Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 162
Filter
1.
Br J Pharmacol ; 161(8): 1793-805, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20726987

ABSTRACT

BACKGROUND AND PURPOSE: Recently, we identified etodolac as a possible ligand for the human intestinal proton-couple peptide transporter (hPEPT1). This raised the possibility that other non-steroidal anti-inflammatory drugs, and especially ibuprofen, could also interact with hPEPT1. Here, we have assessed the interactions of ibuprofen with hPEPT1. EXPERIMENTAL APPROACH: The uptake of [(14)C]Gly-Sar, [(3)H]Ibuprofen and other radio-labelled compounds were investigated in Madin-Darby canine kidney cells (MDCK)/hPEPT1, MDCK/Mock, LLC-PK(1) or Caco-2 cells. The transepithelial transport of ibuprofen and hPEPT1 substrates was investigated in Caco-2 cell monolayers. KEY RESULTS: Ibuprofen concentration dependently inhibited hPEPT1-mediated uptake of Gly-Sar in MDCK/hPEPT1 cells (K(i)(app) = 0.4 mM) but uptake of ibuprofen in Caco-2 cells and MDCK/hPEPT1 cells was not inhibited by hPEPT1 substrates. The maximum uptake rate for Gly-Sar uptake was reduced from 522 pmol·min(-1)·cm(-2) to 181 pmol·min(-1)·cm(-2) and 78 pmol·min(-1)·cm(-2) in the presence of 0.5 mM and 1 mM ibuprofen, respectively. The interaction between ibuprofen and hPEPT1 was thus non-competitive. In LLC-PK1 cells, ibuprofen (1 mM) did not influence the transporter-mediated uptake of glycine or α-methyl-D-glycopyranoside. In Caco-2 cell monolayers the absorptive transport of δ-aminolevulinic acid was reduced by 23% and 48% by ibuprofen (1 and 10 mM), respectively. Likewise the transport of Gly-Sar was reduced by 23% in the presence of ibuprofen (1 mM). CONCLUSIONS AND IMPLICATIONS: Ibuprofen is a non-competitive inhibitor of hPEPT1. As ibuprofen reduced the transepithelial transport of δ-aminolevulinic acid, drug-drug interactions between ibuprofen and hPEPT1 drug substrates at their site of absorption are possible if administered together.


Subject(s)
Biological Transport/drug effects , Ibuprofen/pharmacology , Symporters/antagonists & inhibitors , Animals , Caco-2 Cells , Cell Line , Cell Line, Transformed , Dipeptides/metabolism , Dogs , Drug Interactions , Glycine/metabolism , Humans , Ibuprofen/pharmacokinetics , Methylglucosides/pharmacokinetics , Peptide Transporter 1
2.
Drug Metab Dispos ; 37(3): 602-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19074526

ABSTRACT

A small GTP-binding protein, Rab8, is essential for apical localization of oligopeptide transporter PEPT1/SLC15A1 and sodium/glucose cotransporter SGLT1/SLC5A1 in small intestine; deficiency of rab8 gene results in mislocalization and reduced expression of these transporters. Here, we examined the role of PEPT1 and SGLT1 in vivo in gastrointestinal absorption of a beta-lactam antibiotic, cefixime, and alpha-methyl-d-glycopyranoside (alpha-MDG), respectively, using rab8 gene knockout [rab8(-/-)] mice as experimental animals deficient in those transporters. Plasma concentration of cefixime and alpha-MDG after oral administration in rab8(-/-) mice was much lower than that in wild-type mice, whereas such reduction in oral absorption was not observed for antipyrine, membrane permeation of which is not transporter-mediated. Uptake of cefixime from the apical side of isolated small intestine assessed by means of the everted sac method in wild-type mice was decreased in the presence of excess unlabeled glycylsarcosine, a PEPT1 substrate. In contrast, the uptake in rab8(-/-) mice was much lower than that in wild-type mice and comparable with that of an extracellular marker, mannitol, suggesting that the apical membrane permeability of cefixime was reduced in rab8(-/-) mice. Uptake of cefixime in wild-type mice was pH-dependent, being higher at lower pH, whereas that in rab8(-/-) mice remained at the background level at all pH values examined. These results suggest that PEPT1 and SGLT1 play an important role in gastrointestinal absorption of cefixime and alpha-MDG, respectively, in vivo in mice. The present findings also illustrate the pharmacokinetic influence of the sorting machinery protein Rab8.


Subject(s)
Cefixime/pharmacokinetics , Intestinal Absorption/physiology , Methylglucosides/pharmacokinetics , Sodium-Glucose Transporter 1/physiology , Symporters/physiology , rab GTP-Binding Proteins/physiology , Animals , Female , Intestine, Small/metabolism , Male , Mice , Mice, Knockout , Peptide Transporter 1 , rab GTP-Binding Proteins/genetics
3.
Med Phys ; 35(8): 3471-81, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18777907

ABSTRACT

The authors developed a transmission-dispersion model to estimate dispersion in blood sampling systems and to calculate dispersion-free input functions needed for kinetic analysis. Transport of molecules through catheters was considered in two parts: a central part with convective transmission of molecules and a stagnant layer that molecules may enter and leave. The authors measured dispersion caused by automatic and manual blood sampling using three PET tracers that distribute differently in blood (C15O, H2(15)O, and 11C-methylglucose). For manual sampling, dispersion was negligible. For the automated sampling procedure, characteristic parameters were calibrated for each tracer, and subsequently used in calculating dispersion-free input functions following real bolus injections. This led to shapes of dispersion-free input functions C(i)(t) that had sharper peaks than the measured C(o)(t), and the authors quantified the effect of correcting for dispersion before kinetic modeling. The transmission-dispersion model quantitatively takes apart effects of transmission and dispersion, it has transparent noise properties associated with each component, and it does not require deconvolution to calculate dispersion-free input functions. Once characteristic parameters are estimated, input functions can be corrected before applying kinetic models. This allows bias-free estimation of kinetic parameters such as blood flow.


Subject(s)
Algorithms , Catheterization/methods , Positron-Emission Tomography/methods , Radiopharmaceuticals , Humans , Methylglucosides/blood , Methylglucosides/pharmacokinetics , Models, Biological , Radiopharmaceuticals/blood , Radiopharmaceuticals/pharmacokinetics
4.
J Agric Food Chem ; 56(8): 2852-6, 2008 Apr 23.
Article in English | MEDLINE | ID: mdl-18376843

ABSTRACT

4-(3,4-Dihydroxybenzoyloxymethyl)phenyl- O-beta-D-glucopyranoside (OV-16) is a polyphenolic glycoside isolated from oregano (Origanum vulgare L.), which is a popular Chinese herb and a common spice in Western diet. To understand the biotransformation and pharmacokinetics of OV-16, rats were orally administered OV-16 and oregano decoction. Blood samples were withdrawn at specific time points. The presence of OV-16 and its metabolites protocatechuic acid (PCA) and p-hydroxybenzyl alcohol (HBA) in serum were determined by HPLC method, whereas their conjugated metabolites were assayed indirectly through hydrolysis with beta-glucuronidase and sulfatase. Our results showed that when OV-16 was orally administered, free forms of OV-16, PCA, and HBA were not present in blood and the major metabolites were the glucuronides/sulfates of PCA and HBA sulfate. The serum metabolites of OV-16 exhibited free radical scavenging activity. When oregano decoction was given, the glucuronides and sulfates of PCA were the major metabolites in blood.


Subject(s)
Antioxidants/isolation & purification , Antioxidants/pharmacokinetics , Methylglucosides/metabolism , Methylglucosides/pharmacokinetics , Origanum/chemistry , Animals , Benzyl Alcohols/blood , Chromatography, High Pressure Liquid , Glucuronides/blood , Hydroxybenzoates/blood , Male , Methylglucosides/blood , Rats , Rats, Sprague-Dawley , Sulfates/blood
5.
Acta Physiol (Oxf) ; 193(1): 67-78, 2008 May.
Article in English | MEDLINE | ID: mdl-18005215

ABSTRACT

AIM: To investigate whether the increase in mucosal permeability in the duodenum, induced by luminal hypotonicity, also occurs in the stomach and the jejunum and whether this increase in permeability can be explained by epithelial injury. METHODS: The stomach, duodenum or jejunum of the anaesthetized rat were perfused with a hypotonic solution and effects on mucosal permeability (blood-to-lumen clearance of radioactive probes); luminal alkalinization and net fluid flux were determined in the absence and presence of cyclooxygenase inhibition. RESULTS: The hypotonicity-induced (50 mM NaCl) increase in duodenal mucosal permeability was markedly larger in cyclooxygenase-2-inhibited animals than in controls and associated with a 20% decrease in luminal alkalinization and increased fluid absorption. Perfusion with 50 mM NaCl increased duodenal mucosal permeability to all probes investigated, i.e. (14)C-urea, (14)C-methyl-D-glucose, (51)Cr-EDTA and (14)C-inulin. The percentage increase in permeability was the greatest for inulin and the lowest for urea. Luminal hypotonicity caused superficial villous tip damage in some but not in all duodenal specimens but there was no difference in morphology between controls and cyclooxygenase-2-inhibited animals. Jejunum, but not the stomach, responded to luminal hypotonicity by increasing net fluid absorption, mucosal permeability (greater than sixfold) and the rate of luminal alkalinization (>100%). CONCLUSIONS: The stomach does not respond while the jejunum is more sensitive to hypotonicity-induced increase in mucosal permeability than the duodenum. The hypotonicity-induced increase in duodenal mucosal permeability most probably constitutes a physiological mechanism that entails widening of paracellular pathways, which facilitates the transport of osmolytes into the lumen.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Intestinal Mucosa/drug effects , Upper Gastrointestinal Tract/drug effects , Animals , Cyclooxygenase 2 Inhibitors/pharmacology , Duodenum/drug effects , Duodenum/metabolism , Duodenum/pathology , Gastric Mucosa/metabolism , Hydrogen-Ion Concentration/drug effects , Hypotonic Solutions/pharmacology , Indomethacin/pharmacology , Intestinal Absorption/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Inulin/pharmacokinetics , Isoxazoles/pharmacology , Jejunum/drug effects , Jejunum/metabolism , Male , Methylglucosides/pharmacokinetics , Osmolar Concentration , Permeability/drug effects , Rats , Rats, Inbred Strains , Sodium Chloride/pharmacology , Stomach/drug effects , Upper Gastrointestinal Tract/metabolism , Urea/pharmacokinetics
6.
Am J Physiol Endocrinol Metab ; 292(3): E668-76, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17062842

ABSTRACT

Appropriate partitioning of nutrients between the mother and conceptus is a major determinant of pregnancy success, with placental transfer playing a key role. Insulin-like growth factors (IGFs) increase in the maternal circulation during early pregnancy and are predictive of fetal and placental growth. We have previously shown in the guinea pig that increasing maternal IGF abundance in early to midpregnancy enhances fetal growth and viability near term. We now show that this treatment promotes placental transport to the fetus, fetal substrate utilization, and nutrient partitioning near term. Pregnant guinea pigs were infused with IGF-I, IGF-II (both 1 mg.kg-1.day-1) or vehicle subcutaneously from days 20-38 of pregnancy (term=69 days). Tissue uptake and placental transfer of the nonmetabolizable radio analogs [3H]methyl-D-glucose (MG) and [14C]aminoisobutyric acid (AIB) in vivo was measured on day 62. Early pregnancy exposure to elevated maternal IGF-I increased placental MG uptake by>70% (P=0.004), whereas each IGF increased fetal plasma MG concentrations by 40-50% (P<0.012). Both IGFs increased fetal tissue MG uptake (P<0.048), whereas IGF-I also increased AIB uptake by visceral organs (P=0.046). In the mother, earlier exposure to either IGF increased AIB uptake by visceral organs (P<0.014), whereas IGF-I also enhanced uptake of AIB by muscle (P=0.044) and MG uptake by visceral organs (P=0.016) and muscle (P=0.046). In conclusion, exogenous maternal IGFs in early pregnancy sustainedly increase maternal substrate utilization, placental transport of MG to the fetus, and fetal utilization of substrates near term. This was consistent with the previously observed increase in fetal growth and survival following IGF treatment.


Subject(s)
Food , Maternal-Fetal Exchange/drug effects , Placenta/metabolism , Pregnancy, Animal , Somatomedins/pharmacology , Aminoisobutyric Acids/pharmacokinetics , Animals , Biological Transport , Female , Fetal Weight/drug effects , Gestational Age , Guinea Pigs , Heart/drug effects , Heart/embryology , Litter Size/drug effects , Methylglucosides/pharmacokinetics , Placenta/anatomy & histology , Placenta/drug effects , Pregnancy , Pregnancy, Animal/drug effects , Term Birth
7.
Am J Physiol Renal Physiol ; 291(6): F1213-23, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16788146

ABSTRACT

The product of gene RSC1A1, named RS1, participates in transcriptional and posttranscriptional regulation of the sodium-d-glucose cotransporter SGLT1. Using coexpression in oocytes of Xenopus laevis, posttranscriptional inhibition of human SGLT1 (hSGLT1) and some other transporters by human RS1 (hRS1) was demonstrated previously. In the present study, histidine-tagged hRS1 was expressed in oocytes or Sf9 cells and purified using nickel(II)-charged nitrilotriacetic acid-agarose. hRS1 protein was injected into oocytes expressing hSGLT1 or the human organic cation transporter hOCT2, and the effect on hSGLT1-mediated uptake of methyl-alpha-D-[14C]glucopyranoside ([14C]AMG) or hOCT2-mediated uptake of [14C]tetraethylammonium ([14C]TEA) was measured. Within 30 min after the injection of hRS1 protein, hSGLT1-expressed AMG uptake or hOCT2-expressed TEA uptake was inhibited by approximately 50%. Inhibition of AMG uptake was decreased when a dominant negative mutant of dynamin I was coexpressed and increased after stimulation of PKC. Inhibition remained unaltered when endocytosis was inhibited by chlorpromazine, imipramine, or filipin but was prevented when exocytosis was inhibited by botulinum toxin B or when the release of vesicles from the TGN and endosomes was inhibited by brefeldin A. Inhibition of hSGLT1-mediated AMG uptake and hOCT2-mediated TEA uptake by hRS1 protein were decreased at an enhanced intracellular AMG concentration. The data suggest that hRS1 protein exhibits glucose-dependent, short-term inhibition of hSGLT1 and hOCT2 by inhibiting the release of vesicles from the trans-Golgi network.


Subject(s)
Exocytosis/physiology , Monosaccharide Transport Proteins/metabolism , Sodium-Glucose Transporter 1/metabolism , Animals , Biological Transport/drug effects , Biological Transport/physiology , Brefeldin A/pharmacology , Carbon Radioisotopes , Dynamins/metabolism , Exocytosis/drug effects , Glucose/metabolism , Humans , Insecta , Methylglucosides/pharmacokinetics , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/pharmacology , Oocytes/cytology , Oocytes/metabolism , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2 , Protein Kinase C/metabolism , Protein Synthesis Inhibitors/pharmacology , Rats , Sodium/metabolism , Xenopus laevis , trans-Golgi Network/metabolism
8.
J Am Soc Nephrol ; 16(5): 1257-65, 2005 May.
Article in English | MEDLINE | ID: mdl-15829715

ABSTRACT

Cadmium (Cd2+) poisoning causes severe renal disorders manifested by defects in reabsorptive transport of various compounds. It is reported here that the renal brush-border membrane Na+/glucose co-transporter-1 (SGLT1) is a molecular target for Cd2+ toxicity. In micromolar concentrations, Cd2+ acted as a noncompetitive, partial inhibitor of methyl-D-glucopyranoside uptake in vesicles from COS-7 cells transiently expressing SGLT1. In contrast, only a modest effect in the closely related Na+/myo-inositol co-transporter-1 (SMIT1) was observed. The factor responsible for this difference was the CXXC motif (X can be any residue) at the cytoplasmic end of the eighth transmembrane segment (TM8) of SGLT1. Thus, a mutational transfer of this motif conveyed Cd2+ sensitivity to SMIT1. Moreover, mimicking the inhibitory effect of Cd2+, the biarsenical molecule FlAsH-EDT2 strongly inhibited the SGLT1 that had an engineered tetracysteine motif at the cytoplasmic end of TM8. The experiments also showed that covalent binding of the sulfhydryl reactive biotin-PEO-maleimide to the SGLT1 wild type but not to the mutant lacking the CXXC motif was suppressed by Cd2+. Taken together, these results suggest that in SGLT1, Cd2+ binding to the CXXC motif induces conformational changes that cause a partial inhibition of d-glucose transport.


Subject(s)
Cadmium/toxicity , Kidney/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Monosaccharide Transport Proteins/chemistry , Monosaccharide Transport Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites/drug effects , COS Cells , Chlorocebus aethiops , Cyclohexanes/pharmacology , Cysteine/chemistry , Ionophores/pharmacology , Kidney/drug effects , Ligands , Lipid Bilayers/metabolism , Methylglucosides/pharmacokinetics , Molecular Sequence Data , Protein Structure, Tertiary , Rats , Sodium-Glucose Transporter 1
9.
Am J Physiol Renal Physiol ; 288(5): F988-96, 2005 May.
Article in English | MEDLINE | ID: mdl-15598843

ABSTRACT

Oxidative stress plays an important role in the pathogenesis of renal diseases such as diabetic nephropathy. The metabolism of excessive intracellular glucose may involve a number of processes. One consequence of excessive intracellular glucose levels is an increased rate of oxidative phosphorylation under hyperglycemic conditions, whereas another consequence is an increase in the metabolism of glucose to sorbitol by aldose reductase. In addition, hyperglycemia may result in the activation of NADPH oxidase, the production of superoxide anion, and hydrogen peroxide (H2O2). In this report, we investigate the mechanisms responsible for the H2O2 production that occurs as the consequence of hyperglycemia and the effect of H2O2 on the activity of the Na+/glucose cotransport system (SGLT) in primary cultures of renal proximal tubule cells (PTCs). When primary PTCs were cultured in the presence of high glucose, one consequence was that the Na+/glucose cotransport system was inhibited, as indicated by uptake studies utilizing alpha-methyl-D-glucoside (alpha-MG), a nonmetabolizable analog of D-glucose. Pretreatment of the cultures with either 1) aminoguanidine or pyridoxamine [inhibitors of the accumulation of advanced glycation end products (AGEs)], 2) rotenone (an inhibitor of the mitochondrial electron transport chain), or 3) apocynin or diphenylene iodonium (DPI; inhibitors of NADPH oxidase) blocked the observed changes that occurred as a consequence of the incubation of the PTCs with high glucose. Included among these changes were the observed increase in H2O2 levels, as well as an increase in lipid peroxide production, and a decrease both in the activity of catalase and in the level of glutathione (GSH), endogenous antioxidants. The high glucose-induced decrease in the level of the Na+/glucose cotransporter was similarly prevented by either aminoguanidine, rotenone, or apocynin. Thus the inhibitory effect of high glucose on both the level of the Na+/glucose cotransport system and the activity of the Na+/glucose cotransport system can be explained, at least in part, as being due to the effects of the H2O2, the consequent formation of AGEs, the increase in mitochondrial metabolism, and in NADPH oxidase activity in the PTCs. Other related changes observed in the PTCs that could be reversed by treatment with either aminoguanidine, pyridoxamine, rotenone, apocynin, or DPI included an increase in transforming growth factor-beta1 secretion and the activation of the NF-kappaB signal transduction pathway.


Subject(s)
Glucose/pharmacokinetics , Kidney Tubules, Proximal/metabolism , Monosaccharide Transport Proteins/physiology , Oxidative Stress/physiology , Animals , Cells, Cultured , Diabetic Nephropathies/metabolism , Hydrogen Peroxide/metabolism , Kidney Tubules, Proximal/cytology , Male , Methylglucosides/pharmacokinetics , Monosaccharide Transport Proteins/drug effects , Oxidative Stress/drug effects , Rabbits , Reactive Oxygen Species/metabolism , Sodium/metabolism
10.
J Cell Physiol ; 203(1): 217-25, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15368538

ABSTRACT

Both oxidative stress and epidermal growth factor (EGF) contribute to the initiation and progression of renal proximal tubular dysfunction under pathophysiologic conditions. Thus, this study was performed (1) to examine both the individual, and the combined effects of H2O2 and EGF on alpha-methyl-D-glucopyranoside uptake (alpha-MG uptake) in the primary cultured renal proximal tubule cells (PTCs), and (2) to elucidate the involvement of p44/42 mitogen activated protein kinase (MAPK) and phospholipase A2 in mediating these actions. Both H2O2 and EGF inhibited alpha-MG uptake individually, while the combination of H2O2 and EGF further potentiated the inhibitory effect on alpha-MG uptake, which was elicited by each agent. H2O2 not only caused a rapid increase in the phosphorylation of p44/42 MAPK, but also promoted the translocation of cytosolic phospholipase A2 (cPLA2) from the cytosolic to particulate fraction, and stimulated cellular [3H]-arachidonic acid (AA) release. EGF similarly activates phosphorylation of p44/42 MAPK and stimulates [3H]-AA release. When PTCs were exposed to 100 microM H2O2 and 50 ng/ml EGF simultaneously, a further increase in the phosphorylation of p44/42 MAPK, of [3H]-AA release, and of prostaglandin E2 (PGE2) production was elicited as compared with the effects of each individual agonist alone. Moreover, the additive phosphorylation of p44/42 MAPK, [3H]-AA release, and PGE2 production by H2O2 and EGF was almost completely inhibited by the p44/42 MAPK inhibitor, PD 98059. In conclusion, these results are consistent with the hypothesis that under conditions of oxidative stress, the H2O2-induced inhibition of alpha-MG uptake in the renal proximal tubule is mediated through a modulation of the EGF signaling pathway, promoting further phosphorylation of p44/42 MAPK, activation of PLA2.


Subject(s)
Arachidonic Acid/metabolism , Epidermal Growth Factor/pharmacology , Kidney Tubules, Proximal/metabolism , MAP Kinase Signaling System/drug effects , Methylglucosides/pharmacokinetics , Animals , Cells, Cultured , Drug Interactions , Glucose/metabolism , Hydrogen Peroxide/pharmacology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , MAP Kinase Signaling System/physiology , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oxidants/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Phospholipases A/metabolism , Phospholipases A2 , Rabbits , Sodium/metabolism
11.
J Cell Physiol ; 202(1): 246-54, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15389529

ABSTRACT

A growing body of evidence implicates albumin has an important regulatory function in renal proximal tubule cells (PTCs). In present study, the effect of bovine serum albumin (BSA) on 14C-alpha-methyl-D-glucopyranoside (alpha-MG) uptake and its related signal molecules were examined in the primary cultured rabbit renal PTCs. BSA significantly increased uptake of alpha-MG, a distinctive proximal tubule marker, as well as expression level of Na+/glucose cotransporters (SGLT1 and SGLT2) proteins. The BSA-induced increase of alpha-MG uptake was completely blocked by actinomycin D and cycloheximide. Neomycin or U 73122 (PLC inhibitors), BAPTA/AM or TMB-8 (intracellular Ca2+ mobilization inhibitors) completely abolished BSA-induced increase of alpha-MG uptake. BSA significantly increased IPs accumulation, but did not affect Ca2+ uptake. Effect of BSA on alpha-MG uptake was blocked by PD 98059, but did not SB 203580. BSA increased phosphorylation of p44/42 mitogen activated protein kinase (MAPK) in a time-dependent manner. NAC or catalase (antioxidants) significantly blocked BSA-induced increase of H2O2 formation and alpha-MG uptake. BSA activated NF-kappaB translocation into nucleus. PDTC, SN50, and TLCK (NF-kappaB inhibitors) also completely blocked BSA-induced increase of alpha-MG uptake, NF-kappaB p65 and phospho IkappaB-alpha activation. In conclusion, BSA stimulates alpha-MG uptake and its action is partially correlated with PLC, MAPK, or NF-kappaB signal molecules in primary cultured renal PTCs.


Subject(s)
Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , Methylglucosides/pharmacokinetics , Serum Albumin/metabolism , Signal Transduction/physiology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Animals , Antioxidants/pharmacology , Carbon Radioisotopes , Cells, Cultured , Chelating Agents/pharmacology , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Hydrogen Peroxide/metabolism , I-kappa B Proteins/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Male , Mitogen-Activated Protein Kinase 3/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Phosphorylation/drug effects , Rabbits , Serum Albumin/pharmacology , Serum Albumin, Bovine/pharmacology , Signal Transduction/drug effects , Type C Phospholipases/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
12.
Am J Physiol Renal Physiol ; 286(4): F634-42, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14644748

ABSTRACT

ANG II and Na+-glucose cotransporter have been reported to be associated with the onset of diverse renal diseases. However, the effect of ANG II on Na+-glucose cotransporter activity was not elucidated. The effects of ANG II on alpha-methyl-D-[14C]glucopyranoside (alpha-MG) uptake and its related signal pathways were examined in the primary cultured rabbit renal proximal tubule cells (PTCs). ANG II (>2 h; >10(-9) M) inhibited alpha-MG uptake in a time- and concentration-dependent manner and decreased the protein level of Na+-glucose cotransporters, the expression of which was abrogated by both actinomycin D and cycloheximide exposure. ANG II-induced inhibition of alpha-MG uptake was blocked by losartan, an ANG II type 1 (AT1) receptor blocker, but not by PD-123319, an ANG II type 2 receptor blocker. ANG II-induced inhibition of alpha-MG uptake was blocked by genistein, herbimycin A [tyrosine kinase (TK) inhibitors], mepacrine, and AACOCF3 (phospholipase A2 inhibitors), suggesting the role of TK phosphorylation and arachidonic acid (AA). Indeed, ANG II increased AA release, which was blocked by losartan or TK inhibitors. The effects of ANG II on AA release and alpha-MG uptake also were abolished by staurosporine and bisindolylmaleimide I (protein kinase C inhibitors) or PD-98059 (p44/42 MAPK inhibitor), but not SB-203580 (p38 MAPK inhibitor), respectively. Indeed, ANG II increased p44/42 MAPK activity. ANG II-induced activation of p44/42 MAPK was blocked by staurosporine. In conclusion, ANG II inhibited alpha-MG uptake via PKC-MAPK-cPLA2 signal cascade through the AT1 receptor in the PTCs.


Subject(s)
Angiotensin II/pharmacology , Kidney Tubules, Proximal/metabolism , Methylglucosides/pharmacokinetics , Signal Transduction/drug effects , Vasoconstrictor Agents/pharmacology , Angiotensin II/metabolism , Animals , Arachidonic Acid/metabolism , Carbon Radioisotopes , Cells, Cultured , Cyclic AMP/metabolism , Iodine Radioisotopes , Kidney Tubules, Proximal/drug effects , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , Rabbits , Vasoconstrictor Agents/metabolism
13.
J Nucl Med ; 44(7): 1075-81, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12843224

ABSTRACT

UNLABELLED: This study aimed to synthesize and to evaluate the biologic characteristics of (11)C labeled methyl-D-glucoside, a nonmetabolizable tracer that is selectively transported by sodium-dependent glucose transporters (SGLTs). METHODS: (11)C-Methyl-D-glucoside was prepared by methylation of glucose with (11)C-methyl triflate and was obtained as a mixture of anomers that were separated with high-performance liquid chromatography. The biodistribution of both the D- and L-isomers was determined in mice, and the presence of metabolites in the blood was investigated. The intrarenal distribution of (11)C-methyl-D-glucoside in mouse kidneys was visualized using autoradiography. Transport of alpha-methyl-D-glucoside and beta-methyl-D-glucoside by the human sodium-D-glucose cotransporter hSGLT1 was characterized after expression of hSGLT1 in oocytes of Xenopus laevis. RESULTS: The developed preparation procedure provided (11)C-methyl-D-glucoside in a total synthesis time of 20 min and a yield of 30% (decay corrected). The alpha- and beta-anomers of methyl-D-glucoside were reabsorbed from the primary urinary filtrate and showed only a minimal urinary excretion. Because methyl-L-glucoside was not reabsorbed and the reabsorption of methyl-D-glucoside was blocked by phlorizin, sodium-D-glucose cotransporters were critically involved. beta-Methyl-D-glucoside was accumulated in the kidneys to a higher extent than the alpha-anomer, suggesting that the basolateral efflux from the tubular cells is slower for the beta-anomer. Autoradiography showed that methyl-D-glucoside was accumulated throughout the renal cortex, suggesting that both sodium-D-glucose cotransporters expressed in kidney, SGLT1 and SGLT2, are involved in the uptake. The tracer was found to be metabolically stable and did not accumulate in red blood cells, which indicates that methyl-D-glucoside is not transported by the sodium-independent transporter GLUT1. Electrical measurements in Xenopus oocytes revealed that alpha-methyl-D-glucoside and beta-methyl-D-glucoside are transported by the human SGLT1 transporter with similar maximal transport rates and apparent Michaelis-Menten constant values. CONCLUSION: (11)C-Methyl-D-glucoside is a selective tracer of sodium-dependent glucose transport and can be used to visualize the function of this transporter with PET in vivo.


Subject(s)
Isotope Labeling/methods , Methylglucosides/chemical synthesis , Methylglucosides/pharmacokinetics , Monosaccharide Transport Proteins/metabolism , Oocytes/metabolism , Animals , Autoradiography/methods , Carbon Radioisotopes/blood , Carbon Radioisotopes/chemistry , Carbon Radioisotopes/isolation & purification , Carbon Radioisotopes/pharmacokinetics , Evaluation Studies as Topic , Glucose Transporter Type 1 , Humans , Male , Metabolic Clearance Rate , Methylglucosides/blood , Methylglucosides/isolation & purification , Mice , Organ Specificity , Radiopharmaceuticals/blood , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/isolation & purification , Radiopharmaceuticals/metabolism , Radiotherapy Planning, Computer-Assisted/methods , Recombinant Proteins/metabolism , Tissue Distribution , Xenopus laevis
15.
FEMS Microbiol Lett ; 218(1): 47-50, 2003 Jan 21.
Article in English | MEDLINE | ID: mdl-12583896

ABSTRACT

When protein synthesis is arrested by amino acid starvation, Escherichia coli wild-type strains show stringent control (SC) over stable RNA (sRNA) accumulation as well as a large number of other growth-related processes. One of the events under SC is transport of metabolites. Thus, under amino acid starvation, E. coli fails to accumulate the non-metabolizable glucose analog alpha-methyl-D-glucoside, whereas isogenic relaxed strains continue to take up this glucose analog. Unlike the Bacteria, most wild-type archaeal strains show relaxed control of sRNA accumulation, although a number of stringent strains have been identified. In order to determine whether stringency in the Archaea affects physiological events different from sRNA accumulation, transport of glucose analogs was examined under amino acid starvation in two stringent archaeal strains, Haloferax volcanii and Sulfolobus acidocaldarius. The experiments were performed with 2-deoxy-D-glucose, which was shown to be transported, but metabolized very limitedly. Unlike E. coli, H. volcanii and S. acidocaldarius continued to transport 2-deoxy-D-glucose under amino acid starvation. Thus, in both Archaea glucose analog transport is not under SC, as it is in E. coli.


Subject(s)
Amino Acids/metabolism , Glucose/metabolism , Haloferax volcanii/metabolism , Sulfolobus acidocaldarius/metabolism , Carbon Radioisotopes , Deoxyglucose/pharmacokinetics , Methylglucosides/pharmacokinetics , RNA, Bacterial/metabolism
16.
Kidney Blood Press Res ; 25(5): 308-14, 2002.
Article in English | MEDLINE | ID: mdl-12435877

ABSTRACT

Oxidative stress has been implicated as a primary cause of renal failure in certain renal diseases. Indeed, renal proximal tubule is a very sensitive site to oxidative stress and retains functionally fully characterized transporters. It has been reported that ginsenosides have a beneficial effect on diverse diseases including oxidative stress. However, the protective effect of ginsenosides on oxidative stress has not been elucidated in renal proximal tubule cells. Thus, we examined the effect of ginsenosides on oxidative stress-induced alteration of apical transporters and its related mechanism in renal proximal tubule cells. In the present study, hydrogen peroxide (H(2)O(2)) (>10(-5) M) inhibited alpha-methyl-D-glucopyranoside uptake in a dose-dependent manner (p < 0.05). It also inhibited Pi and Na(+) uptake. At a concentration of 20 microg/ml, total ginsenosides significantly reduced H(2)O(2)-induced inhibition of apical transporters. In contrast, protopanaxadiol (PD) and protopanaxatriol (PT) saponins exhibited a less preventive effect than total ginsenosides (p < 0.05). Furthermore, we examined its action mechanism. H(2)O(2) increased lipid peroxide formation, arachidonic acid (AA) release, and Ca(2+) uptake. These effects on H(2)O(2) were significantly prevented by total ginsenosides and PD or PT sanponins. However, total ginsenosides appear to be more protective than PD and PT saponins (p < 0.05). In conclusion, ginsenosides prevented H(2)O(2)-induced inhibition of apical transporters via a decrease in oxidative stress, AA release, and Ca(2+) uptake in primary cultured renal proximal tubule cells.


Subject(s)
Ginsenosides/pharmacology , Hydrogen Peroxide/pharmacology , Kidney Tubules, Proximal/metabolism , Membrane Transport Proteins/drug effects , Membrane Transport Proteins/metabolism , Animals , Arachidonic Acid/metabolism , Calcium/pharmacokinetics , Dose-Response Relationship, Drug , Hydrogen Peroxide/administration & dosage , Kidney Tubules, Proximal/cytology , Lipid Peroxides/metabolism , Male , Methylglucosides/pharmacokinetics , Oxidative Stress/physiology , Phosphorus/pharmacokinetics , Rabbits , Sapogenins/pharmacology , Sodium/pharmacokinetics , Triterpenes/pharmacology
17.
Am J Physiol Renal Physiol ; 283(1): F73-85, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12060589

ABSTRACT

The present study describes characteristic features of two clonal subpopulations of opossum kidney (OK) cells (OK(LC) and OK(HC)) that are functionally different but morphologically identical. The most impressive differences between OK(HC) and OK(LC) cells are the overexpression of Na+-K+-ATPase and type 3 Na+/H+ exchanger by the former, accompanied by an increased Na+-K+-ATPase activity (57.6 +/- 5.6 vs. 30.0 +/- 0.1 nmol P(i). mg protein(-1). min(-1)); the increased ability to translocate Na+ from the apical to the basolateral surface; and the increased Na+-dependent pH(i) recovery (0.254 +/- 0.016 vs. 0.094 +/- 0.011 pH units/s). Vmax values (in pH units/s) for Na+-dependent pHi recovery in OK(HC) cells (0.00521 +/- 0.0004) were twice (P < 0.05) those in OK(LC) (0.00202 +/- 0.0001), with similar Km values (in mM) for Na+ (OK(LC), 21.0 +/- 5.5; OK(HC), 14.0 +/- 5.6). In addition, we measured the activities of transporters (organic ions, alpha-methyl-D-glucoside, L-type amino acids, and Na+ and enzymes (adenylyl cyclase, aromatic L-amino acid decarboxylase, and catechol-O-methyltransferase). The cells were also characterized morphologically by optical and scanning electron microscopy and karyotyped. It is suggested that OK(LC) and OK(HC) cells constitute an interesting cell model for the study of renal epithelial physiology and pathophysiology, namely, hypertension.


Subject(s)
Amiloride/analogs & derivatives , Kidney/cytology , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Amiloride/pharmacology , Animals , Aromatic-L-Amino-Acid Decarboxylases/metabolism , Biological Transport/drug effects , Biological Transport/physiology , Catechol O-Methyltransferase/metabolism , Clone Cells , Diuretics/pharmacology , Epithelial Cells/chemistry , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Hypertension, Renal/metabolism , Immunoblotting , LLC-PK1 Cells , Leucine/pharmacokinetics , Levodopa/pharmacokinetics , Methylglucosides/pharmacokinetics , Microscopy, Electron, Scanning , Opossums , Parathyroid Hormone/pharmacology , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/analysis , Sodium-Potassium-Exchanging ATPase/analysis , Swine , p-Aminohippuric Acid/pharmacokinetics
18.
Am J Physiol Regul Integr Comp Physiol ; 282(3): R738-43, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11832394

ABSTRACT

In the chicken intestine, the reduction in Na(+) intake led to a decrease in the transport of alpha-methyl-D-glucoside in the ileum (reduction of 42%) and in the rectum (51%). These reductions were reversed within 24 h after resalination and were inversely correlated to the changes in aldosterone plasma concentration. The reduction in intestinal hexose transport in the low Na(+)-fed animals was due to a decrease in the number of Na(+)-dependent D-glucose cotransporters (SGLT1) in the rectum (46%) and in the ileum (38%). Northern blot analysis showed that specific SGLT1 mRNA was expressed in the jejunum, ileum, and rectum. The amount of SGLT1 mRNA was the same in all intestinal regions and was not affected by Na(+) intake, supporting the view that the effects of dietary Na(+) on intestinal hexose transport involve posttranscriptional regulation of SGLT1. This study suggests that changes in SGLT1 expression may be involved in the homeostasis of Na(+).


Subject(s)
Membrane Glycoproteins/metabolism , Monosaccharide Transport Proteins/metabolism , Sodium, Dietary/pharmacology , Animals , Chickens , Enterocytes/metabolism , Immunoblotting , Male , Membrane Glycoproteins/genetics , Methylglucosides/pharmacokinetics , Microvilli/metabolism , Monosaccharide Transport Proteins/genetics , RNA, Messenger/metabolism , Sodium-Glucose Transporter 1
19.
Kidney Blood Press Res ; 25(6): 375-83, 2002.
Article in English | MEDLINE | ID: mdl-12590201

ABSTRACT

Human envenomation caused by bee stings has been reported to cause acute renal failure and the pathogenetic mechanisms of these renal functional changes are still unclear. Bee venom is also a complex mixture of enzymes and proteins. Thus, this study was conducted to examine the effects of bee venom (BV, Apis mellifera) fractions on apical transporters' activity and its related signal pathways in primary cultured renal proximal tubule cells. Whole BV was extracted into three fractions according to solubility [a water-soluble fraction (BVA), an ethylacetate-soluble fraction (BVE), and a hexane-soluble fraction (BVH)]. BVA fraction was further separated to three portions according to molecular weights: BF1 (>20 kD), BF2 (10-20 kD), and BF3 (<10 kD). Each fraction was treated to the PTCs to the ratio of BV (1 microg/ml). BVA (930 ng/ml) significantly decreased cell viability, but BVH (27 ng/ml) and BVE (43 ng/ml) did not. BF3 (710 ng/ml) among BVA fractions predominantly decreased cell viability and inhibited alpha-methyl-D-glucopyranoside (alpha-MG), phosphate (Pi), and Na(+) uptake. In addition, BF3 increased [(3)H] arachidonic acid release, lipid peroxide formation, and Ca(2+) uptake. These effects of BF3 were blocked by mepacrine and AACOCF(3) (phospholipase A(2) inhibitors) or N-acetylcysteine, vitamin C, and vitamin E (antioxidants). In conclusion, BF3 (<10 kD) among BV fractions is the most effective portion in BV-induced inhibition of alpha-MG, P(i), and Na(+) uptake and these effects of BF3 are associated with phospholipase A(2)-oxidative stress-Ca(2+) signal cascade in the primary cultured rabbit renal proximal tubule cells.


Subject(s)
Bee Venoms/pharmacology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Animals , Arachidonic Acid/metabolism , Bee Venoms/metabolism , Biological Transport/drug effects , Calcium/metabolism , Cell Survival , Male , Methylglucosides/pharmacokinetics , Oxidative Stress/drug effects , Phosphates/pharmacokinetics , Phospholipases A/metabolism , Rabbits , Signal Transduction/drug effects , Sodium/pharmacokinetics , Solubility , Tritium
20.
Toxicol Appl Pharmacol ; 177(3): 163-73, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11749115

ABSTRACT

Chronic exposure to cadmium can result in renal glycosuria. Previously, we reported that cadmium reduced the relative abundance of the sodium-glucose cotransporter mRNA (Blumenthal et al., Toxicol. Appl. Pharmacol.149, 49-54, 1998). To investigate this phenomenon further, we isolated full-length cDNA clones encoding both high- and low-affinity sodium-dependent glucose transporters SGLT1 and SGLT2, respectively, from cultured mouse kidney cortical cells. We also amplified a fragment of another putative sodium-glucose cotransporter with homology to the known SAAT1/pSGLT2 or SGLT3 from our cultured cells and named it SGLT3. In order to examine the effect of cadmium on these transporters, primary cultures of mouse kidney cortical cells were exposed to micromolar concentrations of cadmium for 24 h and levels of SGLT1, SGLT2, and SGLT3 mRNA were determined by semiquantitative RT-PCR. Five to 10 microM of cadmium inhibited sodium-dependent uptake of the glucose analog, alpha-methyl D-glucopyranoside and progressively reduced the level of SGLT1. Cadmium also inhibited SGLT2 mRNA by 37%, but no further decline was observed at concentrations of cadmium greater than 5 microM. While cadmium inhibited SGLT1 and SGLT2, it significantly stimulated the expression of SGLT3 by fivefold. These results imply that individual sodium-glucose cotransporter mRNA species are not regulated in a similar fashion. In addition, the isolation of three separate SGLT species from these cultures suggests that, in addition to SGLT1 and SGLT2, glucose reabsorption by renal epithelial cells might involve additional glucose transporters such as SGLT3.


Subject(s)
Cadmium/pharmacology , Kidney Cortex/metabolism , Membrane Glycoproteins/metabolism , Monosaccharide Transport Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Biological Transport/drug effects , Cells, Cultured , Cloning, Molecular , DNA, Complementary/genetics , Dose-Response Relationship, Drug , Gene Expression/drug effects , Glucose/metabolism , Kidney Cortex/cytology , Kidney Cortex/drug effects , Membrane Glycoproteins/genetics , Methylglucosides/pharmacokinetics , Mice , Molecular Sequence Data , Monosaccharide Transport Proteins/genetics , RNA, Messenger , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Sodium/metabolism , Sodium-Glucose Transport Proteins , Sodium-Glucose Transporter 1 , Sodium-Glucose Transporter 2
SELECTION OF CITATIONS
SEARCH DETAIL
...