Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Pharmacol Exp Ther ; 384(1): 197-204, 2023 01.
Article in English | MEDLINE | ID: mdl-36273821

ABSTRACT

The cholesterol-conjugated heteroduplex oligonucleotide (Chol-HDO) is a double-stranded complex; it comprises an antisense oligonucleotide (ASO) and its complementary strand with a cholesterol ligand. Chol-HDO is a powerful tool for achieving target RNA knockdown in the brains of mice after systemic injection. Here, a quantitative model analysis was conducted to characterize the relationship between the pharmacokinetics (PK) and pharmacodynamics (PD), non-coding RNA metastasis-associated lung adenocarcinoma 1 (Malat1) RNA, of Chol-HDO, in a time-dependent manner. The established PK model could describe regional differences in the observed brain concentration-time profiles. Incorporating the PD model enabled the unique knockdown profiles in the brain to be explained in terms of the time delay after single dosing and enhancement following repeated dosing. Moreover, sensitivity analysis of PK exposure/persistency, target RNA turnover, and knockdown potency identified key factors for the efficient and sustained target RNA knockdown in the brain. The simulation of an adequate dosing regimen quantitatively supported the benefit of Chol-HDO in terms of achieving a suitable dosing interval. This was achieved via sufficient and sustained brain exposure and subsequent strong and sustained target RNA knockdown in the brain, even after systemic injection. The present study provides new insights into drug discoveries and development strategies for HDO in patients with neurogenic disorders. SIGNIFICANCE STATEMENT: The quantitative model analysis presented here characterized the PK/PD relationship of Chol-HDO, enabled its simulation under various conditions or assumptions, and identified key factors for efficient and sustained RNA knockdown, such as PK exposure and persistency. Chol-HDO appears to be an efficient drug delivery system for the systemic administration of desired drugs to brain targets.


Subject(s)
Oligonucleotides , RNA , Mice , Animals , Blood-Brain Barrier , Cholesterol , DNA
3.
Drug Metab Pharmacokinet ; 41: 100408, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34710650

ABSTRACT

Göttingen minipigs are increasingly used to evaluate the pharmacokinetic (PK) profiles of drug candidates. However, their accuracy in predicting human PK parameters is unclear. In this study, we investigated the utility of Göttingen minipigs for predicting human PK profiles. We evaluated the PK parameters of 30 compounds with diverse metabolic pathways after intravenous administration in minipigs. Human total clearance (CLtotal) was corrected using the blood to plasma ratio, and the volume of distribution at steady state (Vd(ss)) was corrected with plasma unbound fraction (fup). CLtotal and Vd(ss) were predicted using single-species allometric scaling using data from minipigs and other reported animal models (monkeys, human liver chimeric mice, and rats). The predicted values were compared with actual values reported in humans. Göttingen minipig were superior to rats because of their better predictability of Vd(ss) and CLtotal, as represented by lower absolute average fold error values. However, their predictability for Vd(ss) was inferior to monkey and human liver chimeric mice. Prediction of CLtotal from blood-based minipig data showed excellent correlation with human data, and comparable predictability with monkey and human liver chimeric mice. Thus, Göttingen minipigs can be used as an optional model for preclinical pharmaceutical research for predicting human CLtotal.


Subject(s)
Pharmaceutical Preparations , Administration, Intravenous , Animals , Humans , Liver , Mice , Models, Animal , Rats , Swine , Swine, Miniature
4.
Xenobiotica ; 51(1): 51-60, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32779988

ABSTRACT

The unbound fractions in plasma (f up) in two mouse models of humanized liver mice, PXB and humanized TK-NOG mice, were compared with human f up values using equilibrium dialysis method. A good relationship between f up values obtained from PXB mice and humans was observed; the f up of 34/39 compounds (87.2%) in PXB mice were within 3-fold of human f up. In contrast, a weak correlation was observed between human and humanized TK-NOG mouse f up values; the f up of 15/24 compounds (62.5%) in humanized TK-NOG mice were within 3-fold of human f up. As different profiles of plasma protein binding (PPB) profiles were observed between PXB and humanized TK-NOG mice, f up evaluation is necessary in each mouse model to utilize these humanized liver mice for pharmacological, drug-drug interaction (DDI), and toxicity studies. The unbound fraction in the mixed plasma of human and SCID mouse plasma (85:15) was well correlated with f up in PXB mice (38/39 compounds within a 3-fold). Thus, this artificial PXB mouse plasma could be used to evaluate PPB.


Subject(s)
Pharmaceutical Preparations/metabolism , Animals , Chimera , Disease Models, Animal , Hepatocytes/metabolism , Humans , Liver/metabolism , Mice , Mice, SCID , Protein Binding/physiology
5.
Toxicol Sci ; 170(1): 223-233, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31020312

ABSTRACT

Accumulation of toxic endogenous and/or exogenous substances can trigger tissue injury. Multidrug and toxin extrusion proteins (MATEs) are transporters at renal proximal tubules involved in the secretion of hydrophilic substances into urine. Multidrug and toxin extrusion protein inhibition can lead to nephrotoxicity via accumulation of toxic substances; however, case studies demonstrating causality are rare, except for drug-drug interaction studies. To explore the involvement of MATE inhibition in nephrotoxicity, MATE1 inhibition, cytotoxicity, and mitochondrial toxicity (MT) of 38 in-house compounds that showed toxicity were assessed in in vivo safety evaluations using rats, dogs, and monkeys and compared considering unbound exposures at minimal steady-state concentration (C24h,u) between nephrotoxicity positive and negative compounds. Logarithmic-corrected means of C24h,u normalized by MATE1 IC50 or cytotoxicity EC50 (C24h,u/IC50 and C24h,u/EC50) were higher for nephrotoxic compounds. An exposure cutoff of C24h,u/IC50 > 0.01 filtered nephrotoxicity with a 54% positive predictive value. Of 7 cases filtered with this cutoff, all the cases showed pathological changes at renal proximal tubules expressing MATE1. Furthermore, all cases with > 0.01 reliable exposure for MATE1 inhibition and cytotoxicity exhibited nephrotoxicity. Although compounds potent for MATE1 inhibition and cytotoxicity without and with MT (potentials of 10, 30, and 40 µM, respectively) were correctly classified as nephrotoxic by evaluation of in vitro potency alone, without considering exposures, these results suggest that MATE1 inhibition potency and cytotoxicity can be used to assess nephrotoxicity, especially at proximal tubules, and could be used for safety assessment in early drug discovery.


Subject(s)
Drug Discovery/methods , Drug-Related Side Effects and Adverse Reactions/metabolism , Kidney/drug effects , Mitochondria/drug effects , Organic Cation Transport Proteins/antagonists & inhibitors , Animals , Cell Survival/drug effects , Computer Simulation , Dogs , Drug Evaluation, Preclinical , Drug-Related Side Effects and Adverse Reactions/pathology , Haplorhini , Hep G2 Cells , Humans , Kidney/metabolism , Kidney/pathology , Madin Darby Canine Kidney Cells , Organic Cation Transport Proteins/genetics , Pharmaceutical Preparations/administration & dosage , Rats , Toxicokinetics , Transfection
6.
Xenobiotica ; 49(12): 1379-1387, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30744481

ABSTRACT

1. The prediction of human pharmacokinetic (PK) parameters is an important theme to select drug candidates from preclinical studies. It is essential to improve the prediction accuracy of compound half-life (t1/2) in humans. In this study, the predictability of t1/2 in humans using PXB mice®, chimeric mice with humanised liver, was assessed using 14 compounds showing long t1/2 in humans. 2. After intravenous administration of the compounds to PXB mice, the plasma concentration-time profiles were fitted using one- or two-compartment models and the human clearance (CLt) and distribution volume (Vdss) were predicted from single-species scaling. Using the obtained parameters, the t1/2 in humans was predicted. Using PXB mice, the predicted t1/2 values of 71.4% of the compounds were within two-fold of the actual values. Meanwhile, based on predictions using SCID mice, the host strain of the PXB mice, only 7.1% of tested compounds were within two-fold. 3. In conclusion, we demonstrated the novel utility of PXB mice for human PK predictions of compounds having long t1/2 in humans.


Subject(s)
Liver , Pharmacokinetics , Animals , Chimera , Half-Life , Hepatocytes , Humans , Liver/drug effects , Male , Mice, SCID , Mice, Transgenic
7.
Bioorg Med Chem Lett ; 28(18): 3067-3072, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30098865

ABSTRACT

CCR6 has been implicated in both autoimmune diseases and non-autoimmune diseases. Thus, inhibition of CCR6-dependent cell migration is an attractive strategy for their treatment. An orally available small molecule inhibitor of CCR6 could therefore be a useful biological probe for the pathophysiological studies. Initial SAR study of a hit compound provided potent N-benzenesulfonylpiperidine derivatives that suppressed CCL20-induced Gi signals. By subsequent scaffold morphing of the central ring and further optimization, we identified a novel series of 1,4-trans-1-benzenesulfonyl-4-aminocyclohexanes as potent and selective CCR6 inhibitors with good pharmacokinetic properties. Our compounds showed good correlation between Gi signal inhibitory activity and cell migration inhibitory activity in human CCR6-transfected CHO cells. In addition, representative compound 35 potently inhibited CCR6-dependent cell migration and the increase in ERK phosphorylation in human primary cells. Therefore, the compound could be used effectively as a biological probe against human CCR6.


Subject(s)
Amines/pharmacology , Cyclohexanes/pharmacology , Piperidines/pharmacology , Receptors, CCR6/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Amines/chemical synthesis , Amines/chemistry , Animals , B-Lymphocytes/drug effects , CHO Cells , Cell Movement/drug effects , Cricetulus , Cyclohexanes/chemical synthesis , Cyclohexanes/chemistry , Dose-Response Relationship, Drug , Haplorhini , Humans , Molecular Structure , Piperidines/chemical synthesis , Piperidines/chemistry , Receptors, CCR6/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
8.
SLAS Discov ; 23(2): 154-163, 2018 02.
Article in English | MEDLINE | ID: mdl-28846466

ABSTRACT

Ghrelin O-acyl transferase (GOAT; MBOAT4) catalyzes O-acylation at serine-3 of des-acyl ghrelin. Acyl ghrelin is secreted by stomach X/A-like cells and plays a role in appetite and metabolism. Therefore, GOAT has been expected to be a novel antiobesity target because it is responsible for acyl ghrelin production. Here, we report homogeneous time-resolved fluorescence (HTRF) and enzyme-linked immunosorbent assay (ELISA) methods utilizing human GOAT-expressing microsomes as a novel high-throughput assay system for the discovery of hit compounds and optimization of lead compounds. Hit compounds exemplified by compound A (2-[(2,4-dichlorobenzyl)sulfanyl]-1,3-benzoxazole-5-carboxylic acid) were identified by high-throughput screening using the HTRF assay and confirmed to have GOAT inhibitory activity using the ELISA. Based on the hit compound information, the novel lead compound (compound B, (4-chloro-6-{[2-methyl-6-(trifluoromethyl)pyridin-3-yl]methoxy}-1-benzothiophen-3-yl)acetic acid) was synthesized and exhibited potent GOAT inhibition with oral bioavailability. Both the hit compound and lead compound showed octanoyl-CoA competitive inhibitory activity. Moreover, these two compounds decreased acyl ghrelin production in the stomach of mice after their oral administration. These novel findings demonstrate that GOAT is a druggable target, and its inhibitors are promising antiobesity drugs.


Subject(s)
Acyltransferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Ghrelin/metabolism , Acyl Coenzyme A/metabolism , Acylation/drug effects , Administration, Oral , Animals , Biological Availability , Drug Discovery/methods , Enzyme Inhibitors/pharmacokinetics , High-Throughput Screening Assays/methods , Humans , Male , Mice , Mice, Inbred C57BL , Microsomes/drug effects , Microsomes/metabolism , Stomach/drug effects
9.
J Pharmacol Exp Ther ; 362(3): 441-449, 2017 09.
Article in English | MEDLINE | ID: mdl-28698254

ABSTRACT

In addition to their potent antidiabetic effects, glucagon-like peptide-1 (GLP-1) analogs lower body weight in humans. Hence, agonistic targeting of the GLP-1 receptor could be a valid approach to target obesity. However, quantitative analyses of the pharmacokinetic/pharmacodynamic (PK/PD) relationship between GLP-1 analogs and their antiobesity effect have not been reported in either animals or humans. Therefore, the present study was performed to establish a mechanism-based PK/PD model of GLP-1 receptor agonists using the GLP-1 analog exenatide for the development of promising new antiobesity drugs. Exenatide was administered to high-fat diet-induced obese C57BL/6J mice via subcutaneous bolus and continuous infusion. Food intake and body-weight reductions were observed and depended on the plasma concentrations of exenatide. The homeostatic feedback model, in which food intake is assumed to be regulated by appetite control signals, described the relationship among the plasma concentration-time profile of exenatide, food intake, and body weight. The estimated IC50 of exenatide against food intake was 2.05 pM, which is similar to the reported KD value of exenatide in rat brain and the estimated EC50 value for augmentation of insulin secretion in humans. The PK/PD model simulation indicated that subcutaneous infusion would show a stronger effect on body-weight reduction than bolus dosing would. This novel, quantitative PK/PD model could be used for antiobesity research and development of GLP-1 analogs, GLP-1 secretagogues, GLP-1 degradation inhibitors, and combinations thereof by allowing the estimation of appropriate pharmacokinetic profiles and dosing regimens.


Subject(s)
Anti-Obesity Agents , Glucagon-Like Peptide-1 Receptor/agonists , Models, Biological , Obesity/drug therapy , Peptides , Venoms , Animals , Anti-Obesity Agents/pharmacokinetics , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Body Weight/drug effects , Diet, High-Fat , Exenatide , Infusions, Subcutaneous , Injections, Subcutaneous , Male , Mice, Inbred C57BL , Obesity/metabolism , Peptides/pharmacokinetics , Peptides/pharmacology , Peptides/therapeutic use , Venoms/pharmacokinetics , Venoms/pharmacology , Venoms/therapeutic use
10.
Biochem Biophys Res Commun ; 484(2): 358-364, 2017 03 04.
Article in English | MEDLINE | ID: mdl-28131833

ABSTRACT

Accurate prediction of drug-induced renal toxicity is necessary for development of safer drugs for patients. Cellular assay systems that recapitulate physiologically relevant microenvironments have been proposed for correct estimation of drug responses in the human body. However, establishment of such assay systems for accurate prediction of renal toxicity is challenging because of the lack of readily available in vitro assay systems. In this study, we investigated the cellular response to fluid shear stress, which is a characteristic of the environment in the kidney proximal tubules, using microfluidic devices. The global gene expression profiles of human primary proximal tubule cells under the fluidic conditions revealed upregulation of MATE2-K and activation of Nrf2 signaling in response to fluid shear stress. Network and cell biological analysis additionally showed that expression of MATE2-K is regulated by Nrf2 signaling. These results strongly suggest that fluid shear stress is involved in the expression and maintenance of function of tissue-specific drug transporters in the proximal tubule, where the cells are exposed to continuous shear stress by primary urine. Furthermore, the microfluidic culture of human proximal tubules was demonstrated to be a useful system to analyze the regulatory mechanisms of gene expression in physiologically relevant cell conditions.


Subject(s)
NF-E2-Related Factor 2/metabolism , Organic Cation Transport Proteins/genetics , Stress, Mechanical , Cells, Cultured , Gene Expression Profiling , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism
11.
Xenobiotica ; 47(12): 1052-1063, 2017 Dec.
Article in English | MEDLINE | ID: mdl-27892765

ABSTRACT

1. The aim of the present study was to evaluate the usefulness of chimeric mice with humanised liver (PXB mice) for the prediction of clearance (CLt) and volume of distribution at steady state (Vdss), in comparison with monkeys, which have been reported as a reliable model for human pharmacokinetics (PK) prediction, and with rats, as a conventional PK model. 2. CLt and Vdss values in PXB mice, monkeys and rats were determined following intravenous administration of 30 compounds known to be mainly eliminated in humans via the hepatic metabolism by various drug-metabolising enzymes. Using single-species allometric scaling, human CLt and Vdss values were predicted from the three animal models. 3. Predicted CLt values from PXB mice exhibited the highest predictability: 25 for PXB mice, 21 for monkeys and 14 for rats were predicted within a three-fold range of actual values among 30 compounds. For predicted human Vdss values, the number of compounds falling within a three-fold range was 23 for PXB mice, 24 for monkeys, and 16 for rats among 29 compounds. PXB mice indicated a higher predictability for CLt and Vdss values than the other animal models. 4. These results demonstrate the utility of PXB mice in predicting human PK parameters.


Subject(s)
Pharmaceutical Preparations/metabolism , Pharmacokinetics , Animals , Chimera , Half-Life , Haplorhini , Hepatocytes/metabolism , Humans , Inactivation, Metabolic , Liver/metabolism , Mice , Rats
12.
Int J Pharm ; 452(1-2): 36-41, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23612356

ABSTRACT

Multidrug resistance protein 2 (MRP2, ABCC2) is localized to the apical membrane of hepatocytes and played an important role in the biliary excretion of a broad range of endogenous and xenobiotic compounds and drugs, such as pravastatin. However, the effects of statins on MRP2 in the liver and the precise mechanisms of their actions have been obscure. The goal of this study was to determine the regulatory molecular mechanism for statin-induced MRP2 expression in hepatocytes. In vitro and in vivo studies suggested that pitavastatin increased MRP2 expression. Pitavastatin promoted liver X receptor (LXR) α/ß translocation from the cytosol to nuclei, resulting in LXR activation. Deletion and mutational analysis suggested that the potential sterol regulatory element (SRE) played a major role in the observed modulation of MRP2 expression by pitavastatin. Furthermore pitavastatin increased the protein-DNA complex, and when SRE was mutated, stimulation of the protein-DNA complex by pitavastatin was decreased. It was demonstrated that pitavastatin upregulated MRP2 expression by an SREBP regulatory pathway in hepatocytes and that the actions of statins may lead to improve the biliary excretion of MRP2 substrates.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Multidrug Resistance-Associated Proteins/metabolism , Quinolines/pharmacology , Sterol Regulatory Element Binding Proteins/metabolism , ATP-Binding Cassette Transporters/genetics , Animals , Atorvastatin , Cells, Cultured , Gene Expression Regulation/drug effects , Hep G2 Cells , Heptanoic Acids/pharmacology , Humans , Liver/drug effects , Liver/metabolism , Liver X Receptors , Male , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Orphan Nuclear Receptors/metabolism , Pyrroles/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar
13.
Yakugaku Zasshi ; 132(5): 609-15, 2012.
Article in Japanese | MEDLINE | ID: mdl-22687697

ABSTRACT

The aim of this study was to determine the effects of α-cyano-4-hydroxycinnamic acid (CHC), a lactate efflux inhibitor, and citrate, an alkaline reagent, on statin-induced muscle injury using a human prototypic embryonal rhabdomyosarcoma cell line (RD) as a model of in vitro skeletal muscle and on statin-induced muscle damage in an in vivo study. Statin-induced reduction of cell viability and apoptosis was measured by the 3-(4,5-dimethylthiazol-2-thiazolyl)-2,5-diphenyl tetrazolium bromide (MTT) assay and caspase assay. In an in vivo study, plasma creatine phosphokinase (CPK) level was examined in cerivastatin-treated rats. CHC increased growth inhibition of RD cells induced by cerivastatin, a lipophilic statin, but not these induced by pravastatin, a hydrophilic statin. On the other hand, citrate suppressed cerivastatin-, simvastatin- and atorvastatin-induced reduction of cell viability and caspase activation in RD cells. Moreover, citrate prevented cerivastatin-induced increase in CPK concentration in a concentration-dependent manner. This is first study to evaluate CHC or citrate-induced exacerbation or improvement of statin-induced muscle damage.


Subject(s)
Citric Acid/pharmacology , Coumaric Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Pyridines/adverse effects , Animals , Apoptosis/drug effects , Caspase 1/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Creatine Kinase/blood , Humans , Hydrogen-Ion Concentration , Male , Muscle, Skeletal/pathology , Rats , Rats, Wistar , Rhabdomyosarcoma, Embryonal/enzymology , Rhabdomyosarcoma, Embryonal/pathology
14.
Eur J Pharmacol ; 662(1-3): 9-14, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21554872

ABSTRACT

ATP-binding cassette transporter A1 (ABCA1) is predicted to be involved in the control of apolipoprotein AI-mediated cholesterol efflux: biosynthesis of high-density lipoprotein (HDL). However, the effects of HMG-CoA reductase inhibitors (statins) on ABCA1 in the liver and the precise mechanisms of their actions have been obscure. The aims of this study were to determine whether statins (atorvastatin (Ato) and pitavastatin (Pit)) affect hepatic ABCA1 expression and to clarify the mechanisms of their actions using HepG2 cells and the rat liver. We examined alterations in mRNA and protein levels of ABCA1 and peroxisome proliferator-activated receptors (PPARs) by quantitative real-time polymerase chain reaction (PCR) and Western blot analysis, respectively. In vitro and in vivo studies suggested that Pit increases ABCA1 mRNA level, but not Ato. Pit greatly increased Abca1 mRNA level and also increased the amount of plasma HDL and the mRNA level of PPARα. Clofibrate (PPARα agonist) increased ABCA1 expression in HepG2 cells and rat primary hepatocytes more than did PPAR ß/δ and γ agonists. Pit-induced ABCA1 expression alteration was blocked by GW6471 (PPARα antagonist) and by PPARα knockdown. In this study, we demonstrated that Pit affect ABCA1 expression via PPARα in hepatocytes. The strategy to target a PPARα agonist in the liver can lead to increases in ABCA1 expression and HDL level.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Heptanoic Acids/pharmacology , Pyrroles/pharmacology , Quinolines/pharmacology , ATP Binding Cassette Transporter 1 , Animals , Atorvastatin , Hep G2 Cells , Humans , Male , PPAR alpha/metabolism , Rats , Rats, Wistar
15.
Biochim Biophys Acta ; 1788(11): 2396-403, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19728987

ABSTRACT

Liver X receptors (LXRs) belong to the nuclear hormone receptor superfamily. Multidrug resistance-associated protein 2 (MRP2), multidrug resistance 1 (MDR1) and breast cancer resistance protein (BCRP) play an important role in the efflux of a broad range of endogenous and xenobiotic compounds from hepatocytes. Since the effects of LXR activation on there transporters have been obscure, we investigated the effects of LXR agonists, TO901317 and 25-hydroxycholesterol, on MRP2, MDR1, BCRP expression in HepG2 cells and the rat liver. In an in vitro study, TO901317 increased ABCA1, an LXR target gene, and MRP2 mRNA and protein levels. On the other hand, TO901317 had little effect on MDR1 and BCRP mRNA levels. In an in vivo study, Abca1 and Mrp2 mRNA and protein levels were increased by TO901317, but TO901317 had no effect on Mdr1a and Bcrp mRNA levels in the rat liver. Moreover, TO901317-induced MRP2 mRNA expression was blocked by LXRalpha knockdown. In this study, we demonstrated that LXR activation induced expression of MRP2 but not that of MDR1 and BCRP in hepatocytes. The results suggest that agonists for LXR activate transcription of the MRP2 gene in order to promote excretion of endogenous and xenobiotic compounds from hepatocytes into bile.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP-Binding Cassette Transporters/genetics , Gene Expression Regulation , Liver/metabolism , Multidrug Resistance-Associated Proteins/genetics , Neoplasm Proteins/genetics , Orphan Nuclear Receptors/metabolism , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Animals , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Hydrocarbons, Fluorinated/pharmacology , Hydroxycholesterols/pharmacology , Immunoenzyme Techniques , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver X Receptors , Male , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Orphan Nuclear Receptors/antagonists & inhibitors , Orphan Nuclear Receptors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , Transfection
16.
Life Sci ; 82(17-18): 969-75, 2008 Apr 23.
Article in English | MEDLINE | ID: mdl-18402982

ABSTRACT

In the present study, we examined the mechanisms underlying the cytotoxicity of pitavastatin, a new statin, and we compared the in vitro potencies of muscle cytotoxicity using a prototypic embryonal rhabdomyosarcoma cell line (RD cells), a typical side effect of statins and compared the cholesterol-lowering effects of statins using Hep G2 hepatoma cells. Pitavastatin reduced the number of viable cells and caused caspase-9 and -3/7 activation in a time- and concentration-dependent manner. The comparison of cytotoxities of statins showed that statins significantly reduced cell viability and markedly enhanced activity of caspase-3/7 in concentration-dependent manner. On the other hand, the effects of hydrophilic statins, pravastatin, rosuvastatin were very weak. The rank order of cytotoxicity was cerivastatin > simvastatin acid> fluvastatin > atorvastatin > lovastatin acid > pitavastatin >> rosuvastatin, pravastatin. Statin-induced cytotoxicity is associated with these partition coefficients. On the other hand, the cholesterol-lowering effect of statins did not correlate with these partition coefficients and cytotoxicity. Thus, it is necessary to consider the association between risk of myopathy and cholesterol-lowering effect of a statin for precise use of statins.


Subject(s)
Anticholesteremic Agents/adverse effects , Anticholesteremic Agents/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Muscular Diseases/drug therapy , Anticholesteremic Agents/pharmacokinetics , Caspases/metabolism , Cell Line, Tumor , Cholesterol/blood , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Muscular Diseases/pathology , Quinolines/adverse effects , Quinolines/therapeutic use , Tetrazolium Salts , Thiazoles
SELECTION OF CITATIONS
SEARCH DETAIL
...