-induced expression of CYP2B6 (a typical target gene of CAR) by modulating the phosphorylation status of CAR. In human hepatocytes, metformin robustly suppressed the expression of CYP2B6 induced by both indirect (phenobarbital) and direct CITCO [6-(4-chlorophenyl)imidazo[2,1-b]1,3thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime] activators of human CAR. Mechanistic investigation revealed that metformin specifically enhanced the phosphorylation of threonine-38 of CAR, which blocks CAR nuclear translocation and activation. Moreover, we showed that phosphorylation of CAR by metformin was primarily an AMP-activated protein kinase- and extracellular signal-regulated kinase 1/2-dependent event. Additional two-hybrid and coimmunoprecipitation assays demonstrated that metformin could also disrupt CITCO-mediated interaction between CAR and the steroid receptor coactivator 1 or the glucocorticoid receptor-interacting protein 1. Our results suggest that metformin is a potent repressor of drug-induced CYP2B6 expression through specific inhibition of human CAR activation. Thus, metformin may affect the metabolism and clearance of drugs that are CYP2B6 substrates.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Receptors, Cytoplasmic and Nuclear/drug effects , Signal Transduction/drug effects , AMP-Activated Protein Kinases/physiology , Active Transport, Cell Nucleus/drug effects , Constitutive Androstane Receptor , Cytochrome P-450 CYP2B6 , Dose-Response Relationship, Drug , ErbB Receptors/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , Oximes/pharmacology , Phenobarbital/pharmacology , Phosphorylation , Receptors, Cytoplasmic and Nuclear/metabolism , Thiazoles/pharmacology , p38 Mitogen-Activated Protein Kinases/physiology
5.
Assay Drug Dev Technol ; 10(1): 78-87, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22053711

ABSTRACT

Hepatotoxicity is a major concern for both drug development and toxicological evaluation of environmental chemicals. The assessment of compound-induced hepatotoxicity has traditionally relied on in vivo testing; however, it is being replaced by human in vitro models due to an emphasis on the reduction of animal testing and species-specific differences. Since most cell lines and hybridomas lack the full complement of enzymes at physiological levels found in the liver, primary hepatocytes are the gold standard to study liver toxicities in vitro due to the retention of most of their in vivo activities. Here, we optimized a cell viability assay using plateable cryopreserved human hepatocytes in a 1536-well-plate format. The assay was validated by deriving inhibitory concentration at 50% values for 12 known compounds, including tamoxifen, staurosporine, and phenylmercuric acetate, with regard to hepatotoxicity and general cytotoxicity using multiple hepatocyte donors. The assay performed well, and the cytotoxicity of these compounds was confirmed in comparison to HepG2 cells. This is the first study to report the reliability of using plateable cryopreserved human hepatocytes for cytotoxicity studies in a 1536-well-plate format. These results suggest that plateable cryopreserved human hepatocytes can be scaled up for screening a large compound library and may be amenable to other hepatocytic assays such as metabolic or drug safety studies.


Subject(s)
Cell Culture Techniques/methods , Cryopreservation , Cytotoxins/toxicity , Hepatocytes/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cryopreservation/methods , Dose-Response Relationship, Drug , Hepatocytes/physiology , Humans
6.
J Biomol Screen ; 16(8): 895-902, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21693767

ABSTRACT

Cytochrome P450 (CYP) enzymes are key players in drug metabolism. Therefore, it is essential to understand how these enzymes can be affected by xenobiotics with regards to induction and toxicity to avoid potential drug-drug interactions. Typically, information has been gathered by combining data from multiple experiments, which is time-consuming and labor intensive, and interassay variability may lead to misinterpretation. Monitoring CYP induction and cytotoxicity by xenobiotics using an automated, multiplexed format can decrease workload and increase data confidence. Here the authors demonstrate the ability to monitor CYP1A and CYP3A4 induction, combined with a cytotoxicity measurement, from a single microplate well using cryopreserved human hepatocytes. The assay procedure was automated in a 384-well format, including cell manipulations, compound titration and transfer, and reagent dispensing, using simple robotic instrumentation. EC(50) and E(max) values were derived for multiple known CYP1A and -3A4 inducers. Induction and toxicological responses in the triplex system were validated based on literature values from conventional single-parameter assays. Validation and pharmacology data confirm that multiplexed cell-based CYP assays can simplify workload, save time and effort, and generate biologically relevant data.


Subject(s)
Biological Products/analysis , Drug Evaluation, Preclinical/methods , Hepatocytes , High-Throughput Screening Assays , Automation, Laboratory , Biological Products/pharmacology , Cell Survival/drug effects , Cells, Cultured , Cryopreservation , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , Enzyme Induction , Genes, Reporter , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Kinetics , Luciferases/analysis , Reproducibility of Results
7.
Pharmacogenet Genomics ; 21(4): 231-6, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20739907

ABSTRACT

The arylamine N-acetyltransferase 2 (NAT2) slow acetylation phenotype is an established risk factor for urinary bladder cancer. We reported earlier on this risk association using NAT2 phenotypic categories inferred from NAT2 haplotypes based on seven single nucleotide polymorphisms (SNPs) in a study in Spain. In a subsequent genome-wide scan, we have identified a single common tag SNP (rs1495741) located in the 3' end of NAT2 that is also associated with bladder cancer risk. The aim of this report is to evaluate the agreement between the common tag SNP and the 7-SNP NAT2 inferred phenotype. The agreement between the 7-SNP NAT2 inferred phenotype and the tag SNP, rs1495741, was initially assessed in 2174 individuals from the Spanish Bladder Cancer Study (SBCS), and confirmed in a subset of individuals from the Main and Vermont component the New England Bladder Cancer Study (NEBCS). We also investigated the association of rs1495741 genotypes with NAT2 catalytic activity in cryopreserved hepatocytes from 154 individuals of European background. We observed very strong agreement between rs1495741 and the 7-SNP inferred NAT2 phenotype: sensitivity and specificity for the NAT2 slow phenotype was 99 and 95%, respectively. Our findings were replicated in an independent population from the NEBCS. Estimates for the association between NAT2 slow phenotype and bladder cancer risk in the SBCS and its interaction with cigarette smoking were comparable for the 7-SNP inferred NAT2 phenotype and rs1495741. In addition, rs1495741 genotypes were strongly related to NAT2 activity measured in hepatocytes (P<0.0001). A novel NAT2 tag SNP (rs1495741) predicts with high accuracy the 7-SNP inferred NAT2 phenotype, and thus can be used as a sole marker in pharmacogenetic or epidemiological studies of populations of European background. These findings illustrate the utility of tag SNPs, often used in genome-wide association studies (GWAS), to identify novel phenotypic markers. Further studies are required to determine the functional implications of rs1495741 and the structure and evolution of the haplotype on which it resides.


Subject(s)
Arylamine N-Acetyltransferase/genetics , Phenotype , Polymorphism, Single Nucleotide/genetics , Case-Control Studies , Genotype , Haplotypes , Humans , New England , Spain , Vermont , White People/genetics
8.
Drug Metab Dispos ; 39(1): 151-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20966043

ABSTRACT

The pregnane X receptor (PXR) binds xenobiotics and regulates the expression of several drug-metabolizing enzymes and transporters. Human PXR (hPXR) activation and CYP3A4 induction can be involved in drug-drug interactions, resulting in reduced efficacy or increased toxicity. However, there are known species-specific differences with regard to PXR activation that should be taken into account when animal PXR data are extrapolated to humans. We profiled 2816 clinically used drugs from the National Institutes of Health Chemical Genomics Center Pharmaceutical Collection for their ability to activate hPXR and rat PXR (rPXR) at the cellular level, induce human CYP3A4 at the cellular level, and bind human PXR at the protein level. From 6 to 11% of drugs were identified as active across the four assays, which included assay-specific and pan-active compounds. The lowest concordance was observed between the hPXR and rPXR assays, and many compounds active in both assays nonetheless demonstrated significant potency differences between species. Analysis based on clustering potency values demonstrated the greatest activity correlation between the hPXR activation and CYP3A4 induction assays. Structure-activity relationship analysis identified chemical scaffolds that were pan-active (e.g., dihydropyridine calcium channel blockers) and others that were uniquely active in individual assays (e.g., steroids and fatty acids). These results provide important information on PXR activation by clinically used drugs, highlight the species specificity of PXR activation by xenobiotics, and provide a means of prioritizing compounds for follow-up studies and optimization efforts.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Pharmaceutical Preparations/metabolism , Receptors, Steroid/agonists , Receptors, Steroid/metabolism , Xenobiotics/metabolism , Animals , Cell Line, Tumor , Drug Interactions , Hep G2 Cells , Hepatocytes/metabolism , Humans , Pregnane X Receptor , Rats , Small Molecule Libraries , Species Specificity , Structure-Activity Relationship
9.
J Pharmacol Exp Ther ; 334(2): 540-4, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20430842

ABSTRACT

Human populations exhibit genetic polymorphism in N-acetylation capacity, catalyzed by N-acetyltransferase 2 (NAT2). We investigated the relationship between NAT2 acetylator genotype and phenotype in cryopreserved human hepatocytes. NAT2 genotypes determined in 256 human samples were assigned as rapid (two rapid alleles), intermediate (one rapid and one slow allele), or slow (two slow alleles) acetylator phenotypes based on functional characterization of the NAT2 alleles reported previously in recombinant expression systems. A robust and significant relationship was observed between deduced NAT2 phenotype (rapid, intermediate, or slow) and N-acetyltransferase activity toward sulfamethazine (p < 0.0001) and 4-aminobiphenyl (p < 0.0001) and for O-acetyltransferase-catalyzed metabolic activation of N-hydroxy-4-aminobiphenyl (p < 0.0001), N-hydroxy-2-amino-3,8-dimethylimidazo[4,5-f] quinoxaline (p < 0.01), and N-hydroxy-2-amino-1-methyl-6-phenylimidazo[4,5-b] pyridine (p < 0.0001). NAT2-specific protein levels also significantly associated with the rapid, intermediate, and slow NAT2 acetylator phenotypes (p < 0.0001). As a negative control, p-aminobenzoic acid (an N-acetyltransferase 1-selective substrate) N-acetyltransferase activities from the same samples did not correlate with the three NAT2 acetylator phenotypes (p > 0.05). These results clearly document codominant expression of human NAT2 alleles resulting in rapid, intermediate, and slow acetylator phenotypes. The three phenotypes reflect levels of NAT2 protein catalyzing both N- and O-acetylation. Our results suggest a significant role of NAT2 acetylation polymorphism in arylamine-induced cancers and are consistent with differential cancer risk and/or drug efficacy/toxicity in intermediate compared with rapid or slow NAT2 acetylator phenotypes.


Subject(s)
4-Aminobenzoic Acid/metabolism , Aminobiphenyl Compounds/metabolism , Arylamine N-Acetyltransferase/metabolism , Hepatocytes/metabolism , Acetylation , Arylamine N-Acetyltransferase/genetics , Catalysis , Genotype , Humans , In Vitro Techniques , Polymorphism, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...