Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Drug Metab Dispos ; 46(7): 924-933, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29712725

ABSTRACT

Cerivastatin (CER) was withdrawn from the world market because of lethal rhabdomyolysis. Coadministrations of CER and cyclosporine A (CsA) or gemfibrozil (GEM) have been reported to increase the CER blood concentration. CsA is an inhibitor of organic anion transporting polypeptide (OATP)1B1 and CYP3A4, and GEM and its glucuronide (GEM-glu) inhibit OATP1B1 and CYP2C8. The purpose of this study was to describe the transporter-/enzyme-mediated drug-drug interactions (DDIs) of CER with CsA or GEM based on unified physiologically based pharmacokinetic (PBPK) models and to investigate whether the DDIs can be quantitatively analyzed by a bottom-up approach. Initially, the PBPK models for CER and GEM/GEM-glu were constructed based on the previously reported standard protocols. Next, the drug-dependent parameters were optimized by Cluster Newton Method. Thus, described concentration-time profiles for CER and GEM/GEM-glu agreed well with the clinically observed data. The DDIs were then simulated using the established PBPK models with previously obtained in vitro inhibition constants of CsA or GEM/GEM-glu against the OATP1B1 and cytochrome P450s. DDIs with the inhibitors were underestimated compared with observed data using the geometric means of reported values. To search for better described parameters within the range of in vitro values, sensitivity analyses were performed for DDIs of CER. Using the in vitro parameter sets selected by sensitivity analyses, these DDIs were well reproduced, indicating that the present PBPK models were able to describe adequately the clinical DDIs based on a bottom-up approach. The approaches in this study would be applicable to the prediction of other DDIs involving both transporters and metabolic enzymes.


Subject(s)
Biological Transport/physiology , Drug Interactions/physiology , Pyridines/pharmacokinetics , Cyclosporine/pharmacokinetics , Cytochrome P-450 CYP2C8/metabolism , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme System/metabolism , Gemfibrozil/pharmacokinetics , Glucuronides/pharmacokinetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/drug effects , Liver/metabolism , Liver-Specific Organic Anion Transporter 1/metabolism , Models, Biological
2.
Biol Pharm Bull ; 40(11): 1990-1995, 2017.
Article in English | MEDLINE | ID: mdl-29093349

ABSTRACT

Riboflavin (vitamin B2) plays a role in various biochemical oxidation-reduction reactions. Flavin mononucleotide (FMN) and FAD, the biologically active forms, are made from riboflavin. Riboflavin transporters (RFVTs), RFVT1-3/Slc52a1-3, have been identified. However, the roles of human (h)RFVTs in FMN and FAD homeostasis have not yet been fully clarified. In this study, we assessed the contribution of each hRFVT to riboflavin, FMN and FAD uptake and efflux using in vitro studies. The transfection of hRFVTs increased cellular riboflavin concentrations. The uptake of riboflavin by human embryonic kidney cells transfected with hRFVTs was significantly increased, and the efflux was accelerated in a time-dependent manner. However, the uptake and efflux of FMN and FAD hardly changed. These results strongly suggest that riboflavin, rather than FMN or FAD, passes through plasma membranes via hRFVTs. Our findings could suggest that hRFVTs are involved in riboflavin homeostasis in the cells, and that FMN and FAD concentrations are regulated by riboflavin kinase and FAD synthase.


Subject(s)
Cell Membrane/metabolism , Membrane Transport Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Riboflavin/metabolism , Flavin Mononucleotide/metabolism , Flavin-Adenine Dinucleotide/metabolism , HEK293 Cells , Humans , Membrane Transport Proteins/genetics , Protein Transport , Receptors, G-Protein-Coupled/genetics , Transfection
3.
J Pharm Sci ; 106(9): 2715-2726, 2017 09.
Article in English | MEDLINE | ID: mdl-28479356

ABSTRACT

Quantitative analysis of transporter- and enzyme-mediated complex drug-drug interactions (DDIs) is challenging. Repaglinide (RPG) is transported into the liver by OATP1B1 and then is metabolized by CYP2C8 and CYP3A4. The purpose of this study was to describe the complex DDIs of RPG quantitatively based on unified physiologically based pharmacokinetic (PBPK) models using in vitro Ki values for OATP1B1, CYP3A4, and CYP2C8. Cyclosporin A (CsA) or gemfibrozil (GEM) increased the blood concentrations of RPG. The time profiles of RPG and the inhibitors were analyzed by PBPK models, considering the inhibition of OATP1B1 and CYP3A4 by CsA or OATP1B1 inhibition by GEM and its glucuronide and the mechanism-based inhibition of CYP2C8 by GEM glucuronide. RPG-CsA interaction was closely predicted using a reported in vitro Ki,OATP1B1 value in the presence of CsA preincubation. RPG-GEM interaction was underestimated compared with observed data, but the simulation was improved with the increase of fm,CYP2C8. These results based on in vitro Ki values for transport and metabolism suggest the possibility of a bottom-up approach with in vitro inhibition data for the prediction of complex DDIs using unified PBPK models and in vitro fm value of a substrate for multiple enzymes should be considered carefully for the prediction.


Subject(s)
Carbamates/blood , Cyclosporine/pharmacology , Enzyme Inhibitors/pharmacology , Gemfibrozil/pharmacology , Hypoglycemic Agents/blood , Piperidines/blood , Biological Transport/drug effects , Carbamates/metabolism , Carbamates/pharmacology , Computer Simulation , Cytochrome P-450 CYP2C8/metabolism , Cytochrome P-450 CYP2C8 Inhibitors/pharmacology , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Liver-Specific Organic Anion Transporter 1/metabolism , Models, Biological , Piperidines/metabolism , Piperidines/pharmacology
4.
Sci Rep ; 6: 27557, 2016 06 08.
Article in English | MEDLINE | ID: mdl-27272163

ABSTRACT

Homeostasis of riboflavin should be maintained by transporters. Previous in vitro studies have elucidated basic information about riboflavin transporter RFVT3 encoded by SLC52A3 gene. However, the contribution of RFVT3 to the maintenance of riboflavin homeostasis and the significance in vivo remain unclear. Here, we investigated the physiological role of RFVT3 using Slc52a3 knockout (Slc52a3-/-) mice. Most Slc52a3-/- mice died with hyperlipidemia and hypoglycemia within 48 hr after birth. The plasma and tissue riboflavin concentrations in Slc52a3-/- mice at postnatal day 0 were dramatically lower than those in wild-type (WT) littermates. Slc52a3-/- fetuses showed a lower capacity of placental riboflavin transport compared with WT fetuses. Riboflavin supplement during pregnancy and after birth reduced neonatal death and metabolic disorders. To our knowledge, this is the first report to indicate that Rfvt3 contributes to placental riboflavin transport, and that disruption of Slc52a3 gene caused neonatal mortality with hyperlipidemia and hypoglycemia owing to riboflavin deficiency.


Subject(s)
Membrane Transport Proteins/genetics , Riboflavin Deficiency/genetics , Riboflavin/blood , Animals , Animals, Newborn/genetics , Female , Humans , Hyperlipidemias/genetics , Hyperlipidemias/mortality , Hyperlipidemias/pathology , Hypoglycemia/genetics , Hypoglycemia/mortality , Hypoglycemia/pathology , Mice , Mice, Knockout , Placenta/metabolism , Placenta/pathology , Pregnancy , Riboflavin/genetics , Riboflavin Deficiency/mortality , Riboflavin Deficiency/pathology
5.
Am J Physiol Gastrointest Liver Physiol ; 306(2): G102-10, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24264046

ABSTRACT

Riboflavin, also known as vitamin B2, is transported across the biological membrane into various organs by transport systems. Riboflavin transporter RFVT3 is expressed in the small intestine and has been suggested to localize in the apical membranes of the intestinal epithelial cells. In this study, we investigated the functional involvement of RFVT3 in riboflavin absorption using intestinal epithelial T84 cells and mouse small intestine. T84 cells expressed RFVT3 and conserved unidirectional riboflavin transport corresponding to intestinal absorption. Apical [(3)H]riboflavin uptake was pH-dependent in T84 cells. This uptake was not affected by Na(+) depletion at apical pH 6.0, although it was significantly decreased at apical pH 7.4. The [(3)H]riboflavin uptake from the apical side of T84 cells was prominently inhibited by the RFVT3 selective inhibitor methylene blue and significantly decreased by transfection of RFVT3-small-interfering RNA. In the gastrointestinal tract, RFVT3 was expressed in the jejunum and ileum. Mouse jejunal and ileal permeabilities of [(3)H]riboflavin were measured by the in situ closed-loop method and were significantly reduced by methylene blue. These results strongly suggest that RFVT3 would functionally be involved in riboflavin absorption in the apical membranes of intestinal epithelial cells.


Subject(s)
Intestinal Absorption/physiology , Membrane Transport Proteins/genetics , Membrane Transport Proteins/physiology , Riboflavin/metabolism , Animals , Biological Transport, Active , Cells, Cultured , HEK293 Cells , Humans , Ileum/drug effects , Ileum/metabolism , In Situ Hybridization , Intestinal Absorption/drug effects , Intestinal Absorption/genetics , Intestinal Mucosa/metabolism , Jejunum/drug effects , Jejunum/metabolism , Membrane Transport Proteins/drug effects , Methylene Blue/pharmacology , Mice , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Transfection
6.
Eur J Pharmacol ; 714(1-3): 281-7, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23911957

ABSTRACT

Riboflavin (vitamin B2) acts as an intermediary during various biochemical oxidation-reduction reactions in the liver. Hepatic riboflavin homeostasis is suggested to be maintained through its transporter(s). Riboflavin transporters, RFVT2/Slc52a2 and RFVT3/Slc52a3, have been identified in rodents. However, the role of each RFVT in the hepatic homeostasis of riboflavin has not yet been fully clarified. In this study, we assessed the contribution of each RFVT to riboflavin uptake into the liver using in vitro and in vivo studies. The uptake of riboflavin by mouse primary hepatocytes increased in a time-dependent and a concentration-dependent manner. Riboflavin transport was independent of extracellular Na(+). However, the uptake decreased slightly along with the extracellular pH increases. Real-time PCR analysis revealed that the mRNA level of Slc52a2, or coding for mouse (m)RFVT2, in the mouse liver was 10 times higher than that of Slc52a3 (coding for mRFVT3). The uptake of riboflavin at pH 7.4 by primary hepatocytes was significantly decreased by the transfection of Slc52a2-small interfering RNA (siRNA), but not Slc52a3-siRNA. Furthermore, we also confirmed the contribution of riboflavin transporters in vivo. The riboflavin concentrations in plasma, but not in the liver, were significantly decreased in mice fed on a riboflavin-deficient diet for 8 weeks. The expression of Slc52a2 mRNA was significantly upregulated by riboflavin deprivation. These results strongly suggest that mRFVT2 was involved in hepatic riboflavin homeostasis.


Subject(s)
Homeostasis , Liver/metabolism , Membrane Transport Proteins/metabolism , Riboflavin/metabolism , Animals , Biological Transport , Hepatocytes/metabolism , Liver/cytology , Male , Mice , Mice, Inbred C57BL
7.
J Inherit Metab Dis ; 35(6): 943-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22864630

ABSTRACT

Brown-Vialetto-Van Laere syndrome (BVVLS [MIM 211530]) is a rare neurological disorder characterized by infancy onset sensorineural deafness and ponto-bulbar palsy. Mutations in SLC52A3 (formerly C20orf54), coding for riboflavin transporter 2 (hRFT2), have been identified as the molecular genetic correlate in several individuals with BVVLS. Exome sequencing of just one single case revealed that compound heterozygosity for two pathogenic mutations in the SLC52A2 gene coding for riboflavin transporter 3 (hRFT3), another member of the riboflavin transporter family, is also associated with BVVLS. Overexpression studies confirmed that the gene products of both mutant alleles have reduced riboflavin transport activities. While mutations in SLC52A3 cause decreased plasma riboflavin levels, concordant with a role of SLC52A3 in riboflavin uptake from food, the SLC52A2-mutant individual had normal plasma riboflavin concentrations, a finding in line with a postulated function of SLC52A2 in riboflavin uptake from blood into target cells. Our results contribute to the understanding of human riboflavin metabolism and underscore its role in the pathogenesis of BVVLS, thereby providing a rational basis for a high-dose riboflavin treatment.


Subject(s)
Bulbar Palsy, Progressive/genetics , Bulbar Palsy, Progressive/metabolism , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/metabolism , Mutation, Missense , Receptors, G-Protein-Coupled/genetics , Riboflavin/metabolism , Amino Acid Sequence , Base Sequence , Biological Transport, Active/genetics , Bulbar Palsy, Progressive/diagnosis , Child, Preschool , DNA Mutational Analysis , Female , Hearing Loss, Sensorineural/diagnosis , Humans , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Models, Biological , Molecular Sequence Data , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/metabolism , Sequence Homology, Amino Acid , Syndrome
8.
J Nutr ; 140(7): 1220-6, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20463145

ABSTRACT

We isolated cDNA coding a new human riboflavin transporter (hRFT)3, which exhibits 86.7 and 44.1% amino acid identity with hRFT1 and hRFT2, respectively. It was predicted to have 10 putative membrane-spanning domains. The functional characteristics of hRFT3 were examined and compared with those of its isoforms, hRFT1 and hRFT2. Real-time PCR revealed that hRFT3 mRNA was strongly expressed in the brain and salivary gland. hRFT1 mRNA was strongly expressed in the placenta and small intestine, whereas hRFT2 mRNA was most abundantly expressed in the testis and strongly in the small intestine and prostate. hRFT-mediated uptake of [3H]riboflavin was evaluated using human embryonic kidney 293 cells transiently transfected with the cDNA coding each hRFT. The apparent Michaelis-Menten constants of hRFT1, hRFT2, and hRFT3 for riboflavin were 1.38, 0.98, and 0.33 micromol/L, respectively. The hRFT-mediated [3H]riboflavin uptake was independent of extracellular Na+ and Cl(-). Specific uptake of [3H]riboflavin by hRFT2, but not hRFT1 and hRFT3, decreased as extracellular pH was changed from 5.4 to 8.4. The substrate specificities of the hRFT family were similar. hRFT-mediated uptake of [3H]riboflavin was inhibited by some riboflavin analogs, but not D-ribose, organic ions, or other vitamins. The newly isolated hRFT3 may play an important role in brain riboflavin homeostasis. Its amino acid sequence and functional characteristics are similar to those of hRFT1, but not hRFT2.


Subject(s)
Brain/metabolism , Membrane Transport Proteins/metabolism , Nerve Tissue Proteins/metabolism , Base Sequence , Cell Line , DNA Primers , DNA, Complementary , Humans , Membrane Transport Proteins/genetics , Nerve Tissue Proteins/genetics , RNA, Messenger/genetics , Receptors, G-Protein-Coupled , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...