Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Biochim Biophys Acta ; 1859(9): 1218-1227, 2016 09.
Article in English | MEDLINE | ID: mdl-26994748

ABSTRACT

The ligand-activated nuclear receptor pregnane X receptor (PXR, NR1I2) and the constitutive androstane receptor (CAR, NR1I3) are two master transcriptional regulators of many important drug metabolizing enzymes and transporter genes (DMET) in response to xenobiotics including many drugs. The peroxisome proliferator-activated receptor alpha (PPARα, NR1C1), the target of lipid lowering fibrate drugs, primarily regulates fatty acid catabolism and energy-homeostasis. Recent research has shown that there are substantial overlaps in the regulated genes of these receptors. For example, both CAR and PXR also modulate the transcription of key enzymes involved in lipid and glucose metabolism and PPARα also functions as a direct transcriptional regulator of important DMET genes including cytochrome P450s CYP3A4 and CYP2C8. Despite their important and widespread influence on liver metabolism, comparative data are scarce, particularly at a global level and in humans. The major objective of this study was to directly compare the genome-wide transcriptional changes elucidated by the activation of these three nuclear receptors in primary human hepatocytes. Cultures from six individual donors were treated with the prototypical ligands for CAR (CITCO), PXR (rifampicin) and PPARα (WY14,643) or DMSO as vehicle control. Genomewide mRNA profiles determined with Affymetrix microarrays were analyzed for differentially expressed genes and metabolic functions. The results confirmed known prototype target genes and revealed strongly overlapping sets of coregulated but also distinctly regulated and novel responsive genes and pathways. The results further specify the role of PPARα as a regulator of drug metabolism and the role of the xenosensors PXR and CAR in lipid metabolism and energy homeostasis. This article is part of a Special Issue entitled: Xenobiotic nuclear receptors: New Tricks for An Old Dog, edited by Dr. Wen Xie.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Hepatocytes/metabolism , PPAR alpha/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/genetics , Transcriptome , Adult , Constitutive Androstane Receptor , Cytochrome P-450 Enzyme System/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation , Gene Ontology , Genome-Wide Association Study , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Inactivation, Metabolic/genetics , Ligands , Male , Metabolic Networks and Pathways/genetics , Middle Aged , Molecular Sequence Annotation , Oximes/pharmacology , PPAR alpha/agonists , PPAR alpha/metabolism , Pregnane X Receptor , Primary Cell Culture , Pyrimidines/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/agonists , Receptors, Steroid/metabolism , Rifampin/pharmacology , Signal Transduction , Thiazoles/pharmacology
2.
BMC Genomics ; 16: 790, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26467653

ABSTRACT

BACKGROUND: Interpreting large-scale studies from microarrays or next-generation sequencing for further experimental testing remains one of the major challenges in quantitative biology. Combining expression with physical or genetic interaction data has already been successfully applied to enhance knowledge from all types of high-throughput studies. Yet, toolboxes for navigating and understanding even small gene or protein networks are poorly developed. RESULTS: We introduce two Cytoscape plug-ins, which support the generation and interpretation of experiment-based interaction networks. The virtual pathway explorer viPEr creates so-called focus networks by joining a list of experimentally determined genes with the interactome of a specific organism. viPEr calculates all paths between two or more user-selected nodes, or explores the neighborhood of a single selected node. Numerical values from expression studies assigned to the nodes serve to score identified paths. The pathway enrichment analysis tool PEANuT annotates networks with pathway information from various sources and calculates enriched pathways between a focus and a background network. Using time series expression data of atorvastatin treated primary hepatocytes from six patients, we demonstrate the handling and applicability of viPEr and PEANuT. Based on our investigations using viPEr and PEANuT, we suggest a role of the FoxA1/A2/A3 transcriptional network in the cellular response to atorvastatin treatment. Moreover, we find an enrichment of metabolic and cancer pathways in the Fox transcriptional network and demonstrate a patient-specific reaction to the drug. CONCLUSIONS: The Cytoscape plug-in viPEr integrates -omics data with interactome data. It supports the interpretation and navigation of large-scale datasets by creating focus networks, facilitating mechanistic predictions from -omics studies. PEANuT provides an up-front method to identify underlying biological principles by calculating enriched pathways in focus networks.


Subject(s)
Metabolic Networks and Pathways/genetics , Protein Interaction Maps/genetics , Software , User-Computer Interface , Computational Biology , Gene Regulatory Networks/genetics , Humans
3.
Arch Toxicol ; 89(11): 2089-103, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25182422

ABSTRACT

In addition to its well-characterized role in the regulation of drug metabolism and transport by xenobiotics, pregnane X receptor (PXR) critically impacts on lipid homeostasis. In mice, both ligand-dependent activation and knockout of PXR were previously shown to promote hepatic steatosis. To elucidate the respective pathways in human liver, we generated clones of human hepatoma HepG2 cells exhibiting different PXR protein levels, and analyzed effects of PXR activation and knockdown on steatosis and expression of lipogenic genes. Ligand-dependent activation as well as knockdown of PXR resulted in increased steatosis in HepG2 cells. Activation of PXR induced the sterol regulatory element-binding protein (SREBP) 1-dependent lipogenic pathway via PXR-dependent induction of SREBP1a, which was confirmed in primary human hepatocytes. Inhibiting SREBP1 activity by blocking the cleavage-dependent maturation of SREBP1 protein impaired the induction of lipogenic SREBP1 target genes and triglyceride accumulation by PXR activation. On the other hand, PXR knockdown resulted in up-regulation of aldo-keto reductase (AKR) 1B10, which enhanced the acetyl-CoA carboxylase (ACC)-catalyzed reaction step of de novo lipogenesis. In a cohort of human liver samples histologically classified for non-alcoholic fatty liver disease, AKR1B10, SREBP1a and SREBP1 lipogenic target genes proved to be up-regulated in steatohepatitis, while PXR protein was reduced. In summary, our data suggest that activation and knockdown of PXR in human hepatic cells promote de novo lipogenesis and steatosis by induction of the SREBP1 pathway and AKR1B10-mediated increase of ACC activity, respectively, thus providing mechanistic explanations for a putative dual role of PXR in the pathogenesis of steatohepatitis.


Subject(s)
Fatty Liver/pathology , Hepatocytes/pathology , Receptors, Steroid/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Acetyl-CoA Carboxylase/metabolism , Aldehyde Reductase/genetics , Aldo-Keto Reductases , Fatty Liver/genetics , Gene Knockdown Techniques , Gene Silencing , Hep G2 Cells , Humans , Ligands , Lipogenesis/genetics , Pregnane X Receptor , Receptors, Steroid/metabolism , Triglycerides/metabolism , Up-Regulation/genetics
4.
Cell Physiol Biochem ; 33(6): 2003-13, 2014.
Article in English | MEDLINE | ID: mdl-25012546

ABSTRACT

BACKGROUND: RNA interference (RNAi) has tremendous potential for investigating gene function and for developing new therapies. Primary human hepatocytes (PHH) are the "gold standard" for studying the regulation of hepatic metabolism in vitro. However, application of RNAi in PHH has some technical hurdles. The objective of this study was to develop effective and robust protocol for transduction of PHH with lentiviral vectors. METHODS: We used lentiviral vectors to transduce PHH for introduction of short hairpin RNAs (shRNAs) targeting constitutive androstane receptor (CAR), peroxisome proliferator activated receptor alpha (PPARα), and microRNA, miR-143. Infection efficiency was quantitatively analyzed by flow cytometry and microscopy. Target gene expression was assessed using quantitative real-time (qRT-PCR) method. RESULTS: Lentiviral vector transduction resulted in ≥95% of infected cells at low multiplicity of infection (MOI) of 3, which did not impair cellular viability. We demonstrated the feasibility of this technique in studies on targeting nuclear receptors, PPARα and CAR, with shRNAs as well as in lentivirus-mediated overexpression and knock-down of miRNA-143 experiments. CONCLUSIONS: We developed an efficient and robust protocol with standardized procedures for virus production, method of titer determination, and infection procedure for RNAi in primary human hepatocytes based on delivery of shRNAs, microRNAs or anti-microRNAs in different laboratory settings. This approach should be useful to study not only the regulation via nuclear receptors but also other biological, pharmacological, and toxicological aspects of drug metabolism.


Subject(s)
Hepatocytes/metabolism , Lentivirus/genetics , RNA Interference , RNA, Small Interfering/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Cell Line, Tumor , Cells, Cultured , Constitutive Androstane Receptor , Flow Cytometry , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Microscopy, Fluorescence , PPAR alpha/genetics , Primary Cell Culture , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transduction, Genetic
5.
J Pharmacol Exp Ther ; 348(3): 393-400, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24259679

ABSTRACT

The acylated phloroglucinol, hyperforin, the main active ingredient of St. John's Wort, exerts antidepressant properties via indirect inhibition of serotonin reuptake by selectively activating the canonical transient receptor potential channel 6 (TRPC6). Hyperforin treatment can lead to drug-drug interactions due to potent activation of the nuclear receptor PXR (NR1I2), a key transcriptional regulator of genes involved in drug metabolism and transport. It was previously shown that synthetic acylated phloroglucinol derivatives activate TRPC6 with similar potency as hyperforin. However, their interaction potential with PXR remained unknown. Here we investigated five synthetic TRPC6-activating phloroglucinol derivatives and four TRPC6-nonactivating compounds compared with hyperforin and rifampicin for their potential to activate PXR in silico and in vitro. Computational PXR pharmacophore modeling did not indicate potent agonist or antagonist interactions for the TRPC6-activating derivatives, whereas one of them was suggested by docking studies to show both agonist and antagonist interactions. Hyperforin and rifampicin treatment of HepG2 cells cotransfected with human PXR expression vector and a CYP3A4 promoter-reporter construct resulted in potent PXR-dependent induction, whereas all TRPC6-activating compounds failed to show any PXR activation or to antagonize rifampicin-mediated CYP3A4 promoter induction. Hyperforin and rifampicin treatment of primary human hepatocytes resulted in highly correlated induction of PXR target genes, whereas treatment with the phloroglucinol derivatives elicited moderate gene expression changes that were only weakly correlated with those of rifampicin and hyperforin treatment. These results show that TRPC6-activating phloroglucinols do not activate PXR and should therefore be promising new candidates for further drug development.


Subject(s)
Phloroglucinol/analogs & derivatives , Phloroglucinol/pharmacology , Receptors, Steroid/agonists , Terpenes/pharmacology , Cytochrome P-450 CYP3A/genetics , Genes, Reporter , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Luciferases/genetics , Luciferases/metabolism , Molecular Docking Simulation , Phloroglucinol/chemistry , Pregnane X Receptor , Promoter Regions, Genetic , Receptors, Steroid/antagonists & inhibitors , Receptors, Steroid/chemistry , Terpenes/chemistry , Transcription, Genetic , Transcriptome
6.
Mol Pharmacol ; 83(3): 709-18, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23295386

ABSTRACT

The nuclear receptor peroxisome proliferator-activated receptor (PPAR)α is known primarily as a regulator of fatty acid metabolism, energy balance, and inflammation, but evidence suggests a wider role in regulating the biotransformation of drugs and other lipophilic chemicals. We investigated whether PPARα directly regulates the transcription of cytochrome P450 3A4, the major human drug-metabolizing enzyme. Using chromatin immunoprecipitation in human primary hepatocytes as well as electrophoretic mobility shift and luciferase reporter-gene assays, we identified three functional PPARα-binding regions (PBR-I, -II, and -III) within ∼12 kb of the CYP3A4 upstream sequence. Furthermore, a humanized CYP3A4/3A7 mouse model showed in vivo induction of CYP3A4 mRNA and protein by [4-chloro-6-(2,3-xylidino)-2-pyrimidinylthio]acetic acid (WY14,643) in liver but not in intestine, whereas hepatic occupancy of PBRs by PPARα was ligand independent. Using lentiviral gene knock-down and treatment with WY14,643 in primary human hepatocytes, PPARα was further shown to affect the expression of a distinct set of CYPs, including 1A1, 1A2, 2B6, 2C8, 3A4, and 7A1, but not 2C9, 2C19, 2D6, or 2E1. Interestingly, the common phospholipid 1-palmitoyl-2-oleoyl-sn-glycerol-3-phosphocholine (16:0/18:1-PC), previously proposed to reflect nutritional status and shown to be a specific endogenous ligand of PPARα, induced CYP3A4 (up to 4-fold) and other biotransformation genes in hepatocytes with similar selectivity and potency as WY14,643. These data establish PPARα as a direct transcriptional regulator of hepatic CYP3A4. This finding warrants investigation of both known and newly developed PPARα-targeted drugs for their drug-drug interaction potential. Furthermore, our data suggest that nutritional status can influence drug biotransformation capacity via endogenous phospholipid signaling.


Subject(s)
Cytochrome P-450 CYP3A/genetics , Liver/metabolism , PPAR alpha/genetics , Transcription, Genetic/genetics , Animals , Chromatin Immunoprecipitation/methods , Cytochrome P-450 CYP3A/metabolism , Hepatocytes/drug effects , Hepatocytes/enzymology , Hepatocytes/metabolism , Humans , Ligands , Liver/enzymology , Mice , PPAR alpha/metabolism , Phospholipids/metabolism , Promoter Regions, Genetic/drug effects , Protein Binding/drug effects , Pyrimidines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic/drug effects
7.
FEBS J ; 278(3): 506-18, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21166998

ABSTRACT

Low temperature is an important environmental factor affecting the performance and distribution of plants. During the so-called process of cold acclimation, many plants are able to develop low-temperature tolerance, associated with the reprogramming of a large part of their metabolism. In this study, we present a systems biology approach based on mathematical modelling to determine interactions between the reprogramming of central carbohydrate metabolism and the development of freezing tolerance in two accessions of Arabidopsis thaliana. Different regulation strategies were observed for (a) photosynthesis, (b) soluble carbohydrate metabolism and (c) enzyme activities of central metabolite interconversions. Metabolism of the storage compound starch was found to be independent of accession-specific reprogramming of soluble sugar metabolism in the cold. Mathematical modelling and simulation of cold-induced metabolic reprogramming indicated major differences in the rates of interconversion between the pools of hexoses and sucrose, as well as the rate of assimilate export to sink organs. A comprehensive overview of interconversion rates is presented, from which accession-specific regulation strategies during exposure to low temperature can be derived. We propose this concept as a tool for predicting metabolic engineering strategies to optimize plant freezing tolerance. We confirm that a significant improvement in freezing tolerance in plants involves multiple regulatory instances in sucrose metabolism, and provide evidence for a pivotal role of sucrose-hexose interconversion in increasing the cold acclimation output.


Subject(s)
Arabidopsis/metabolism , Cold Temperature , Systems Biology/methods , Acclimatization , Arabidopsis/enzymology , Arabidopsis/growth & development , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Carbohydrate Metabolism , Photosynthesis/physiology , Starch/metabolism , Sucrose/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...