Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
J Pharm Pharm Sci ; 24: 475-483, 2021.
Article in English | MEDLINE | ID: mdl-34516949

ABSTRACT

PURPOSE: Organic anion transporting polypeptide (OATP) 1B3 transports many clinically important drugs, including statins, from blood into the liver. It exclusively expresses in human liver under normal physiological conditions. There is no rodent ortholog of human OATP1B3. Tissue targeting of therapeutic molecules mediated by transporters, including liver-targeting via liver-specific OATPs, is an emerging area in drug development. Sandwich-cultured primary hepatocytes (SCH) are a well characterized in vitro model for assessment of hepatic drug uptake and biliary excretion. The current study was designed to develop a novel rat SCH model expressing human OATP1B3 to study the hepatic disposition of OATP1B3 substrates. METHODS: Primary rat hepatocytes transduced with adenoviral vectors expressing FLAG-tagged OATP1B3 (Ad-OATP1B3), a control vector Ad-LacZ, or that were non-transduced were cultured in a sandwich configuration. FLAG immunoblot and immunofluorescence-staining determined expression and localization of OATP1B3. Uptake of [3H]-cholecystokinin octapeptide (CCK-8), a specific OATP1B3 substrate, was determined. Taurocholate (TC) is a substrate routinely used in SCH to assess biliary excretion via bile canaliculi (BC) and is also a substrate of OATP1B3. [3H]-TC accumulation in cells+BC, cells, biliary excretion index (BEI) and in vitro Clbiliary were determined using B-CLEAR® technology. RESULTS: OATP1B3 protein was extensively expressed and primarily localized on the plasma membrane in day 4 Ad-OATP1B3-transduced rat SCH. [3H]-CCK-8 accumulation in cells+BC was significantly greater (~5-13 folds, p<0.001) in day 4 SCH with vs. without Ad-OATP1B3-transduction. Expressing OATP1B3 in rat SCH significantly increased [3H]-TC accumulation in cells+BC and cells, without affecting BEI and in vitro Clbiliary. CONCLUSIONS: Rat SCH expressing human OATP1B3-is a novel in vitro model allowing simultaneous assessment of hepatic uptake, hepatocellular accumulation and biliary excretion process of a human OATP1B3 substrate. This model could be a potential tool for screening for liver-targeting compounds mediated by OATP1B3.


Subject(s)
Cell Culture Techniques , Hepatocytes/metabolism , Liver/metabolism , Models, Biological , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Adenoviridae/genetics , Animals , Drug Evaluation, Preclinical , Genetic Vectors , Male , Rats, Wistar , Sincalide/pharmacology , Solute Carrier Organic Anion Transporter Family Member 1B3/genetics
2.
J Ethnopharmacol ; 280: 114469, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34329714

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Ginkgo biloba L. is a traditional Chinese medicine for hyper lipaemia. Ginkgo flavonols and terpene lactones are responsible for the lipid-lowering effect in non-alcoholic fatty liver disease (NAFLD). However, the pharmacokinetics of ginkgo flavonols and terpene lactones in NAFLD was not clarified. AIM OF THE STUDY: To investigate the effects of Ginkgo biloba L. leaves extracts (EGB) and NAFLD on hepatocyte organic anion transporting polypeptide (Oatp)1b2, and to assess the pharmacokinetics of EGB active ingredients in NAFLD rats. MATERIALS AND METHODS: Male rats were fed with a high-fat diet to induce NAFLD models. The pharmacokinetic characteristics of EGB active ingredients were studied in NAFLD rats after two or four weeks of treatment with 3.6, 10.8, and 32.4 mg/kg EGB. The effects of NAFLD and EGB were investigated on the systemic exposure of pitavastatin, a probe substrate of Oatp1b2. The inhibitory effects of ginkgo flavonols and terpene lactones on OATP1B1-mediated uptake of 3H-ES were tested in hOATP1B1-HEK293 cells. RESULTS: The plasma exposure of ginkgolides and flavonols in NAFLD rats increased in a dose-dependent manner following oral administration of EGB at 3.6-32.4 mg/kg. The half-lives of ginkgolides A, B, C, and bilobalide (2-3 h) were shorter than quercetin, kaempferol, and isorhamnetin (approximately 20 h). NAFLD reduced the plasma pitavastatin exposure by about 50 % due to the increased Oatp1b2 expression in rat liver. Increased EGB (from 3.6 to 32.4 mg/kg) substantially increased the Cmax and AUC0-t of pitavastatin by 1.8-3.2 and 1.3-3.0 folds, respectively. In hOATP1B1-HEK293 cells, kaempferol and isorhamnetin contributed to the inhibition of OATP1B1-mediated uptake of 3H-ES with IC50 values of 3.28 ± 1.08 µM and 46.12 ± 5.25 µM, respectively. CONCLUSIONS: NAFLD and EGB can alter the activity of hepatic uptake transporter Oatp1b2 individually or in combination. The pharmacokinetic herb-disease-drug interaction found in this research will help inform the clinical administration of EGB or Oatp1b2 substrates.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Non-alcoholic Fatty Liver Disease/physiopathology , Plant Extracts/pharmacology , Quinolines/pharmacokinetics , Animals , Area Under Curve , Diet, High-Fat , Disease Models, Animal , Dose-Response Relationship, Drug , Ginkgo biloba , HEK293 Cells , Herb-Drug Interactions , Humans , Liver-Specific Organic Anion Transporter 1/metabolism , Male , Plant Extracts/administration & dosage , Rats , Rats, Sprague-Dawley , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism
3.
Drug Metab Dispos ; 48(12): 1264-1270, 2020 12.
Article in English | MEDLINE | ID: mdl-33037044

ABSTRACT

Organic anion-transporting polypeptide (OATP) 1B1/3-mediated drug-drug interaction (DDI) potential is evaluated in vivo with rosuvastatin (RST) as a probe substrate in clinical studies. We calibrated our assay with RST and estradiol 17-ß-D-glucuronide (E217ßG)/cholecystokinin-8 (CCK8) as in vitro probes for qualitative and quantitative prediction of OATP1B-mediated DDI potential for RST. In vitro OATP1B1/1B3 inhibition using E217ßG and CCK8 yielded higher area under the curve (AUC) ratio (AUCR) values numerically with the static model, but all probes performed similarly from a qualitative cutoff-based prediction, as described in regulatory guidances. However, the magnitudes of DDI were not captured satisfactorily. Considering that clearance of RST is also mediated by gut breast cancer resistance protein (BCRP), inhibition of BCRP was also incorporated in the DDI prediction if the gut inhibitor concentrations were 10 × IC50 for BCRP inhibition. This combined static model closely predicted the magnitude of RST DDI with root-mean-square error values of 0.767-0.812 and 1.24-1.31 with and without BCRP inhibition, respectively, for in vitro-in vivo correlation of DDI. Physiologically based pharmacokinetic (PBPK) modeling was also used to simulate DDI between RST and rifampicin, asunaprevir, and velpatasvir. Predicted AUCR for rifampicin and asunaprevir was within 1.5-fold of that observed, whereas that for velpatasvir showed a 2-fold underprediction. Overall, the combined static model incorporating both OATP1B and BCRP inhibition provides a quick and simple mathematical approach to quantitatively predict the magnitude of transporter-mediated DDI for RST for routine application. PBPK complements the static model and provides a framework for studying molecules when a dynamic model is needed. SIGNIFICANCE STATEMENT: Using 22 drugs, we show that a static model for organic anion-transporting polypeptide (OATP) 1B1/1B3 inhibition can qualitatively predict potential for drug-drug interaction (DDI) using a cutoff-based approach, as in regulatory guidances. However, consideration of both OATP1B1/3 and gut breast cancer resistance protein inhibition provided a better prediction of the magnitude of the transporter-mediated DDI of these inhibitors with rosuvastatin. Based on these results, we have proposed an empirical mechanistic-static approach for a more reliable prediction of transporter-mediated DDI liability with rosuvastatin that drug development teams can leverage.


Subject(s)
Liver-Specific Organic Anion Transporter 1/metabolism , Models, Biological , Rosuvastatin Calcium/pharmacokinetics , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Area Under Curve , Cholecystokinin/pharmacokinetics , Drug Evaluation, Preclinical , Drug Interactions , Estradiol/analogs & derivatives , Estradiol/pharmacokinetics , HEK293 Cells , Humans , Liver-Specific Organic Anion Transporter 1/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Peptide Fragments/pharmacokinetics , Solute Carrier Organic Anion Transporter Family Member 1B3/antagonists & inhibitors
4.
Br J Cancer ; 123(12): 1715-1719, 2020 12.
Article in English | MEDLINE | ID: mdl-32989230

ABSTRACT

Androgen receptor (AR) signalling drives neoplastic growth and therapy resistance in prostate cancer. Recent clinical data show that docetaxel combined with androgen deprivation therapy improves outcome in hormone-sensitive disease. We studied whether testosterone and AR signalling interferes with docetaxel treatment efficacy in castration-resistant prostate cancer (CRPC). We found that testosterone supplementation significantly impaired docetaxel tumour accumulation in a CRPC model, resulting in decreased tubulin stabilisation and antitumour activity. Furthermore, testosterone competed with docetaxel for uptake by the drug transporter OATP1B3. Irrespective of docetaxel-induced tubulin stabilisation, AR signalling by testosterone counteracted docetaxel efficacy. AR-pathway activation could also reverse long-term tumour regression by docetaxel treatment in vivo. These results indicate that to optimise docetaxel efficacy, androgen levels and AR signalling need to be suppressed. This study lends evidence for continued maximum suppression of AR signalling by combining targeted therapeutics with docetaxel in CRPC.


Subject(s)
Androgen Antagonists/therapeutic use , Antineoplastic Agents/therapeutic use , Docetaxel/therapeutic use , Drug Resistance, Neoplasm/physiology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/metabolism , Testosterone/pharmacology , Acetylation , Androgen Antagonists/pharmacokinetics , Androgen Receptor Antagonists/therapeutic use , Animals , Antineoplastic Agents/pharmacokinetics , Cell Death , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival , Disease Progression , Docetaxel/pharmacokinetics , Drug Interactions , Humans , In Situ Nick-End Labeling , Male , Mice , Mice, Nude , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Prostate-Specific Antigen/biosynthesis , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/drug effects , Signal Transduction/drug effects , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Testosterone/administration & dosage , Testosterone/antagonists & inhibitors , Testosterone/metabolism , Tubulin/drug effects , Tubulin/metabolism
5.
Clin Transl Sci ; 13(1): 47-52, 2020 01.
Article in English | MEDLINE | ID: mdl-31468718

ABSTRACT

As the research into the organic anion transporting polypeptides (OATPs) continues to grow, it is important to ensure that the data generated are accurate and reproducible. In the in vitro evaluation of OATP1B1/1B3 inhibition, there are many variables that can contribute to variability in the resulting inhibition constants, which can then, in turn, contribute to variable results when clinical predictions (R-values) are performed. Currently, the only experimental condition recommended by the US Food and Drug Administration (FDA) is the inclusion of a pre-incubation period.1 To identify other potential sources of variability, a descriptive analysis of available in vitro inhibition data was completed. For each of the 21 substrate/inhibitor pairs evaluated, cell type and pre-incubation were found to have the greatest effect on half-maximal inhibitory concentration (IC50 ) variability. Indeed, when only HEK293 cells and co-incubation conditions were included, the observed variability for the entire data set (highest IC50 /lowest) was reduced from 12.4 to 5.2. The choice of probe substrate used in the study also had a significant effect on inhibitor constant variability. Interestingly, despite the broad range of inhibitory constants identified, these two factors showed little effect on the calculated R-values relative to the FDA evaluation cutoff of 1.1 triggering a clinical evaluation for the inhibitors evaluated. However, because of the small data set available, further research is needed to confirm these preliminary results and define best practice for the study of OATPs.


Subject(s)
Drug Evaluation, Preclinical/methods , Liver-Specific Organic Anion Transporter 1/antagonists & inhibitors , Solute Carrier Organic Anion Transporter Family Member 1B3/antagonists & inhibitors , Cell Culture Techniques/methods , Cell Culture Techniques/standards , Cyclosporine/pharmacology , Datasets as Topic , Drug Evaluation, Preclinical/standards , Drug Interactions , Gemfibrozil/pharmacology , Guidelines as Topic , HEK293 Cells , Humans , Inhibitory Concentration 50 , Liver-Specific Organic Anion Transporter 1/metabolism , Reproducibility of Results , Rifampin/pharmacology , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , United States , United States Food and Drug Administration/standards
6.
Drug Metab Dispos ; 47(10): 1040-1049, 2019 10.
Article in English | MEDLINE | ID: mdl-31399508

ABSTRACT

Rhinacanthin-C is a major active constituent in Rhinacanthus nasutus (L.) Kurz, a plant widely used in herbal remedies. Its potential for pharmacokinetic herb-drug interaction may exist with drug transporters and drug metabolizing enzymes. This study assessed the possibility for rhinacanthin-C-mediated drug interaction by determining its inhibitory effects against major human efflux and influx drug transporters as well as various human cytochrome P450(CYP) isoforms. Rhinacanthin-C demonstrated a moderate permeability through the Caco-2 monolayers [Papp (AP-to-BL) = 1.26 × 10-6 cm/s]. It significantly inhibited transport mediated by both P-glycoprotein (P-gp) (IC50 = 5.20 µM) and breast cancer resistance protein (BCRP) (IC50 = 0.83 µM) across Caco-2 and BCRP-overexpressing Madin-Darby canine kidney II cells (MDCKII) cells. This compound also strongly inhibited uptake mediated by organic anion-transporting polypeptide 1B1 (OATP1B1) (IC50 = 0.70 µM) and OATP1B3 (IC50 = 3.95 µM) in OATP1B-overexpressing HEK cells. In addition to its inhibitory effect on these drug transporters, rhinacanthin-C significantly inhibited multiple human CYP isoforms including CYP2C8 (IC50 = 4.56 µM), 2C9 (IC50 = 1.52 µM), 2C19 (IC50 = 28.40 µM), and 3A4/5 (IC50 = 53 µM for midazolam and IC50 = 81.20 µM for testosterone), but not CYP1A2, 2A6, 2B6, 2D6, and 2E1. These results strongly support a high propensity for rhinacanthin-C as a perpetrator of clinical herb-drug interaction via inhibiting various influx and efflux drug transporters (i.e., P-gp, BCRP, OATP1B1, and OATP1B3) and CYP isoforms (i.e., CYP2C8, CYP2C9, and CYP2C19). Thus, the potential for significant pharmacokinetic herb-drug interaction should be addressed when herbal products containing rhinacanthin-C are to be used in conjunction with other prescription drugs.


Subject(s)
Acanthaceae/chemistry , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Herb-Drug Interactions , Naphthoquinones/pharmacology , Prescription Drugs/pharmacology , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Caco-2 Cells , Cell Membrane Permeability/drug effects , Cytochrome P-450 Enzyme System/metabolism , Dogs , HEK293 Cells , Humans , Inhibitory Concentration 50 , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Liver-Specific Organic Anion Transporter 1/antagonists & inhibitors , Liver-Specific Organic Anion Transporter 1/metabolism , Madin Darby Canine Kidney Cells , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Recombinant Proteins/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3/antagonists & inhibitors , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism
7.
J Agric Food Chem ; 67(32): 9079-9087, 2019 Aug 14.
Article in English | MEDLINE | ID: mdl-31353905

ABSTRACT

Organic anion transporting polypeptides (OATPs) 1B1 and 1B3 are two highly homologous transporters expressed in the human liver. However, epigallocatechin gallate (EGCG), which is the most predominant catechin in green tea, has opposite effects on the function of OATP1B1 and OATP1B3. In the present study, the critical structural domains and amino acid residues for the activation of OATP1B3 by EGCG have been determined by characterizing the function of a series of OATP1B3-derived chimeric transporters, site-directed mutagenesis, and kinetic studies. Our results showed that G45 and F555 in transmembrane domains 1 and 10 are the most important amino acid residues for OATP1B3 activation. Kinetic studies showed that the activation of OATP1B3 by EGCG at a low substrate concentration was due to its increased substrate binding affinity. However, EGCG caused increased Km and decreased Vmax for 1B3-G45A and 1B3-F555H. The flexibility at position 45 and aromaticity at position 555 might be important for OATP1B3 activation. While 1B3-G45A and 1B3-F555H could not be activated by EGCG, their transport activity for EGCG was comparable to that of wild-type OATP1B3. In conclusion, the present study elucidated the molecular mechanism for OATP1B3 activation by EGCG.


Subject(s)
Catechin/analogs & derivatives , Plant Extracts/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3/chemistry , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Amino Acid Motifs , Camellia sinensis/chemistry , Catechin/chemistry , Catechin/metabolism , HEK293 Cells , Humans , Kinetics , Liver/metabolism , Liver-Specific Organic Anion Transporter 1/chemistry , Liver-Specific Organic Anion Transporter 1/genetics , Liver-Specific Organic Anion Transporter 1/metabolism , Models, Molecular , Plant Extracts/chemistry , Protein Domains , Solute Carrier Organic Anion Transporter Family Member 1B3/genetics
8.
Chem Biol Interact ; 310: 108745, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31299240

ABSTRACT

Ursodeoxycholic acid (UDCA) is a major effective constituent of bear bile powder, which is widely used as function food in China and is documented in the Chinese pharmacopoeia as a traditional Chinese medicine. UDCA has been developed as the only accepted therapy by the US FDA for primary biliary cholangitis. Recently, the US FDA granted accelerated approval to obeticholic acid (OCA), a semisynthetic bile acid derivative from chenodeoxycholic acid, for primary biliary cholangitis. However, some perplexing toxicities of UDCA have been reported in the clinic. The present work aimed to investigate the difference between UDCA and OCA in regard to potential metabolic activation through acyl glucuronidation and hepatic accumulation of consequent acyl glucuronides. Our results demonstrated that the metabolic fates of UDCA and OCA were similar. Both UDCA and OCA were predominantly metabolically activated by conjugation to the acyl glucuronide in human liver microsomes. UGT1A3 played a predominant role in the carboxyl glucuronidation of both UDCA and OCA, while UGT2B7 played a major role in their hydroxyl glucuronidation. Further uptake studies revealed that OATP1B1- and 1B3-transfected cells could selectively uptake UDCA acyl glucuronide, but not UDCA, OCA, and OCA acyl glucuronide. In summary, the liver disposition of OCA is different from that of UDCA due to hepatic uptake, and liver accumulation of UDCA acyl glucuronide might be related to the perplexing toxicities of UDCA.


Subject(s)
Chenodeoxycholic Acid/analogs & derivatives , Glucuronides/metabolism , Liver-Specific Organic Anion Transporter 1/metabolism , Microsomes, Liver/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Ursodeoxycholic Acid/metabolism , Animals , Biological Transport , Chenodeoxycholic Acid/metabolism , Humans , Medicine, Chinese Traditional , Ursidae , Ursodeoxycholic Acid/analogs & derivatives , Ursodeoxycholic Acid/toxicity
9.
Acta Pharmacol Sin ; 40(6): 833-849, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30327544

ABSTRACT

ShenMai, an intravenous injection prepared from steamed Panax ginseng roots (Hongshen) and Ophiopogon japonicus roots (Maidong), is used as an add-on therapy for coronary artery disease and cancer; saponins are its bioactive constituents. Since many saponins inhibit human organic anion-transporting polypeptides (OATP)1B, this investigation determined the inhibition potencies of circulating ShenMai saponins on the transporters and the joint potential of these compounds for ShenMai-drug interaction. Circulating saponins and their pharmacokinetics were characterized in rats receiving a 30-min infusion of ShenMai at 10 mL/kg. Inhibition of human OATP1B1/1B3 and rat Oatp1b2 by the individual saponins was investigated in vitro; the compounds' joint inhibition was also assessed in vitro and the data was processed using the Chou-Talalay method. Plasma protein binding was assessed by equilibrium dialysis. Altogether, 49 saponins in ShenMai were characterized and graded into: 10-100 µmol/day (compound doses from ShenMai; 7 compounds), 1-10 µmol/day (17 compounds), and <1 µmol/day (25 compounds, including Maidong ophiopogonins). After dosing, circulating saponins were protopanaxadiol-type ginsenosides Rb1, Rb2, Rc, Rd, Ra1, Rg3, Ra2, and Ra3, protopanaxatriol-type ginsenosides Rg1, Re, Rg2, and Rf, and ginsenoside Ro. The protopanaxadiol-type ginsenosides exhibited maximum plasma concentrations of 2.1-46.6 µmol/L, plasma unbound fractions of 0.4-1.0% and terminal half-lives of 15.6-28.5 h (ginsenoside Rg3, 1.9 h), while the other ginsenosides exhibited 0.1-7.7 µmol/L, 20.8-99.2%, and 0.2-0.5 h, respectively. The protopanaxadiol-type ginsenosides, ginsenosides without any sugar attachment at C-20 (except ginsenoside Rf), and ginsenoside Ro inhibited OATP1B3 more potently (IC50, 0.2-3.5 µmol/L) than the other ginsenosides (≥22.6 µmol/L). Inhibition of OATP1B1 by ginsenosides was less potent than OATP1B3 inhibition. Ginsenosides Rb1, Rb2, Rc, Rd, Ro, Ra1, Re, and Rg2 likely contribute the major part of OATP1B3-mediated ShenMai-drug interaction potential, in an additive and time-related manner.


Subject(s)
Drugs, Chinese Herbal/pharmacokinetics , Ginsenosides/pharmacokinetics , Liver-Specific Organic Anion Transporter 1/antagonists & inhibitors , Solute Carrier Organic Anion Transporter Family Member 1B3/antagonists & inhibitors , Administration, Intravenous , Animals , Drug Combinations , Drug Interactions , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/metabolism , Ginsenosides/administration & dosage , Ginsenosides/blood , Ginsenosides/metabolism , Humans , Liver-Specific Organic Anion Transporter 1/metabolism , Male , Ophiopogon/chemistry , Panax/chemistry , Protein Binding , Rats, Sprague-Dawley , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism
10.
Phytother Res ; 32(8): 1647-1650, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29672961

ABSTRACT

The use and significance of baicalin, the main bioactive component found in Radix Scutellaria, have been on the rise due to its interesting pharmacological properties. Baicalin, a low passive permeability compound, is directly absorbed from the upper intestine and its hepatic elimination is dominant. However, interaction but no transport studies have implicated organic anion­transporting polypeptides in its cellular uptake. By using mammalian cells stably expressing the uptake transporters of interest, we are showing that baicalin is a potent substrate of Organic anion­transporting polypeptide 2B1 (OATP2B1) and less potent substrate of OATP1B3. OATP2B1 and OATP1B3 transport baicalin and may play a role in the hepatic uptake of baicalin formed in the intestine.


Subject(s)
Flavonoids/metabolism , Organic Anion Transporters/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Animals , Biological Transport , Dogs , HEK293 Cells , Humans , Intestinal Mucosa/metabolism , Liver/metabolism , Madin Darby Canine Kidney Cells
SELECTION OF CITATIONS
SEARCH DETAIL