Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Mol Med Rep ; 27(2)2023 Feb.
Article in English | MEDLINE | ID: mdl-36633126

ABSTRACT

Obesity is a serious medical condition worldwide, and a major risk factor for type 2 diabetes, metabolic syndrome, cancer and cardiovascular disease. In addition to changes in dietary habits and physical activity, consuming supplements to maintain good health and prevent obesity is important in modern society. Raspberry ketone (RK) is a natural phenolic ketone found in the European red raspberry (Rubus idaeus L.) and is hypothesized to prevent obesity when administered orally. The present study found that RK was reduced to rhododendrol (ROH) in human liver microsomes and cytosol. The present study investigated whether the metabolite ROH had anti­adipogenic effects using mouse 3T3­L1 cells. The effects of ROH or RK on lipid accumulation during differentiation of 3T3­L1 pre­adipocyte into adipocyte were determined using Oil Red O staining. CCAAT enhancer­binding protein α (C/EBPα) and peroxisome proliferator­activated receptor γ (PPARγ) mRNA and protein expression were examined using reverse transcription­quantitative PCR and western blotting analysis, respectively. The present study revealed that ROH suppressed lipid accumulation in the cells, similar to RK. In addition, ROH suppressed the mRNA expression levels of C/EBPα and PPARγ in 3T3­L1 adipocytes. Furthermore, ROH suppressed PPARγ protein expression in 3T3­L1 adipocytes. These findings suggested that ROH is an active metabolite with an anti­adipogenic effect, which may contribute to the anti­obesity effect of orally administered RK. The present study indicated that it is important to understand the biological activity of the metabolites of orally administered compounds.


Subject(s)
Adipocytes , Adipogenesis , Anti-Obesity Agents , Butanols , Animals , Humans , Mice , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/drug effects , Butanols/pharmacology , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Obesity/prevention & control , PPAR gamma/genetics , PPAR gamma/metabolism , Anti-Obesity Agents/pharmacology
2.
Drug Metab Pharmacokinet ; 35(2): 187-190, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32007355

ABSTRACT

Recently, genomic biomarkers have been widely used clinically for prediction of the efficacy and safety of pharmacotherapy and diagnosis and prognosis of pathological conditions. Therefore, genomic biomarkers are anticipated to accelerate not only precision medicine for pharmacotherapy but also development of molecularly targeted drugs. Because the design of clinical studies involving biomarkers may differ from conventional clinical study designs, a concept paper focused on clinical studies and patient selection methods based on genomic biomarkers is desired to prompt innovative drug development. Thus, this concept paper aimed to compile and present current scientific information from the related guidelines regarding application of genomic biomarkers to clinical trials and studies for drug development. We hope that this concept paper will prompt the development of guidelines for biomarker application to drug development by industry, regulatory authorities, the medical profession, and academia.


Subject(s)
Patient Selection , Research Design , Biomarkers/analysis , Clinical Studies as Topic , Drug Development , Humans , Precision Medicine
3.
J Appl Toxicol ; 38(2): 274-283, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28949055

ABSTRACT

Recent studies have shown that sparse distribution of regulatory T cells (Tregs) in the skin might be involved in the onset of severe cutaneous adverse drug reactions such as Stevens-Johnson syndrome and toxic epidermal necrolysis. Treg migration toward epithelial cells is regulated by certain chemokines, including TARC/CCL17 and MDC/CCL22. In this study, we analyzed the effect of allopurinol (APN), a drug known to cause severe adverse reactions, on the expression of factors affecting Treg migration and the mechanisms involved. APN inhibited the tumor necrosis factor (TNF)-α- and interferon (IFN)-γ-associated expression of TARC/CCL17 and MDC/CCL22 mRNA in HaCaT cells in a dose-dependent manner. Consistent with this, APN also suppressed TNF-α- and IFN-γ-induced production of TARC/CCL17 and MDC/CCL22 proteins and the migration of C-C chemokine receptor type 4-positive cells. Activity of the transcription factors NF-κB and STAT1, which are involved in TARC/CCL17 and MDC/CCL22 expression, was also investigated. APN inhibited activation of NF-κB, but not that of STAT1. Furthermore, it restricted p38 MAPK phosphorylation. These results suggest that APN inhibits TNF-α- and IFN-γ-induced TARC/CCL17 and MDC/CCL22 production through downregulation of p38 MAPK and NF-κB signaling, resulting in the sparse distribution of Tregs in the skin of patients with APN-associated Stevens-Johnson syndrome/toxic epidermal necrolysis.


Subject(s)
Allopurinol/toxicity , Cell Movement/drug effects , Chemokine CCL17/antagonists & inhibitors , Chemokine CCL22/antagonists & inhibitors , Keratinocytes/drug effects , NF-kappa B/metabolism , T-Lymphocytes, Regulatory/drug effects , Chemokine CCL17/metabolism , Chemokine CCL22/metabolism , HEK293 Cells , Humans , K562 Cells , Keratinocytes/immunology , Oxypurinol/toxicity , Stevens-Johnson Syndrome/etiology , Stevens-Johnson Syndrome/immunology , T-Lymphocytes, Regulatory/immunology
4.
Mol Cell Biol ; 37(10)2017 05 15.
Article in English | MEDLINE | ID: mdl-28265001

ABSTRACT

The nuclear receptor CAR (NR1I3) regulates hepatic drug and energy metabolism as well as cell fate. Its activation can be a critical factor in drug-induced toxicity and the development of diseases, including diabetes and tumors. CAR inactivates its constitutive activity by phosphorylation at threonine 38. Utilizing receptor for protein kinase 1 (RACK1) as the regulatory subunit, protein phosphatase 2A (PP2A) dephosphorylates threonine 38 to activate CAR. Here we demonstrate that CAR undergoes homodimer-monomer conversion to regulate this dephosphorylation. By coexpression of two differently tagged CAR proteins in Huh-7 cells, mouse primary hepatocytes, and mouse livers, coimmunoprecipitation and two-dimensional gel electrophoresis revealed that CAR can form a homodimer in a configuration in which the PP2A/RACK1 binding site is buried within its dimer interface. Epidermal growth factor (EGF) was found to stimulate CAR homodimerization, thus constraining CAR in its inactive form. The agonistic ligand CITCO binds directly to the CAR homodimer and dissociates phosphorylated CAR into its monomers, exposing the PP2A/RACK1 binding site for dephosphorylation. Phenobarbital, which is not a CAR ligand, binds the EGF receptor, reversing the EGF signal to monomerize CAR for its indirect activation. Thus, the homodimer-monomer conversion is the underlying molecular mechanism that regulates CAR activation, by placing phosphorylated threonine 38 as the common target for both direct and indirect activation of CAR.


Subject(s)
Hepatocytes/metabolism , Neuropeptides/metabolism , Protein Phosphatase 2/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Cells, Cultured , Constitutive Androstane Receptor , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Gene Expression Regulation/drug effects , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Ligands , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphorylation , Protein Multimerization , Receptors for Activated C Kinase
5.
J Biol Chem ; 286(41): 35763-35769, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21873423

ABSTRACT

The nuclear constitutive active/androstane receptor (CAR) is inactivated and sequestered in the cytoplasm when Thr-38 is phosphorylated. Here, we have demonstrated that activated ERK1/2 interacts with phosphorylated CAR to repress dephosphorylation of Thr-38. The phosphorylation-dependent interaction between CAR and ERK1/2 was examined by co-immunoprecipitation experiments of ectopically expressed FLAG-tagged CAR T38A and CAR T38D mutants with endogenous phospho-ERK1/2 in Huh-7 cells. Phospho-ERK1/2 coprecipitated only the phosphorylation-mimicking CAR T38D mutant; this coprecipitation was mediated by the interaction with the xenochemical response signal peptide near the C terminus of CAR. This interaction increased after EGF treatment and decreased after treatment with the MEK inhibitor U0126 as well as after knockdown of MEK1/2 by shRNA in Huh-7 cells. The phosphorylation levels of Thr-38 of CAR decreased in U0126-treated Huh-7 cells. Thus, activated ERK1/2 interacts with CAR and represses dephosphorylation of Thr-38, providing a cell signal-regulated mechanism for CAR activation.


Subject(s)
Cytoplasm/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Amino Acid Substitution , Animals , Butadienes/pharmacology , Cell Line , Constitutive Androstane Receptor , Cytoplasm/genetics , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/genetics , MAP Kinase Kinase 2/metabolism , Male , Mice , Mice, Inbred C3H , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Mutation, Missense , Nitriles/pharmacology , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Sorting Signals/physiology , Protein Structure, Tertiary , Protein Transport/drug effects , Protein Transport/physiology , Receptors, Cytoplasmic and Nuclear/genetics
6.
Drug Metab Dispos ; 38(1): 177-86, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19797611

ABSTRACT

Hepatocyte growth factor (HGF), an antimitogenic factor for HepG2 cells, increased mRNA and protein levels of UGT1A1 and CYP2B6, as well as the endogenous cyclin-dependent kinase (CDK) inhibitors p16, p21, and p27 in HepG2 cells but not in HuH6, Caco2, or MCF7 cells. Treatment with 1,4-diamino-2,3-dicyano-1,4-bis(methylthio)butadiene (U0126) (an extracellular signal-regulated kinase inhibitor) suppressed the HGF-induced expression of UGT1A1 and CYP2B6, as well as p16, p21, and p27 in HepG2 cells. The CDK inhibitor roscovitine also enhanced the expression of UGT1A1, CYP2B6, and CYP3A4. Transfection of anti-CDK2 siRNA led to elevated levels of UGT1A1, CYP2B6, and CYP3A4 in HepG2 and SW480 cells, whereas anti-CDK4 small interfering RNA (siRNA) did not significantly enhance the expression of these enzymes. In fact, CDK2 activity was decreased in HGF-treated HepG2 cells. In cells arrested in S phase by a thymidine block and then released into a synchronous cell cycle, there was a clear dissociation among the activation of CDK2 and the expression of UGT1A1, CYP2B6, and CYP3A4. Furthermore, the induction of CYP3A4 but not UGT1A1 or CYP2B6 mRNA expression by roscovitine was repressed in pregnane X receptor (PXR) siRNA-transfected HepG2 cells. Transfection with constitutive androstane receptor siRNA or PXR siRNA in HepG2 cells did not repress the HGF-stimulated expression of UGT1A1 mRNA. Taken together, our results show that the expression of UGT1A1 and CYP2B6 is negatively regulated through a CDK2 signaling pathway linked to cell cycle progression in HepG2 and SW480 cells, the mechanism of which may differ from that of CYP3A4 expression through PXR phosphorylated by CDK2.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Cell Cycle/physiology , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Glucuronosyltransferase/metabolism , Hepatocyte Growth Factor/pharmacology , Oxidoreductases, N-Demethylating/metabolism , Aryl Hydrocarbon Hydroxylases/genetics , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Enzyme Induction/drug effects , Enzyme Induction/physiology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression/drug effects , Gene Expression/genetics , Glucuronosyltransferase/genetics , Hep G2 Cells , Histones/metabolism , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Oxidoreductases, N-Demethylating/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Pregnane X Receptor , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , RNA, Small Interfering/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Roscovitine , Signal Transduction/drug effects , Signal Transduction/physiology
7.
J Biol Chem ; 284(50): 34785-92, 2009 Dec 11.
Article in English | MEDLINE | ID: mdl-19858220

ABSTRACT

Upon activation by therapeutics, the nuclear xenobiotic/ constitutive active/androstane receptor (CAR) regulates various liver functions ranging from drug metabolism and excretion to energy metabolism. CAR can also be a risk factor for developing liver diseases such as hepatocellular carcinoma. Here we have characterized the conserved threonine 38 of human CAR as the primary residue that regulates nuclear translocation and activation of CAR. Protein kinase C phosphorylates threonine 38 located on the alpha-helix spanning from residues 29-42 that constitutes a part of the first zinc finger and continues into the region between the zinc fingers. Molecular dynamics study has revealed that this phosphorylation may destabilize this helix, thereby inactivating CAR binding to DNA as well as sequestering it in the cytoplasm. We have found, in fact, that helix-stabilizing mutations reversed the effects of phosphorylation. Immunohistochemical study using an anti-phospho-threonine 38 peptide antibody has, in fact, demonstrated that the classic CAR activator phenobarbital dephosphorylates the corresponding threonine 48 of mouse CAR in the cytoplasm of mouse liver and translocates CAR into the nucleus. These results define CAR as a cell signal-regulated constitutive active nuclear receptor. These results also provide phosphorylation/dephosphorylation of the threonine as the primary drug target for CAR activation.


Subject(s)
Active Transport, Cell Nucleus/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Threonine/metabolism , Amino Acid Sequence , Animals , Constitutive Androstane Receptor , GABA Modulators/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Liver/cytology , Liver/metabolism , Mice , Mice, Knockout , Models, Molecular , Molecular Sequence Data , Phenobarbital/metabolism , Phosphorylation , Point Mutation , Protein Structure, Tertiary , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , Zinc Fingers
8.
FEBS Lett ; 583(5): 885-9, 2009 Mar 04.
Article in English | MEDLINE | ID: mdl-19302787

ABSTRACT

Constitutive androstane receptor (CAR) is a transcription factor regulating the expression of several genes related to drug metabolism. CAR expression was elevated in human HepG2 and SW480 cells by serum starvation. From reporter gene assays, mutagenesis, RNA interference, and chromatin immunoprecipitation assays, we identified the serum response element at -142/-139 in the CAR gene transactivated by Elk-1. Whereas treatment with U0126 (ERK inhibitor) enhanced CAR expression, SP600125 (stress-activated protein kinase inhibitor, SAPK) suppressed the phosphorylation of Elk-1 caused by serum-starvation stress and the elevation of CAR mRNA, suggesting that CAR expression may be mediated by phosphorylated Elk-1 via the SAPK signaling pathway.


Subject(s)
Receptors, Cytoplasmic and Nuclear/metabolism , Stress, Physiological , Transcription Factors/metabolism , Up-Regulation , ets-Domain Protein Elk-1/metabolism , 5' Flanking Region/genetics , Anthracenes/pharmacology , Base Sequence , Cell Line, Tumor , Constitutive Androstane Receptor , Culture Media, Serum-Free , Humans , Molecular Sequence Data , Phosphorylation/drug effects , Protein Binding , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction , Transcription Factors/genetics , Transcriptional Activation/genetics , ets-Domain Protein Elk-1/genetics
9.
Eur J Nutr ; 47(4): 192-200, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18560790

ABSTRACT

BACKGROUND: While naturally occurring inulin has anti-hyperlipidemic effects in animals and humans, health effects of synthetic inulin with different degrees of fructose polymerization remain poorly understood. AIM OF THE STUDY: Our study aimed at distinguishing health effects of synthetic inulin with different degrees of fructose polymerization (DP) from those of resistant maltodextrin and clofibrate. METHODS: We examined effects of synthetic inulin on serum and liver lipid profiles and blood biochemical parameters in rats fed a high-fat and high-sucrose (HF, cafeteria) diet when compared to resistant maltodextrin and clofibrate. RESULTS: Treatment with inulin (average DP = 6-8, 16-17 and 23) and resistant maltodextrin for 3 weeks reduced the elevation in liver levels of triacylglycerol and total cholesterol of rats fed the cafeteria diet but not the standard diet. In these groups, inulin (average DP = 16-17) significantly reduced the portal plasma glucose level. Moreover, the levels of portal plasma propionate and circulating serum adiponectin, which were decreased in cafeteria rats, recovered to nearly normal levels after administration of inulin (average DP = 16-17). In addition, the dietary inulin suppressed elevation in levels of portal plasma insulin and circulating serum leptin and induction of acetyl-CoA carboxylase and fatty acid synthase mRNAs in the liver of cafeteria rats, consistent with the reduction of liver lipids. The dietary inulin and clofibrate markedly reduced triacylglycerol levels in serum very low density lipoprotein (VLDL) and liver and epididymal adipose tissue weights of cafeteria rats; the extent of suppression by the dietary inulin was higher than that by clofibrate. No additive or synergistic effect of the dietary inulin and clofibrate was found in decrease in circulating serum VLDL and liver lipid levels. CONCLUSION: These observations indicate that the dietary inulin may prevent the development of metabolic disease such as hyperlipidemia and hyperinsulinemia caused by intake of cafeteria diet, in association with suppression of liver lipogenesis.


Subject(s)
Clofibrate/pharmacology , Dietary Fats/administration & dosage , Dietary Sucrose/administration & dosage , Inulin/pharmacology , Liver/drug effects , Metabolic Syndrome/prevention & control , Polysaccharides/pharmacology , Animals , Biomarkers/blood , Blood Chemical Analysis , Blood Glucose/metabolism , Insulin/blood , Lipids/blood , Liver/metabolism , Male , Metabolic Syndrome/blood , Random Allocation , Rats , Rats, Wistar
10.
Biochem Biophys Res Commun ; 369(4): 1027-33, 2008 May 16.
Article in English | MEDLINE | ID: mdl-18331826

ABSTRACT

Constitutive androstane receptor (CAR) is a transcription factor to regulate the expression of several genes related to drug-metabolism. Here, we demonstrate that CAR protein accumulates during G1 in human SW480 and HepG2 cells. After the G1/S phase transition, CAR protein levels decreased, and CAR was hardly detected in cells by the late M phase. CAR expression in both cell lines was suppressed by RNA interference-mediated suppression of CDK4. Depletion of CAR by RNA interference in both cells and by hepatocyte growth factor treatment in HepG2 cells resulted in decreased MDM2 expression that led to p21 upregulation and repression of HepG2 cell growth. Thus, our results demonstrate that CAR expression is an early G1 event regulated by CDK4 that contributes to MDM2 expression; these findings suggest that CAR may influence the expression of genes involved in not only the metabolism of endogenous and exogenous substances but also in the cell proliferation.


Subject(s)
Cell Proliferation , G1 Phase/genetics , Gene Expression Regulation , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Constitutive Androstane Receptor , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , G1 Phase/drug effects , Gene Expression , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , RNA Interference , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , S Phase , Serine Endopeptidases/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation
11.
Naunyn Schmiedebergs Arch Pharmacol ; 377(4-6): 597-605, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18172616

ABSTRACT

Human UDP-glucuronosyltransferase (UGT)1A1 is a critical enzyme responsible for detoxification and metabolism of endogenous and exogenous lipophilic compounds, such as potentially neurotoxic bilirubin and the anticancer drug irinotecan SN-38, via conjugation with glucuronic acid. A 290-bp distal enhancer module, phenobarbital-responsive enhancer module of UGT1A1 (gtPBREM), fully accounts for constitutive androstane receptor (CAR)-, pregnane X receptor (PXR)-, glucocorticoid receptor (GR)-, and aryl hydrocarbon receptor (AhR)-mediated activation of the UGT1A1 gene. This study indicates that hepatocyte nuclear factor 1alpha (HNF1alpha) bound to the proximal promoter motif not only enhances the basal reporter activity of UGT1A1, including the distal (-3570/-3180) and proximal (-165/-1) regions, but also influences the transcriptional regulation of UGT1A1 by CAR, PXR, GR, and AhR to markedly enhance reporter activities. Moreover, we assessed the influence of the TA repeat polymorphism and gtPBREM T-3279G mutation on transcriptional activation of UGT1A1 by CAR, PXR, GR, and AhR. Transcriptional activation of the A(TA)(7)TAA mutant by CAR, the PXR activator rifampicin, the GR activator dexamethasone, and the AhR activator benzo[a]pyrene was more reduced than that of the T-3279G variant, and the activity of the UGT1A1 promoter with both T-3279G and A(TA)(7)TAA mutations was still lower. Thus, UGT1A1 gene promoter variations, including the TA repeat polymorphism and T-3279G gtPBREM, have important clinical implications.


Subject(s)
Gene Expression Regulation, Enzymologic , Glucuronosyltransferase/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Transcription, Genetic , Adult , Aged , Constitutive Androstane Receptor , Female , Humans , Hyperbilirubinemia/genetics , Male , Middle Aged , Polymorphism, Genetic , Pregnane X Receptor , Promoter Regions, Genetic , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Steroid/metabolism , Transcription Factors/metabolism
12.
Drug Metab Dispos ; 36(2): 294-302, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17967931

ABSTRACT

Rats that consumed a high-fat and high-sucrose (HF1) diet or a high-fat (HF2) diet developed hepatic steatosis. The alteration in nutritional status affected hepatic cytochrome P450 and UDP-glucuronosyltransferase (UGT) levels. Messenger RNA and protein levels of UGT1A1 and UGT1A6 in the liver but not the jejunum were increased in male rats fed the HF1 diet. These protein levels did not increase in HF2-fed male rats or HF1-fed female rats. In contrast, the CYP1A2 protein level was decreased in the HF1 but not HF2 diet group, whereas CYP2E1 and CYP4A protein levels were elevated in the HF2 but not HF1 diet group. No significant difference in the organic anion transporter polypeptide (Oatp) 1, Oatp2, multidrug resistance-associated protein (Mrp) 2, or Mrp3 protein levels was found between the standard and HF1 diet groups of male rats. Consumption of the HF1 diet affected the in vivo metabolism of acetaminophen (APAP) such that the area under the APAP-glucuronide plasma concentration-time curve was elevated 2.1-fold in male rats but not female rats. In liver cell nuclei of male rats but not female rats, constitutive androstane receptor (CAR) and proliferator-activated receptor alpha (PPARalpha) protein levels were significantly enhanced by intake of the HF1 diet. Additionally, administration of the PPARalpha agonist clofibrate to male rats up-regulated UGT1A1 and UGT1A6 and down-regulated CYP1A2 in the liver. Taken together, these results indicate that nutritional status may gender-specifically influence the expression and activation of CAR and PPARalpha in liver cell nuclei, and this effect appears to be associated with alterations in UGT1A1 and UGT1A6 expression.


Subject(s)
Dietary Fats/administration & dosage , Dietary Sucrose/administration & dosage , Glucuronosyltransferase/metabolism , PPAR alpha/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Acetaminophen/blood , Acetaminophen/pharmacokinetics , Animals , Clofibrate/pharmacology , Constitutive Androstane Receptor , Cytochrome P-450 Enzyme System/metabolism , Female , Glucuronosyltransferase/genetics , Jejunum/drug effects , Jejunum/metabolism , Liver/drug effects , Liver/growth & development , Liver/metabolism , Male , Microsomes, Liver/metabolism , Organic Anion Transporters/metabolism , PPAR alpha/genetics , Phenanthrolines/pharmacology , Phenobarbital/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics
13.
Drug Metab Dispos ; 34(10): 1677-87, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16815962

ABSTRACT

Inulin enzymatically synthesized from sucrose is a dietary component that completely escapes glucide digestion. Supplementing inulin to a high-fat and high-sucrose diet (HF) ameliorated hypertriglycemia and hepatic steatosis in 8-week-fed rats by suppressing elevated levels of serum triacylglycerols, fatty acids, and glucose, and the accumulation of hepatic triacylglycerols and fatty acids. Inulin intake prevented phenobarbital (PB)- and dexamethasone-induced liver injuries in the HF group. No significant alteration in the baseline expression of CYP2B, CYP2C11, CYP3A, and NADPH-cytochrome P450 (P450) reductase mRNAs and proteins was found. In contrast, baseline and PB-treated expressions of CYP2E1 mRNA were reduced in HF-fed rats. The induction of P450s in response to PB was affected by the nutritional status of the rats; mRNA levels of CYP2B1 and CYP3A1 after PB treatment, as assessed by quantitative real-time polymerase chain reaction analysis were reduced in the inulin-supplemented HF (HF+I) group, compared with those in the HF group. Western blot analysis detected the corresponding changes of CYP2B and CYP3A proteins. These alterations were correlated with changes in hepatic thiobarbituric acid-reactive substances. Furthermore, no significant difference in the expression of nuclear receptors constitutive androstane receptor, pregnane X receptor, and retinoid X receptor alpha and coactivator peroxisome proliferator-activated receptor-gamma coactivator 1alpha proteins was found in the hepatic nucleus between the HF and HF+I groups, but the expression of hepatocyte nuclear factor alpha (HNF4alpha) protein was significantly reduced in the HF+I group. Taken together, these results indicate that inulin intake ameliorates PB-induced liver injury, associated with a decline in lipid accumulation and PB-induced expression of CYP2B and CYP3A, which may be related by a reduction in the nuclear expression of HNF4alpha.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Fatty Liver/drug therapy , Hepatocyte Nuclear Factor 4/genetics , Inulin/therapeutic use , Xenobiotics/toxicity , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Constitutive Androstane Receptor , Cytochrome P-450 Enzyme System/metabolism , Dexamethasone/administration & dosage , Dexamethasone/toxicity , Dietary Fats/administration & dosage , Dietary Fats/toxicity , Dietary Sucrose/administration & dosage , Dietary Sucrose/toxicity , Fatty Liver/blood , Fatty Liver/chemically induced , Gene Expression/drug effects , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Immunoblotting , Inulin/administration & dosage , Inulin/pharmacology , Male , PPAR alpha/metabolism , Phenobarbital/administration & dosage , Phenobarbital/toxicity , Pregnane X Receptor , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Retinoid X Receptors/metabolism , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Xenobiotics/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...