Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Clin Nutr ; 30(4): 524-32, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21324568

ABSTRACT

BACKGROUND & AIMS: Enteroendocrine cells are thought to directly sense nutrients via α-gustducin coupled taste receptors (originally identified in the oral epithelium) to modulate the secretion of glucagon-like peptide-1 (GLP-1) and peptide YY (PYY). METHODS: We measured mRNA expression of α-gustducin and T1R3 along the human gut; immunohistochemistry was used to confirm co-localization with GLP-1. Functional implication of sweet taste receptors in glucose-stimulated secretion of GLP-1 and PYY was determined by intragastric infusion of glucose with or without lactisole (a sweet taste receptor antagonist) in 16 healthy subjects. RESULTS: α-gustducin was expressed in a region-specific manner (predominantly in the proximal gut and less in ileum and colon, P < 0.05). Both, T1R3 and α-gustducin were co-localized with GLP-1. Glucose-stimulated secretions of GLP-1 (P = 0.026) and PYY (P = 0.034) were reduced by blocking sweet receptors with lactisole. CONCLUSION: Key proteins implicated in taste signaling are present in the human gut and co-localized with GLP-1 suggesting that these proteins are functionally linked to peptide secretion from enteroendocrine cells. Glucose-stimulated secretion of GLP-1 and PYY is reduced by a sweet taste antagonist, suggesting the functional involvement of gut-expressed sweet taste receptors in glucose-stimulated secretion of both peptides in humans.


Subject(s)
Colon/drug effects , Glucagon-Like Peptide 1/metabolism , Glucose/metabolism , Ileum/drug effects , Peptide YY/metabolism , Adult , Aged , Benzene Derivatives/administration & dosage , Enteroendocrine Cells/metabolism , Female , Glucagon-Like Peptide 1/genetics , Humans , Immunohistochemistry , Male , Middle Aged , Peptide YY/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Taste , Transducin/antagonists & inhibitors , Transducin/genetics , Transducin/metabolism
2.
Nanotechnology ; 22(12): 125702, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21317490

ABSTRACT

The addition of carbon nanotubes (CNTs) to polymeric matrices or master batches has the potential to provide composites with novel properties. However, composites with a uniform dispersion of CNTs have proved to be difficult to manufacture, especially at an industrial scale. This paper reports on processing methods that overcome problems related to the control and reproducibility of dispersions. By using a high pressure homogenizer and a three-roll calendaring mill in combination, CNT reinforced epoxies were fabricated by mould casting with a well dispersed nanofiller content from 0.1 to 2 wt%. The influence of the nano-carbon reinforcements on toughness and electrical properties of the CNT/epoxies was studied. A substantial increase of all mechanical properties already appeared at the lowest CNT content of 0.1 wt%, but further raising the nanofiller concentration only led to moderate further changes. The most significant enhancement was obtained for fracture toughness, reaching up to 82%. The low percolation thresholds were confirmed by electrical conductivity measurements on the same composites yielding a threshold value of only about 0.01 wt%. As corroborated by a thorough microscopic analysis of the composites, mechanical and electrical enhancement points to the formation of an interconnected network of agglomerated CNTs.

3.
Neuroscience ; 170(1): 361-71, 2010 Sep 29.
Article in English | MEDLINE | ID: mdl-20603190

ABSTRACT

A variety of neurological diseases are characterized by disturbances of the blood-brain barrier (BBB) and its transporters. We recently introduced fibroblast growth factor treated cortical organotypic slice cultures of mice as a model for in vitro studies of the blood-brain barrier and have now further characterized the maintenance and function of transport-proteins typically expressed in the endothelium of cerebral blood vessels. The glucose transporter GLUT-1 is present in blood vessels of slice cultures derived from postnatal day 4 to 21 mice after 3 days in vitro. The endothelial multidrug resistance P-glycoprotein (P-gp) which is involved in the control of pharmacological substance transport across the blood-brain barrier is also maintained in blood vessels, most prominently in slice cultures derived from postnatal day 14 and 21 mice. To assess P-gp function, we tested rhodamine 123 transport in presence or absence of the P-gp inhibitor verapamil. Rhodamine 123-fluorescence accumulated rapidly in the vascular lumen both in acute slices and in slices cultured for 3 days in vitro. Our results provide evidence that endothelial transporters and their functional properties can be maintained in organotypic cortical slices cultures, thus making it an attractive model system for the study of the blood-brain barrier.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Blood-Brain Barrier/metabolism , Cerebral Cortex/metabolism , Endothelial Cells/metabolism , Glucose Transporter Type 1/metabolism , Animals , Animals, Newborn , Cerebral Cortex/blood supply , Cerebral Cortex/cytology , Endothelial Cells/cytology , Mice , Mice, Inbred C57BL , Organ Culture Techniques
4.
Clin Pharmacol Ther ; 88(1): 52-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20220749

ABSTRACT

Drug safety is of great importance to public health. The detrimental effects of drugs not only limit their application but also cause suffering in individual patients and evoke distrust of pharmacotherapy. For the purpose of identifying drugs that could be suspected of causing adverse reactions, we present a structure-activity relationship analysis of adverse drug reactions (ADRs) in the central nervous system (CNS), liver, and kidney, and also of allergic reactions, for a broad variety of drugs (n = 507) from the Swiss drug registry. Using decision tree induction, a machine learning method, we determined the chemical, physical, and structural properties of compounds that predispose them to causing ADRs. The models had high predictive accuracies (78.9-90.2%) for allergic, renal, CNS, and hepatic ADRs. We show the feasibility of predicting complex end-organ effects using simple models that involve no expensive computations and that can be used (i) in the selection of the compound during the drug discovery stage, (ii) to understand how drugs interact with the target organ systems, and (iii) for generating alerts in postmarketing drug surveillance and pharmacovigilance.


Subject(s)
Decision Support Techniques , Decision Trees , Drug-Related Side Effects and Adverse Reactions , Algorithms , Artificial Intelligence , Central Nervous System Diseases/chemically induced , Chemical and Drug Induced Liver Injury/etiology , Computer Simulation , Databases, Factual , Drug Evaluation, Preclinical , Drug Hypersensitivity/etiology , Forecasting , Humans , Kidney Diseases/chemically induced , Pharmaceutical Preparations/chemistry , Reproducibility of Results , Small Molecule Libraries , Software
5.
Regul Pept ; 141(1-3): 120-8, 2007 Jun 07.
Article in English | MEDLINE | ID: mdl-17276524

ABSTRACT

BACKGROUND: GLP-1 is secreted into the circulation after food intake. The main biological effects of GLP-1 include stimulation of glucose dependent insulin secretion and induction of satiety feelings. Recently, it was demonstrated in rats and humans that GLP-1 can stimulate renal excretion of sodium. Based on these data, the existence of a renal GLP-1 receptor (GLP-1R) was postulated. However, the exact localization of the GLP-1R and the mechanism of this GLP-1 action have not yet been investigated. METHODS: Primary porcine proximal tubular cells were isolated from porcine kidneys. Expression of GLP-1R was measured at the mRNA level by quantitative RT-PCR. Protein expression of GLP-1R was verified with immunocytochemistry, immunohistochemistry and Western blot analysis. Functional studies included transport assessments of sodium and glucose using three different GLP-1 concentrations (200 pM, 2 nM and 20 nM), 200 pM exendin-4 (GLP-1 analogue) and an inhibitor of the dipeptidylpeptidase IV (DPPIV) enzyme (P32/98 at 10 microM). Finally, the expression of NHE3, the predominant Na(+)/H(+) exchanger in proximal tubular cells, was also investigated. RESULTS: GLP-1R, NHE3 and DPPIV were expressed at the mRNA level in porcine proximal tubular kidney cells. GLP-1R expression was confirmed at the protein level. Staining of human and pig kidney cortex revealed that GLP-1R was predominantly expressed in proximal tubular cells. Functional assays demonstrated an inhibition of sodium re-absorption with GLP-1 after 3 h of incubation. Exendin-4 and GLP-1 in combination with P32/98 co-administration had no clear influence on glucose and sodium uptake and transport. CONCLUSION: GLP-1R is functionally expressed in porcine proximal tubular kidney cells. Addition of GLP-1 to these cells resulted in a reduced sodium re-absorption. GLP-1 had no effect on glucose re-absorption. We conclude that GLP-1 modulates sodium homeostasis in the kidney most likely through a direct action via its GLP-1R in proximal tubular cells.


Subject(s)
Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Receptors, Glucagon/metabolism , Animals , Blotting, Western , Cell Culture Techniques , Cells, Cultured , Dipeptidyl-Peptidase IV Inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Exenatide , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor , Glucose/analysis , Glucose/metabolism , Immunohistochemistry , Kinetics , Pentanoic Acids/pharmacology , Peptides/pharmacology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sodium/antagonists & inhibitors , Sodium/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/metabolism , Swine , Thiazolidines/pharmacology , Venoms/pharmacology
6.
Br J Pharmacol ; 150(3): 361-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17179942

ABSTRACT

BACKGROUND AND PURPOSE: P-glycoprotein (P-gp) is an important efflux transporter that supports the barrier function of the gut against invading antigens and against administered drugs. Since glucocorticoids, such as budesonide, are frequently used during inflammatory bowel disease we investigated how budesonide influences P-gp expression in different intestinal cell lines. EXPERIMENTAL APPROACH: LS180 and Caco-2 cells were incubated with budesonide and changes in P-gp expression were determined on mRNA, protein and functional level. The mRNA expression levels of glucocorticoid receptor (GR) and pregnane X receptor (PXR) were determined in these cell lines. PXR receptor was transiently transfected into Caco-2 cells. KEY RESULTS: Budesonide showed an induction of P-gp in LS180 cells and a down-regulation in Caco-2 cells. Expression levels of nuclear receptors revealed high expression of PXR only in LS180 cells and exclusive expression of GR in Caco-2 cells. Mifepristone, an anti-glucocorticoid, could not reverse the down-regulation of P-gp by budesonide in Caco-2 cells. In PXR-transfected Caco-2 cells the budesonide-mediated down-regulation of P-gp was abolished. Furthermore the expression of cytochrome P450 3A4 (CYP3A4), another PXR target gene, was induced in PXR-transfected Caco-2 cells after budesonide treatment. CONCLUSIONS AND IMPLICATIONS: Budesonide has the potential to influence MDR1 expression in vitro. In LS180 cells, the induction of MDR1 by budesonide probably is mediated via PXR. The mechanism of the down-regulation in Caco-2 cells still remains unclear, but GR does not seem to be involved. Further studies are required to evaluate how budesonide alters P-gp expression in vivo.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Budesonide/pharmacology , Gene Expression/drug effects , Glucocorticoids/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Caco-2 Cells , Cell Line , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Down-Regulation/drug effects , Humans , Pregnane X Receptor , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/physiology , Receptors, Steroid/drug effects , Receptors, Steroid/physiology
7.
Planta Med ; 72(6): 514-20, 2006 May.
Article in English | MEDLINE | ID: mdl-16773535

ABSTRACT

In this study the influence of green tea extract (GTE) or its component epigallocatechin gallate (EGCG) on the expression of different cytochrome P450 (CYP) isoenzymes was investigated in the human gastrointestinal epithelial cell lines LS-180 and Caco-2. Additionally, the effect of GTE or EGCG on functional activity of different CYP isoenzymes was investigated in vitro. mRNA expression levels were determined by quantitative RT-PCR and compared with protein levels. In LS-180 cells GTE, but not EGCG, significantly induced CYP1A2 mRNA expression, whereas neither CYP1A1 nor CYP3A4 mRNA expression was modulated by GTE or EGCG. In Caco-2 cells CYP1A1 as well as CYP1A2 mRNA expression was significantly increased in a dose-dependent manner by GTE and EGCG. However, EGCG alone was about 3-5-fold less effective than GTE. mRNA expression of CYP1A1 or CYP1A2 induced by the promutagen benzo[a]pyrene was significantly down-regulated by EGCG but not by GTE. CYP1A protein levels in response to GTE in Caco-2 and LS-180 cells confirmed the mRNA expression results. CYP activity was measured with CYP1A2 or CYP3A4 expressed in insect cell membranes using a luminescent method. GTE or EGCG significantly inhibited CYP1A2 and CYP3A4 function in a dose-dependent manner. Therefore, it appears that green tea moderately modulates the expression of drug-metabolizing enzymes but non-specifically inhibits the function of human CYPs. Since CYP enzymes play an important role in detoxification processes, these results might be of relevance for the prediction of the outcome of future clinical studies.


Subject(s)
Colon/cytology , Cytochrome P-450 CYP1A1/drug effects , Phytotherapy , Plant Extracts/pharmacology , Tea , Caco-2 Cells/drug effects , Catechin/administration & dosage , Catechin/analogs & derivatives , Catechin/pharmacology , Catechin/therapeutic use , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic , Humans , Plant Extracts/administration & dosage , Plant Extracts/therapeutic use , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured/drug effects
8.
Planta Med ; 72(8): 697-702, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16739069

ABSTRACT

The chemokine interleukin (IL)-8 is a cytokine involved in neutrophil attraction and activation and elevated levels have been observed in intestinal inflammation. Anti-inflammatory activities have been attributed to green tea or its major constituent (-)-epigallocatechin gallate (EGCG). In this study, we investigated the effects of a defined green tea extract (GTE) or EGCG on basal or IL-1beta-induced IL-8 expression and secretion in the human gastrointestinal epithelial cell line Caco-2. mRNA expression levels were determined by quantitative RT-PCR. GTE significantly induced IL-8 mRNA expression, which was not mediated indirectly via an induction of IL-1beta mRNA expression. EGCG only exerted a weak although significant induction of IL-8 mRNA expression at the highest concentration. Intracellular and extracellular protein levels were analyzed by an enzyme-linked immunosorbent assay. GTE and EGCG significantly decreased secreted IL-8 concentrations. Determination of intracellular and secreted IL-8 concentrations after 24 h, 48 h, and 72 h of incubation suggested that GTE specifically inhibited IL-8 secretion while inducing DE NOVO synthesis of IL-8. The IL-1beta-mediated increase of IL-8 secretion was significantly inhibited by GTE in a dose-dependent manner. At the highest concentration, GTE inhibited IL-1beta-induced IL-8 secretion to a similar extent as found for brefeldin A, an inhibitor of vesicular transport. These results suggest that GTE may exert an anti-inflammatory activity in enterocytes, which may be useful for the treatment of intestinal inflammation.


Subject(s)
Antioxidants/pharmacology , Camellia sinensis , Interleukin-8/biosynthesis , Phytotherapy , Plant Extracts/pharmacology , Antioxidants/administration & dosage , Antioxidants/therapeutic use , Caco-2 Cells/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic , Humans , Interleukin-8/genetics , Plant Extracts/administration & dosage , Plant Extracts/therapeutic use , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
9.
Gut ; 55(3): 395-402, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16150853

ABSTRACT

BACKGROUND/AIMS: The apical sodium dependent bile acid transporter ASBT (SLC10A2) contributes substantially to the enterohepatic circulation of bile acids by their reabsorption from the intestine. In the rat, its adaptive regulation was observed in the kidneys, cholangiocytes, and terminal ileum after bile duct ligation. Whether adaptive regulation of the human intestinal ASBT exists during obstructive cholestasis is not known. METHODS: Human ASBT mRNA expression along the intestinal tract was analysed by real time polymerase chain reaction in biopsies of 14 control subjects undergoing both gastroscopy and colonoscopy. Their duodenal ASBT mRNA expression was compared with 20 patients with obstructive cholestasis. Additionally, in four patients with obstructive cholestasis, duodenal ASBT mRNA expression was measured after reconstitution of bile flow. RESULTS: Normalised ASBT expression in control subjects was highest (mean arbitrary units (SEM)) in the terminal ileum (1010 (330)). Low ASBT expression was found in colonic segments (8.3 (5), 4.9 (0.9), 4.8 (1.7), and 1.1 (0.2) in the ascending, transverse, descending, and sigmoid colon, respectively). Duodenal ASBT expression in control subjects (171.8 (20.3)) was found to be approximately fourfold higher compared with patients with obstructive cholestasis (37.9 (6.5); p<0.0001). Individual ASBT mRNA expression was inversely correlated with bile acid and bilirubin plasma concentrations. In four cholestatic patients, average ASBT mRNA increased from 76 (18) before to 113 (18) after relief of cholestasis (NS). Immunohistochemical assessment indicated that ASBT protein was expressed on the apical surface of duodenal epithelial cells. CONCLUSION: Obstructive cholestasis in humans leads to downregulation of ASBT mRNA expression in the distal part of the human duodenum.


Subject(s)
Cholestasis/metabolism , Ileum/metabolism , Organic Anion Transporters, Sodium-Dependent/biosynthesis , Symporters/biosynthesis , Adaptation, Physiological , Bile Acids and Salts/blood , Bilirubin/blood , Cholestasis/blood , Cholestasis/surgery , Down-Regulation , Female , Gene Expression Regulation , Humans , Male , Organic Anion Transporters, Sodium-Dependent/genetics , PPAR alpha/biosynthesis , PPAR alpha/genetics , Polymerase Chain Reaction/methods , Postoperative Period , RNA, Messenger/genetics , Symporters/genetics
10.
Planta Med ; 71(2): 135-41, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15729621

ABSTRACT

Green tea extracts (GTE) might modulate ABC transporter gene expression or function. This may be relevant in the treatment of cancer or in influencing intestinal drug permeability. To gain more insight on the influence of a GTE on secretory transport proteins we investigated the influence of GTE and several green tea components on the mRNA expression level of P-glycoprotein (P-gp) and multidrug resistance-associated protein 2 (MRP2) in human gastrointestinal epithelial LS-180 cells. Furthermore, the functional activity of MRP2, using glutathione methylfluorescein (GS-MF) or [3H]methotrexate (MTX) as substrate, was investigated in canine kidney cells stably overexpressing human MRP2 (MDCK-MRP2). GTE, at a concentration of 0.01 mg/mL, did not increase mRNA expression of P-gp or MRP2 in LS-180 cells. Functional assays in MDCK-MRP2 cells using GS-MF did not show any effect of 0.01 mg/mL GTE on MRP2 activity. In the same cell line the cellular accumulation of MTX (a specific substrate of MRP2) was significantly increased with the MRP-specific inhibitor MK-571 or with 1 mg/mL GTE, but not with 0.1 mg/mL. The green tea components (-)-epigallocatechin gallate, (-)-epigallocatechin, theanine, or caffeine, each in corresponding concentrations to the respective concentration of GTE, did not show any effect on MRP2 function. These data demonstrate that the mRNA expression patterns of P-gp and MRP2 in LS-180 cells are not altered by 0.01 mg/mL of GTE. However, MRP2 function was inhibited by 1 mg/mL GTE, whereas none of the green tea components tested were responsible for this effect.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camellia sinensis , Membrane Transport Modulators , Membrane Transport Proteins/antagonists & inhibitors , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Phytotherapy , Plant Extracts/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Line, Tumor/drug effects , DNA Primers , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Humans , Membrane Transport Proteins/genetics , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Plant Extracts/administration & dosage , Plant Extracts/therapeutic use , Plant Leaves , RNA, Messenger/analysis
11.
Planta Med ; 68(9): 804-7, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12357391

ABSTRACT

The biflavone amentoflavone is an ingredient of Hypericum perforatum L. (Clusiaceae), a plant which is widely used for the treatment of mild to moderate depression. Amentoflavone inhibits the binding of flumazenil to the benzodiazepine binding site of the GABA A -receptor (IC(50) = 14.9 nM). Since it has to pass the blood-brain barrier (BBB) before reaching this receptor, the penetration of [(3)H]-amentoflavone through BBB was studied in an in vitro model consisting of primary cell cultures of porcine brain capillary endothelial cells (BCEC). Concentration-dependent uptake (37-2000 nM) was neither saturable nor temperature-sensitive indicating passive diffusion as the major uptake mechanism. This finding was confirmed by transport experiments through BCEC monolayers (> 2 % of applied dose was transported after 30 min). Co-administration of Hypericum extract increased amentoflavone transport significantly (amentoflavone alone: permeability coefficient P(app) = 4.59.10(-6) cm/s; co-administrated sucrose: P(app) = 3.22.10(-6)cm/s; amentoflavone together with hypericum: P(app) = 6.74.10(-6)cm/s, co-administrated sucrose P(app) = 5.49.10(-6)cm/s) indicating that Hypericum constituents enhance amentoflavone transport possibly by modulating paracellular permeability. Experiments with the P-glycoprotein (P-gp) overexpressing cell line P388-MDR showed that amentoflavone uptake was significantly enhanced by addition of the P-gp inhibitor verapamil, suggesting a P-gp mediated back-transport out of the cells. In conclusion, our findings show, that amentoflavone is able to pass the blood-brain barrier in vitro by passive diffusion.


Subject(s)
Biflavonoids , Blood-Brain Barrier/drug effects , Flavonoids/pharmacology , Hypericum , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Biological Transport/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Plant Extracts/pharmacology , Swine , Tritium/metabolism , Tumor Cells, Cultured , Verapamil/pharmacology
12.
Mol Pharmacol ; 58(6): 1357-67, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11093774

ABSTRACT

To identify specific transporters that drive xenobiotics from central nervous system to blood, the accumulation of fluorescent drugs was studied in isolated capillaries from rat and pig brain using confocal microscopy and quantitative image analysis. Luminal accumulation of daunomycin and of fluorescent derivatives of cyclosporine A (CSA) and ivermectin was concentrative, specific, and energy-dependent (inhibition by NaCN). Transport was reduced by PSC 833, ivermectin, verapamil, CSA, and vanadate, but not by leukotriene C(4) (LTC(4)), indicating the involvement of P-glycoprotein. Luminal accumulation of the fluorescent organic anions sulforhodamine 101 and fluorescein methotrexate was also concentrative, specific, and energy-dependent. LTC(4), chlorodinitrobenzene, and vanadate reduced transport of these compounds, but PSC 833 and verapamil did not, indicating the involvement of a multidrug resistance-associated protein (Mrp). Immunostaining localized P-glycoprotein and Mrp2 to the luminal surface of the capillary endothelium and quantitative polymerase chain reaction showed Mrp1 and Mrp2 expression. Finally, the HIV protease inhibitors saquinavir and ritonavir were potent inhibitors of transport mediated by both P-glycoprotein and Mrp. These results validate a new method for studying drug transport in isolated brain capillaries and implicate both P-glycoprotein and one or more members of the Mrp family in drug transport from central nervous system to blood.


Subject(s)
Brain/metabolism , Capillary Permeability , Xenobiotics/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Biological Transport , Blood-Brain Barrier , Brain/blood supply , Capillaries/metabolism , Fluorescent Antibody Technique , In Vitro Techniques , Microscopy, Confocal , Rats , Swine
13.
Br J Pharmacol ; 129(2): 251-6, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10694230

ABSTRACT

1. Transepithelial transport of a fluorescent derivative of octreotide (NBD-octreotide) was studied in freshly isolated, functionally intact renal proximal tubules from killifish (Fundulus heteroclitus). 2. Drug accumulation in the tubular lumen was visualized by means of confocal microscopy and was measured by image analysis. Secretion of NBD-octreotide into the tubular lumen was demonstrated and exhibited the all characteristics of specific and energy-dependent transport. Steady state luminal fluorescence averaged about five times cellular fluorescence and was reduced to cellular levels when metabolism was inhibited by NaCN. 3. NBD-octreotide secretion was inhibited in a concentration-dependent manner by unlabelled octreotide, verapamil and leukotriene C(4) (LTC(4)). Conversely, unlabelled octreotide reduced in a concentration dependent manner the p-glycoprotein (Pgp)-mediated secretion of a fluorescent cyclosporin A derivative (NBDL-CS) and the mrp2-mediated secretion of fluorescein methotrexate (FL-MTX). 4. This inhibition was not due to impaired metabolism or toxicity since octreotide had no influence on the active transport of fluorescein (FL), a substrate for the classical renal organic anion transport system. 5. The data are consistent with octreotide being transported across the brush border membrane of proximal kidney tubules by both Pgp and mrp2.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/physiology , ATP-Binding Cassette Transporters/physiology , Drug Resistance, Multiple/genetics , Kidney Tubules, Proximal/metabolism , Killifishes/physiology , Octreotide/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP-Binding Cassette Transporters/antagonists & inhibitors , Animals , Calcium Channel Blockers/pharmacology , Fluorescent Dyes , In Vitro Techniques , Kidney Tubules, Proximal/drug effects , Leukotriene C4/pharmacology , Methotrexate/pharmacokinetics , Microscopy, Fluorescence , Time Factors , Verapamil/pharmacology
14.
Pharm Res ; 16(10): 1570-5, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10554099

ABSTRACT

PURPOSE: The mechanism of excretion of the anthelmintic drug ivermectin was investigated in a novel experimental model of functionally intact proximal tubules isolated from a teleost fish (Fundulus heteroclitus). METHODS: Secretion into the lumens of freshly isolated proximal tubules was studied by means of confocal laser scanning microscopy and digital image analysis using ivermectin and fluorescent labelled ivermectin (BODIPY-ivermectin; BI) as substrates. RESULTS: The tubular cells rapidly accumulated BI from the medium and attained steady state within 25 minutes. Luminal fluorescence in the steady state was 5-7 times higher as compared to intracellular fluorescence. The secretion of BI into the tubular lumens was inhibited in a dose dependent manner by unlabelled ivermectin and inhibitors of the renal excretory membrane pump p-glycoprotein, namely SDZ PSC-833 and verapamil, but not by leukotriene C4, a substrate of the renal export protein mrp2. Accumulation inside the tubular cells was not affected by the added inhibitors. Ivermectin inhibited the renal secretion of the fluorescent cyclosporin derivative NBDL-CS, a substrate of p-glycoprotein, but not the secretion of the mrp2-substrate fluorescein-methotrexate, nor the secretion of fluorescein, a substrate of the classical renal organic anion transporter. CONCLUSIONS: The data are consistent with BI and ivermectin interacting in teleost kidney tubules exclusively with p-glycoprotein, but not with one of the other known excretory transport systems. In addition, the studies demonstrate that freshly isolated functionally intact kidney tubules from killifish are a useful tool to differentiate the substrate specificity of renal transport systems with respect to drug elimination.


Subject(s)
Anthelmintics/pharmacokinetics , Ivermectin/pharmacokinetics , Kidney Tubules, Proximal/metabolism , Killifishes/metabolism , Macrophage Inflammatory Proteins , ATP Binding Cassette Transporter, Subfamily B, Member 1/pharmacology , Animals , Biological Transport, Active , Calcium Channel Blockers/pharmacology , Chemokines, CC , Cyclosporins/pharmacology , Cytokines/metabolism , Epithelium/metabolism , In Vitro Techniques , Microscopy, Confocal , Microscopy, Fluorescence , Verapamil/pharmacology
15.
Mol Pharmacol ; 56(2): 383-9, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10419558

ABSTRACT

We used renal proximal tubules from a teleost fish (killifish; Fundulus heteroclitus), fluorescent substrates and confocal microscopy to study the interactions between human immunodeficiency virus protease inhibitors and drug-transporting ATPases. Both saquinavir and ritonavir inhibited luminal accumulation of a fluorescent cyclosporin A derivative (a substrate for P-glycoprotein) and of fluorescein methotrexate [a substrate for multidrug resistance-associated protein 2 (Mrp2)]. Of the two protease inhibitors, ritonavir was the more potent inhibitor of transport by a factor of at least 20. Ritonavir was at least as good an inhibitor of P-glycoprotein- and Mrp2-mediated transport as cyclosporin A and leukotriene C4, respectively. Inhibition of P-glycoprotein- and Mrp2-mediated transport was not due to toxicity or impaired metabolism, because neither saquinavir nor ritonavir inhibited transport of fluorescein on the renal organic anion system. Experiments with a fluorescent saquinavir derivative showed strong secretion into the tubular lumen that was inhibited by verapamil, leukotriene C4, saquinavir, and ritonavir. Together, the data demonstrate that saquinavir, and especially ritonavir, are potent inhibitors of P-glycoprotein- and Mrp2-mediated transport. The experiments with the fluorescent saquinavir derivative suggest that these protease inhibitors may also be substrates for both P-glycoprotein and Mrp2.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adenosine Triphosphatases/metabolism , HIV Protease Inhibitors/pharmacokinetics , Mitochondrial Proteins , Ribosomal Proteins/metabolism , Ritonavir/pharmacokinetics , Saccharomyces cerevisiae Proteins , Saquinavir/pharmacokinetics , Animals , Biological Transport , Cyclosporine/pharmacokinetics , Fluoresceins/pharmacokinetics , Killifishes , Methotrexate/analogs & derivatives , Methotrexate/pharmacokinetics
16.
Drug Metab Dispos ; 27(8): 937-41, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10421622

ABSTRACT

Multidrug resistance-associated protein (MRP) is a transport system that is involved in the elimination of xenobiotics and biologically active endogenous substrates. Recently, the presence of MRP has been demonstrated in cultured brain capillary endothelial cells (BCECs). The time-dependent, functional expression of MRP in porcine BCECs was investigated to assess the value of this cell culture model for drug transport at the blood-brain barrier. Western blot analysis was used to investigate MRP expression in freshly isolated porcine BCECs and compared to MRP expression at days 8 and 10 in culture. Subcellular localization of MRP was investigated by immunocytochemistry with an MRP-specific monoclonal antibody, MRPr1. Functional activity of MRP was assessed by efflux studies with the fluorescent MRP substrate glutathione-methylfluorescein (GS-MF). No significant MRP expression was detected in freshly isolated endothelial cells. However, MRP expression is up-regulated in cell culture in a time-dependent manner. Immunostaining revealed predominantly perinuclear and, to a lesser degree, plasma membrane localization of MRP. At 10 degrees C GS-MF efflux was significantly decreased, indicating the involvement of an energy-dependent transport system. Efflux of GS-MF was apparently inhibited by MK571, a specific inhibitor for MRP. Porcine BCECs demonstrate up-regulation of functional MRP expression during culture, as observed in human tissue, and therefore might serve as a useful in vitro system for studying MRP-mediated blood-brain barrier transport.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Brain Chemistry/physiology , Endothelium, Vascular/metabolism , ATP-Binding Cassette Transporters/antagonists & inhibitors , Animals , Blotting, Western , Bronchodilator Agents/pharmacology , Capillaries/cytology , Capillaries/metabolism , Carcinoma, Small Cell/metabolism , Endothelium, Vascular/cytology , Humans , Immunohistochemistry , In Vitro Techniques , Lung Neoplasms/metabolism , Multidrug Resistance-Associated Proteins , Propionates/pharmacology , Quinolines/pharmacology , Subcellular Fractions/metabolism , Swine , Tumor Cells, Cultured , Up-Regulation/drug effects
17.
Pharm Res ; 16(3): 402-7, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10213371

ABSTRACT

PURPOSE: Secretory systems contribute to drug absorption in the gastrointestinal tract. The purpose of this study was the identification of members of the ATP binding cassette superfamily of secretory transport proteins that may potentially modulate drug absorption in Caco-2 cells, which are an important cellular model predicting enteral absorption of drugs. METHODS: Kinetic studies as well as PCR- and Western blot studies with confluent epithelial layers of human Caco-2 cells. RESULTS: The study demonstrates functional expression of multidrug resistance related protein (MRP) and P-glycoprotein (P-gp) in Caco-2 cells: 1) Efflux studies with the MRP specific substrate glutathion-methylfluorescein (GS-MF) showed functional activity of MRP in Caco-2 cells preloaded with the metabolic precursor of GS-MF, chloromethylfluoresceine-diacetate, CMFDA. Excretion of GS-MF was decreased in presence of the MRP-blocker M K-571.2) Transport experiments with cyclosporin A demonstrated the functional activity of P-gp. Cellular accumulation was increased in presence of the P-gp blocking agent SDZ-PSC 833. 3) The expression of the 190 kDa protein MRP and the 170 kDa protein P-gp in Caco-2 cells was shown by Western blot analysis with specific monoclonal antibodies. 4) The expression of MRP-mRNA in Caco-2 cells was detected by RT-PCR and compared with the MRP over-expressing cell line H69AR. MRP primers recognize specifically human MRP1 (GenBank accession number L05628), but not all other published sequences of MRP (MRP2-MRP6). P-gp expression on mRNA-level was also confirmed by RT-PCR. CONCLUSIONS: The data demonstrate that besides P-gp, multidrug resistance related protein (MRP) is functionally expressed in Caco-2 cells and contributes to the active excretion of substrates in this cell line.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP-Binding Cassette Transporters/metabolism , ATP Binding Cassette Transporter, Subfamily B/drug effects , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/drug effects , ATP-Binding Cassette Transporters/genetics , Blotting, Western , Caco-2 Cells , Cyclosporine/pharmacokinetics , Fluoresceins/pharmacokinetics , Humans , Intestinal Absorption , Molecular Sequence Data , Multidrug Resistance-Associated Proteins , Propionates/pharmacology , Quinolines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
18.
Biochem Pharmacol ; 57(7): 833-5, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10075089

ABSTRACT

Sister of P-glycoprotein (spgp) is a gene that is closely related to the P-glycoprotein family (Pgps). This class of proteins belongs to the superfamily of ATP-binding cassette transporters and is known for its involvement in pharmacological drug interactions. Therefore, this study investigated the distribution of spgp expression in different tissues known for their high levels of Pgps expression such as brain, liver, kidney, small- and large-gut mucosa. Analysis was done by using the reverse transcription-polymerase chain reaction. In addition to a high expression in the liver, we were able to demonstrate a significant spgp expression in brain grey cortex, small- and large-gut mucosa. Although Pgps are expressed in the kidney and brain capillary endothelial cells, no expression of spgp was detected in these tissues, which might indicate that spgp has no function in the blood-brain barrier and is not involved in the renal excretion of drugs.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Animals , Base Sequence , Brain/metabolism , DNA Primers/genetics , DNA, Complementary/genetics , Endothelium, Vascular/metabolism , Gene Expression , Intestinal Mucosa/metabolism , Kidney/metabolism , Liver/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Swine , Tissue Distribution
19.
Biochem Pharmacol ; 57(10): 1147-52, 1999 May 15.
Article in English | MEDLINE | ID: mdl-11230802

ABSTRACT

The effect of P-glycoprotein inhibition on the uptake of the HIV type 1 protease inhibitor saquinavir into brain capillary endothelial cells was studied using porcine primary brain capillary endothelial cell monolayers as an in vitro test system. As confirmed by polymerase chain reaction and Western blot analysis, this system functionally expressed class I P-glycoprotein (pgp1A). P-Glycoprotein isoforms pgp1B or pgp1D could not be detected. The uptake of saquinavir into endothelial cells could be described as the result of a diffusional term of uptake and an oppositely directed saturable extrusion process. Net uptake of saquinavir into cultured brain endothelial cells could be increased significantly up to 2-fold by SDZ PSC 833 in a dose-dependent manner, with an IC(50) of 1.13 microM. In addition, the HIV protease inhibitor ritonavir inhibited p-glycoprotein-mediated extrusion of saquinavir with an IC(50) of 0.2 microM, indicating a high affinity of ritonavir for p-glycoprotein. In conclusion, we showed that the HIV protease inhibitor ritonavir is a more potent inhibitor of P-glycoprotein than the multidrug resistance (MDR)-reversing agent SDZ PSC 833. The inclusion of this drug in combination regimens may greatly facilitate brain uptake of HIV protease inhibitors, which is especially important in patients suffering from AIDS dementia complex.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Cyclosporins/pharmacology , HIV Protease Inhibitors/pharmacology , Ritonavir/pharmacology , Biological Transport/drug effects , Caco-2 Cells , Carbon Radioisotopes , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , HIV Protease Inhibitors/pharmacokinetics , Humans , Saquinavir/pharmacokinetics
20.
Drug Metab Dispos ; 26(11): 1144-8, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9806958

ABSTRACT

Peripherally administered beta-lactam antibiotics, which are structural analogs of tripeptides, may cause neurotoxic reactions or induce seizures. Previous in vivo studies provided evidence for brain uptake of these antibiotics. In the present work, we studied the extent and mechanism of the uptake of benzylpenicillin and glycylsarcosine by brain microvessel endothelial cells in vitro, using freshly isolated and cultured porcine brain capillary endothelial cells. Characterization of the cell culture model demonstrated the functional expression of the system transporting the neutral amino acids leucine and phenylalanine. The initial rate of uptake of benzylpenicillin was >3-fold greater than the rate of uptake of the extracellular marker sucrose (ratio, 3.29 +/- 0.37), whereas uptake of glycylsarcosine did not differ from that of sucrose. The differences in cellular uptake correlated with the octanol/buffer partition coefficients for glycylsarcosine and benzylpenicillin (1.16 x 10(-3) for glycylsarcosine and 6.83 x 10(-2) for benzylpenicillin). The concentration-dependent uptake of benzylpenicillin (1-2000 microM) was not saturable and was not sensitive to shifts in pH or temperature. The permeability-surface area product for the uptake of benzylpenicillin at pH 7.4 was determined from these experiments and was found to be 8.1 x 10(-5) ml/sec/g of brain. This value was very close to the value determined in in vivo studies. Uptake of benzylpenicillin and glycylsarcosine was not reduced in the presence of 1 mM ceftibuten or 100 microM probenecid. The findings with cultured cell monolayers were confirmed using freshly isolated endothelial cells. These in vitro data are compatible with benzylpenicillin, but not glycylsarcosine, being able to penetrate endothelial cells. Uptake of benzylpenicillin by brain capillary endothelial cells occurs by a slow nonsaturable process, with no evidence for carrier-mediated transport.


Subject(s)
Brain/blood supply , Capillaries/metabolism , Dipeptides/pharmacokinetics , Endothelium, Vascular/metabolism , Penicillin G/pharmacokinetics , Penicillins/pharmacokinetics , Animals , Biological Transport , Blood-Brain Barrier , Capillaries/cytology , Cells, Cultured , Endothelium, Vascular/cytology , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...