Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Eur J Pharm Sci ; 156: 105577, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33011235

ABSTRACT

The barrier properties of the brain capillary endothelium, the blood-brain barrier (BBB) restricts uptake of most small and all large molecule drug compounds to the CNS. There is a need for predictive human in vitro models of the BBB to enable studies of brain drug delivery. Here, we investigated whether human induced pluripotent stem cell (hiPSC) line (BIONi010-C) could be differentiated to brain capillary endothelial- like cells (BCEC) and evaluated their potential use in drug delivery studies. BIONi010-C hIPSCs were differentiated according to established protocols. BCEC monolayers displayed transendothelial electrical resistance (TEER) values of 5,829±354 Ω∙cm2, a Papp,mannitol of 1.09±0.15 ∙ 10-6 cm∙s-1 and a Papp,diazepam of 85.7 ± 5.9 ∙ 10-6 cm ∙s-1. The Pdiazepam/Pmannitol ratio of ~80, indicated a large dynamic passive permeability range. Monolayers maintained their integrity after medium exchange. Claudin-5, Occludin, Zonulae Occludens 1 and VE-Cadherin were expressed at the cell-cell contact zones. Efflux transporters were present at the mRNA level, but functional efflux of substrates was not detected. Transferrin-receptor (TFR), Low density lipoprotein receptor-related protein 1 (LRP1) and Basigin receptors were expressed at the mRNA-level. The presence and localization of TFR and LRP1 were verified at the protein level. A wide range of BBB-expressed solute carriers (SLC's) were detected at the mRNA level. The presence and localization of SLC transporters GLUT1 and LAT1 was verified at the protein level. Functional studies revealed transport of the LAT1 substrate [3H]-L-Leucine and the LRP1 substrate angiopep-2. In conclusion, we have demonstrated that BIONi010-C-derived BCEC monolayers exhibited, BBB properties including barrier tightness and integrity, a high dynamic range, expression of some of the BBB receptor and transporter expression, as well as functional transport of LAT1 and LRP1 substrates. This suggests that BIONi010-C-derived BCEC monolayers may be useful for studying the roles of LAT-1 and LRP1 in brain drug delivery.


Subject(s)
Blood-Brain Barrier , Induced Pluripotent Stem Cells , Biological Transport , Cell Line , Humans , Large Neutral Amino Acid-Transporter 1/genetics
2.
Fluids Barriers CNS ; 17(1): 32, 2020 Apr 22.
Article in English | MEDLINE | ID: mdl-32321539

ABSTRACT

BACKGROUND: The complexity of the neurovascular unit (NVU) poses a challenge in the investigations of drug transport across the blood-brain barrier (BBB) and the function of the brain capillary endothelium. Several in vitro models of the brain capillary endothelium have been developed. In vitro culture of primary endothelial cells has, however, been reported to alter the expression levels of various brain endothelial proteins. Only a limited number of studies have addressed this in detail. The aim of the present study was to investigate mRNA levels of selected BBB transporters and markers in in vitro models of the BBB based on bovine primary endothelial cells and compare these to the levels estimated in freshly isolated bovine brain capillaries. METHODS: Brain capillaries were isolated from bovine cerebral cortex grey matter. Capillaries were seeded in culture flasks and endothelial cells were obtained using a brief trypsinization. They were seeded onto permeable supports and cultured in mono-, non-contact- or contact co-culture with/without primary rat astrocytes. mRNA-expression levels of the selected BBB markers and transporters were evaluated using qPCR and monolayer integrity of resulting monolayers was evaluated by measuring the transendothelial electrical resistance (TEER). RESULTS: The capillary mRNA transcript profile indicated low expression of ABCC1 and CLDN1. The mRNA expression levels of TPA, OCLN, ABCB1, SLC2A1, SLC16A1 and SLC7A5 were significantly decreased in all culture configurations compared to freshly isolated bovine brain capillaries. ALP, VWF, ABCC1 and ABCC4 were upregulated during culture, while the mRNA expression levels of F11R, TJP1, CLDN5, CLDN1 and ABCG2 were found to be unaltered. The mRNA expression levels of VWF, ALP, ABCB1 and ABCC1 were affected by the presence of rat astrocytes. CONCLUSION: The endothelial mRNA transcript profile in bovine capillaries obtained in this study correlated nicely with profiles reported in mice and humans. Cultured endothelial cells drastically downregulated the mRNA expression of the investigated SLC transporters but maintained expression of efflux transporter and junctional protein mRNA, implying that the bovine in vitro BBB models may serve well to investigate basic barrier biology and in vivo permeation of passively permeating compounds and efflux transporter substrates but may be less well suited for investigations of SLC-mediated transport.


Subject(s)
Blood-Brain Barrier/metabolism , Capillaries/metabolism , Cerebral Cortex/blood supply , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Membrane Transport Proteins/metabolism , RNA, Messenger/metabolism , Animals , Cattle , Cell Culture Techniques , Cells, Cultured , Coculture Techniques , Down-Regulation , Rats , Rats, Sprague-Dawley , Solute Carrier Proteins/metabolism
3.
Mol Neurobiol ; 56(8): 5304-5314, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30603956

ABSTRACT

Endothelial dysfunction is a key element in cerebral small vessel disease (CSVD), which may cause stroke and cognitive decline. Cyclic nucleotide signaling modulates endothelial function. The cyclic adenosine monophosphate-degrading enzyme phosphodiesterase 3 (PDE3) is an important treatment target which may be modulated by microRNAs (miRNAs) important for regulating gene expression. We aimed to identify PDE3-targeting miRNAs to highlight potential therapeutic targets for endothelial dysfunction and CSVD. PDE3-targeting miRNAs were identified by in silico analysis (TargetScan, miRWalk, miRanda, and RNA22). The identified miRNAs were ranked on the basis of TargetScan context scores and their expression (log2 read counts) in a human brain endothelial cell line (hCMEC/D3) described recently. miRNAs were subjected to co-expression meta-analysis (CoMeTa) to create miRNA clusters. The pathways targeted by the miRNAs were assigned functional annotations via the KEGG pathway and COOL. hCMEC/D3 cells were transfected with miRNA mimics miR-27a-3p and miR-222-3p, and the effect on PDE3A protein expression was analyzed by Western blotting. Only PDE3A is expressed in hCMEC/D3 cells. The in silico prediction identified 67 PDE3A-related miRNAs, of which 49 were expressed in hCMEC/D3 cells. Further analysis of the top two miRNA clusters (miR-221/miR-222 and miR-27a/miR-27b/miR-128) indicated a potential link to pathways relevant to cerebral and vascular integrity and repair. hCMEC/D3 cells transfected with miR-27a-3p and miR-222-3p mimics had reduced relative expression of PDE3A protein. PDE3A-related miRNAs miR-221/miR-222 and miR-27a/miR-27b/miR-128 are potentially linked to pathways essential for immune regulation as well as cerebral and vascular integrity/function. Furthermore, relative PDE3A protein expression was reduced by miR27a-3p and miR-222-3p.


Subject(s)
Brain/blood supply , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Endothelial Cells/metabolism , MicroRNAs/metabolism , Microvessels/cytology , Cell Line , Cell Survival , DNA, Complementary/genetics , Gene Expression Regulation , Humans , MicroRNAs/genetics , Neurons/metabolism
4.
Cell Death Dis ; 5: e1483, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25341037

ABSTRACT

Sirtuins are NAD+ dependent deacetylases and/or ADP-ribosyl transferases active on histone and non-histone substrates. The first sirtuin was discovered as a transcriptional repressor of the mating-type-loci (Silent Information Regulator sir2) in the budding yeast, where it was shown to extend yeast lifespan. Seven mammalian sirtuins (SIRT1-7) have been now identified with distinct subcellular localization, enzymatic activities and substrates. These enzymes regulate cellular processes such as metabolism, cell survival, differentiation, DNA repair and they are implicated in the pathogenesis of solid tumors and leukemias. The purpose of the present study was to investigate the role of sirtuin expression, activity and inhibition in the survival of pediatric sarcoma cell lines.We have analyzed the expression of SIRT1 and SIRT2 in a series of pediatric sarcoma tumor cell lines and normal cells, and we have evaluated the activity of the sirtuin inhibitor and p53 activator tenovin-6 (Tv6) in synovial sarcoma and rhabdomyosarcoma cell lines. We show that SIRT1 is overexpressed in synovial sarcoma biopsies and cell lines in comparison with normal mesenchymal cells. Tv6 induced apoptosis as well as impaired autophagy flux. Using siRNA to knock down SIRT1 and SIRT2, we show that the expression of both proteins is crucial for the survival of rhabdomyosarcoma cells and that the loss of SIRT1 expression results in a decreased LC3II expression. Our results show that SIRT1 and SIRT2 expressions are crucial for the survival of synovial sarcomas and rhabdomyosarcomas, and demonstrate that the pharmacological inhibition of sirtuins impairs the autophagy process and induces tumor cell death.


Subject(s)
Sarcoma/pathology , Sirtuin 1/antagonists & inhibitors , Sirtuin 2/antagonists & inhibitors , Acetylation/drug effects , Animals , Autophagy/drug effects , Autophagy/genetics , Benzamides/pharmacology , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Child , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Mice, SCID , Microtubule-Associated Proteins/metabolism , Niacinamide/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , Sarcoma/genetics , Sarcoma, Synovial/genetics , Sarcoma, Synovial/pathology , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sirtuin 2/genetics , Sirtuin 2/metabolism , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
5.
Br J Pharmacol ; 170(5): 1041-52, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23962042

ABSTRACT

BACKGROUND AND PURPOSE: Intestinal nutrient transporters may mediate the uptake of drugs. The aim of this study was to investigate whether sertraline interacts with the intestinal proton-coupled amino acid transporter 1 PAT1 (SLC36A1). EXPERIMENTAL APPROACH: In vitro investigations of interactions between sertraline and human (h)PAT1, hSGLT1 (sodium-glucose linked transporter 1) and hPepT1 (proton-coupled di-/tri-peptide transporter 1) were conducted in Caco-2 cells using radiolabelled substrates. In vivo pharmacokinetic investigations were conducted in male Sprague-Dawley rats using gaboxadol (10 mg·kg(-1), p.o.) as a PAT1 substrate and sertraline (0-30.6 mg·kg(-1)). Gaboxadol was quantified by hydrophilic interaction chromatography followed by MS/MS detection. KEY RESULTS: Sertraline inhibited hPAT1-mediated L-[(3)H]-Pro uptake in Caco-2 cells. This interaction between sertraline and PAT1 appeared to be non-competitive. The uptake of the hSGLT1 substrate [(14)C]-α-methyl-D-glycopyranoside and the hPepT1 substrate [(14)C]-Gly-Sar in Caco-2 cells was also decreased in the presence of 0.3 mM sertraline. In rats, the administration of sertraline (0.1-10 mM, corresponding to 0.3-30.6 mg·kg(-1), p.o.) significantly reduced the maximal gaboxadol plasma concentration and AUC after its administration p.o. CONCLUSIONS AND IMPLICATIONS: Sertraline is an apparent non-competitive inhibitor of hPAT1-mediated transport in vitro. This inhibitory effect of sertraline is not specific to hPAT1 as substrate transport via hPepT1 and hSGLT1 was also reduced in the presence of sertraline. In vivo, sertraline reduced the amount of gaboxadol absorbed, suggesting that the inhibitory effect of sertraline on PAT1 occurs both in vitro and in vivo. Hence, sertraline could alter the bioavailability of drugs absorbed via PAT1.


Subject(s)
Amino Acid Transport Systems, Neutral/antagonists & inhibitors , Amino Acid Transport Systems/antagonists & inhibitors , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Sertraline/pharmacology , Symporters/antagonists & inhibitors , Administration, Oral , Amino Acid Transport Systems/metabolism , Amino Acid Transport Systems, Neutral/metabolism , Animals , Area Under Curve , Biological Availability , Caco-2 Cells , Chromatography/methods , Dose-Response Relationship, Drug , Humans , Hydrophobic and Hydrophilic Interactions , Intestinal Mucosa/metabolism , Isoxazoles/administration & dosage , Isoxazoles/blood , Isoxazoles/pharmacokinetics , Male , Peptide Transporter 1 , Proline/metabolism , Rats , Rats, Sprague-Dawley , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 1/metabolism , Symporters/metabolism , Tandem Mass Spectrometry , Xenopus laevis
6.
Br J Pharmacol ; 167(3): 654-65, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22577815

ABSTRACT

BACKGROUND AND PURPOSE: Intestinal absorption via membrane transporters may determine the pharmacokinetics of drug compounds. The hypothesis is that oral absorption of gaboxadol (4,5,6,7-tetrahydroisoxazolo [5,4-c] pyridine-3-ol) in rats occurs via the proton-coupled amino acid transporter, rPAT1 (encoded by the gene rSlc36a1). Consequently, we aimed to elucidate the in vivo role of rPAT1 in the absorption of gaboxadol from various intestinal segments obtained from Sprague-Dawley rats. EXPERIMENTAL APPROACH: The absorption of gaboxadol was investigated following its administration into four different intestinal segments. The intestinal expression of rSlc36a1 mRNA was measured by quantitative real-time PCR. Furthermore, the hPAT1-/rPAT1-mediated transport of gaboxadol or L-proline was studied in hPAT1-expressing Xenopus laevis oocytes, Caco-2 cell monolayers and excised segments of the rat intestine. KEY RESULTS: The absorption fraction of gaboxadol was high (81.3-91.3%) following its administration into the stomach, duodenum and jejunum, but low (4.2%) after administration into the colon. The pharmacokinetics of gaboxadol were modified by the co-administration of L-tryptophan (an hPAT1 inhibitor) and L-proline (an hPAT1 substrate). The in vitro carrier-mediated uptake rate of L-proline in the excised intestinal segments was highest in the mid jejunum and lowest in the colon. The in vitro uptake and the in vivo absorption correlated with the expression of rSlc36a1 mRNA along the rat intestine. CONCLUSIONS AND IMPLICATIONS: These results suggest that PAT1 mediates the intestinal absorption of gaboxadol and therefore determines its oral bioavailability. This has implications for the in vivo role of PAT1 and may have an influence on the design of pharmaceutical formulations of PAT1 substrates.


Subject(s)
Amino Acid Transport Systems, Neutral/metabolism , Amino Acid Transport Systems/metabolism , Gastrointestinal Tract/metabolism , Isoxazoles/pharmacokinetics , Symporters/metabolism , Administration, Oral , Amino Acid Transport Systems/genetics , Amino Acid Transport Systems, Neutral/genetics , Animals , Biological Availability , Biological Transport , Caco-2 Cells , Humans , Intestinal Absorption , Isoxazoles/administration & dosage , Male , Proline/pharmacokinetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Symporters/genetics , Tryptophan/pharmacology , Xenopus laevis
7.
Br J Pharmacol ; 161(3): 589-600, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20880398

ABSTRACT

BACKGROUND AND PURPOSE The intestinal proton-coupled amino acid transporter, SLC36A1, transports zwitterionic α-amino acids and drugs such as vigabatrin, gaboxadol and δ-aminolevulinic acid. We hypothesize that SLC36A1 might also transport some dipeptides. The aim of the present study was to investigate SLC36A1-mediated transport of Gly-Gly and Gly-Gly mimetics, and to investigate Gly-Sar transport via SLC36A1 and the proton-coupled dipeptide/tripeptide transporter, SLC15A1 in Caco-2 cells. EXPERIMENTAL APPROACH Transport of a compound via SLC36A1 was determined by its ability to induce an increase in the inward current of two-electrode voltage clamped SLC36A1 cRNA-injected Xenopus laevis oocytes. SLC36A1-mediated L-[³H]Pro uptake in Caco-2 cells was measured in the absence and presence of Gly-Gly or Gly-Sar. In addition, apical [¹4C]Gly-Sar uptake was measured in the absence and presence of the SLC36A1 inhibitor 5-hydroxy-L-tryptophan (5-HTP) or the SLC15A1 inhibitor L-4,4'-biphenylalanyl-L-proline (Bip-Pro). KEY RESULTS In SLC36A1-expressing oocytes, an inward current was induced by Gly-Sar, Gly-Gly, δ-aminolevulinic acid, ß-aminoethylglycine, δ-aminopentanoic acid, GABA, Gly and Pro, whereas Val, Leu, mannitol, 5-HTP and the dipeptides Gly-Ala, Gly-Pro and Gly-Phe did not evoke currents. In Caco-2 cell monolayers, the apical uptake of 30 mM Gly-Sar was inhibited by 20 and 22% in the presence of 5-HTP or Bip-Pro, respectively, and by 48% in the presence of both. CONCLUSION AND IMPLICATIONS Our results suggest that whereas Gly-Gly amid bond bioisosteres are widely accepted by the hPAT1 carrier, dipeptides in general are not; and therefore, Gly-Sar might structurally define the size limit of dipeptide transport via SLC36A1.


Subject(s)
Amino Acid Transport Systems/metabolism , Biological Transport/drug effects , Dipeptides/metabolism , Glycylglycine/analogs & derivatives , Glycylglycine/metabolism , Symporters/metabolism , 5-Hydroxytryptophan/pharmacology , Amino Acid Transport Systems/antagonists & inhibitors , Animals , Caco-2 Cells , Humans , Intestinal Absorption/drug effects , Oocytes/metabolism , Patch-Clamp Techniques/methods , Symporters/antagonists & inhibitors , Xenopus laevis
8.
Br J Pharmacol ; 159(6): 1339-53, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20128809

ABSTRACT

BACKGROUND AND PURPOSE: delta-Aminolevulinic acid (ALA) is used in cancer patients for photodynamic diagnosis or therapy. Oral administration of ALA has been used in patients with prostate and bladder cancer. The present aim was to investigate the mechanism of intestinal absorption of ALA and its transport via the amino acid transporter SLC36A1. EXPERIMENTAL APPROACH: In vitro investigations of ALA affinity for and uptake via SLC36A1 and SLC15A1 were performed in Caco-2 cell monolayers. Interaction of ALA with SLC15A1 was investigated in MDCK/SLC15A1 cells, whereas interactions with SLC36A1 were investigated in COS-7 cells transiently expressing SLC36A1. KEY RESULTS: ALA inhibited SLC36A1-mediated L-[(3)H]Pro and SLC15A1-mediated [(14)C]Gly-Sar uptake in Caco-2 cell monolayers with IC(50) values of 11.3 and 2.1 mM respectively. In SLC36A1-expressing COS-7 cells, the uptake of [(14)C]ALA was saturable with a K(m) value of 6.8 +/- 3.0 mM and a V(max) of 96 +/- 13 pmol x cm(-2) x min(-1). Uptake of [(14)C]ALA was pH and concentration dependent, and could be inhibited by glycine, proline and GABA. In a membrane potential assay, translocation of ALA via SLC36A1 was concentration dependent, with a K(m) value of 3.8 +/- 1.0 mM. ALA is thus a substrate for SLC36A1. In Caco-2 cells, apical [(14)C]ALA uptake was pH dependent, but Na(+) independent, and completely inhibited by 5-hydroxy-L-tryptophan and L-4,4'-biphenylalanyl-l-proline. CONCLUSIONS AND IMPLICATIONS. ALA was a substrate for SLC36A1, and the apical absorption in Caco-2 cell was only mediated by SLC36A1 and SLC15A1. This advances our understanding of intestinal absorption mechanisms of ALA, as well as its potential for drug interactions.


Subject(s)
Amino Acid Transport Systems/metabolism , Aminolevulinic Acid/pharmacology , Intestinal Mucosa/metabolism , Photosensitizing Agents/pharmacology , Symporters/metabolism , Aminolevulinic Acid/pharmacokinetics , Animals , Biological Transport , COS Cells , Caco-2 Cells , Chlorocebus aethiops , Dose-Response Relationship, Drug , Humans , Intestinal Absorption , Membrane Potentials/drug effects , Oligopeptides/pharmacokinetics , Oligopeptides/pharmacology , Peptide Transporter 1 , Photosensitizing Agents/pharmacokinetics , Substrate Specificity , Transfection
9.
Br J Pharmacol ; 157(8): 1380-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19594759

ABSTRACT

BACKGROUND AND PURPOSE: Gaboxadol has been in development for treatment of chronic pain and insomnia. The clinical use of gaboxadol has revealed that adverse effects seem related to peak serum concentrations. The aim of this study was to investigate the mechanism of intestinal absorption of gaboxadol in vitro and in vivo. EXPERIMENTAL APPROACH: In vitro transport investigations were performed in Caco-2 cell monolayers. In vivo pharmacokinetic investigations were conducted in beagle dogs. Gaboxadol doses of 2.5 mg.kg(-1) were given either as an intravenous injection (1.0 mL.kg(-1)) or as an oral solution (5.0 mL.kg(-1)). KEY RESULTS: Gaboxadol may be a substrate of the human proton-coupled amino acid transporter, hPAT1 and it inhibited the hPAT1-mediated L-[(3)H]proline uptake in Caco-2 cell monolayers with an inhibition constant K(i) of 6.6 mmol.L(-1). The transepithelial transport of gaboxadol was polarized in the apical to basolateral direction, and was dependent on gaboxadol concentration and pH of the apical buffer solution. In beagle dogs, the absorption of gaboxadol was almost complete (absolute bioavailability, F(a), of 85.3%) and T(max) was 0.46 h. Oral co-administration with 2.5-150 mg.kg(-1) of the PAT1 inhibitor, L-tryptophan, significantly decreased the absorption rate constant, k(a), and C(max), and increased T(max) of gaboxadol, whereas the area under the curve and clearance of gaboxadol were constant. CONCLUSIONS AND IMPLICATIONS: The absorption of gaboxadol across the luminal membrane of the small intestinal enterocytes is probably mediated by PAT1. This knowledge is useful for reducing gaboxadol absorption rates in order to decrease peak plasma concentrations.


Subject(s)
Amino Acid Transport Systems/antagonists & inhibitors , Amino Acid Transport Systems/metabolism , Intestinal Absorption/drug effects , Isoxazoles/pharmacokinetics , Symporters/antagonists & inhibitors , Symporters/metabolism , Tryptophan/pharmacology , Administration, Oral , Animals , Area Under Curve , Biological Availability , Caco-2 Cells , Dogs , Drug Interactions , Humans , Hydrogen-Ion Concentration , Injections, Intravenous , Isoxazoles/pharmacology , Male , Tissue Distribution
10.
J Environ Qual ; 37(6): 2202-11, 2008.
Article in English | MEDLINE | ID: mdl-18948473

ABSTRACT

Avermectins are widely used to treat livestock for parasite infections. Ivermectin, which belongs to the group of avermectins, is particularly hazardous to the environment, especially to crustaceans and to soil-dwelling organisms. Sorption is one of the key factors controlling transport and bioavailability. Therefore, batch studies have been conducted to characterize the sorption and desorption behavior of ivermectin in three European soils (Madrid, York, and artificial soil). The solid-water distribution coefficient (K(d)) for ivermectin sorption to the tested soils were between 57 and 396 L kg(-1) (determined at 0.1 microg g(-1)), while the organic carbon-normalized sorption coefficients (K(oc)) ranged from 4.00 x 10(3) to 2.58 x 10(4) L kg(-1). The Freundlich sorption coefficient (K(F)) was 396 (after 48 h) for the artificial soil over a concentration range of 0.1 to 50 microg g(-1), with regression constants indicating a concentration-dependent sorption. The obtained data and data in the literature are inconclusive with regard to whether hydrophobic partitioning or more specific interactions are involved in sorption of avermectins. For abamectin, hydrophobic partitioning seems to be one of the dominant types of binding, while hydrophobicity is less important for ivermectin, which is probably due to the lower lipophilicity of the molecule. Furthermore, the presence of cations such as Ca(2+) leads to decreasing sorption. Thus, it is presumed that ivermectin binds to soil by formation of complexes with immobile, inorganic soil matter. In contrast to abamectin, hysteresis could be excluded for ivermectin in the studied soils for the evaluation of sorption and desorption. The sorption mechanism is highly dependent on physicochemical properties of the avermectin.


Subject(s)
Antiparasitic Agents/chemistry , Ivermectin/chemistry , Soil Pollutants/chemistry , Soil/analysis , Molecular Structure
11.
Curr Drug Targets ; 4(5): 373-88, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12816347

ABSTRACT

Two human di/tri-peptide transporters, hPepT1 and hPepT2 have been identified and functionally characterized. In the small intestine hPepT1 is exclusively expressed, whereas both PepT1 and PepT2 are expressed in the proximal tubule. The transport via di/tri-peptide transporters is proton-dependent, and the transporters thus belong to the Proton-dependent Oligopeptide Transporter (POT)-family. The transporters are not drug targets per se, however due to their uniquely broad substrate specificity; they have proved to be relevant drug targets at the level of drug transport. Drug molecules such as oral active beta-lactam antibiotics, bestatin, prodrugs of aciclovir and ganciclovir have oral bioavailabilities, which largely are a result of their interaction with PepT1. In the last few years an increasing number of studies concerned with regulation of di/tri-peptide transporter capacity have appeared. Studies on receptor-mediated regulation has shown that both PepT1 and PepT2 is down-regulated by long-term exposure to epidermal growth factor (EGF) due to a decreased gene transcription. PepT1-mediated transport is up-regulated by certain substrates and in response to fasting and starvation at the level of increased gene transcription. PepT1-mediated transport is up-regulated by short-term exposure to receptor agonists such as EGF, insulin, leptin, and clonidine, and down-regulated by VIP. Overall, the regulation of di/tri-peptide transport may be contributed to 1) changes in apical proton-motive force 2) recruitment of di/tri-peptide transporters from vesicular storages 3) changes in gene transcription/mRNA stability. The aim of the present review is to discuss physiological, patho-physiological and drug-induced regulation of di/tri-peptide transporter mediated transport.


Subject(s)
Carrier Proteins/physiology , Oligopeptides/metabolism , Symporters/physiology , Animals , Biological Transport/drug effects , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Diabetes Mellitus/metabolism , Dipeptides/metabolism , Drug Delivery Systems , Enterocolitis/metabolism , Humans , Intestine, Small/metabolism , Kidney Tubules, Proximal/metabolism , Neoplasms/metabolism , Peptide Transporter 1 , Symporters/chemistry , Symporters/metabolism
12.
Acta Physiol Scand ; 178(2): 139-48, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12780388

ABSTRACT

AIMS: Little is known about the physiological regulation of the human intestinal di/tri-peptide transporter, hPepT1. In the present study we evaluated the effects of epidermal growth factor (EGF) and insulin on hPepT1-mediated dipeptide uptake in the intestinal cell line Caco-2. METHODS: Caco-2 cells were grown on filters for 23-27 days. Apical dipeptide uptake was measured using [14C]glycylsarcosine([14C]Gly-Sar). HPepT1 mRNA levels were investigated using RT-PCR, cytosolic pH was determined using the pH-sensitive fluorescent probe BCECF. RESULTS: Basolateral application of EGF increased [14C]Gly-Sar uptake with an ED50 value of 0.77 +/- 0.25 ng mL-1 (n = 3-6) and a maximal stimulation of 33 +/- 2% (n = 3-6). Insulin stimulated [14C]Gly-Sar uptake with an ED50 value of 3.5 +/- 2.0 ng mL-1 (n = 3-6) and a maximal stimulation of approximately 18% (n = 3-6). Gly-Sar uptake followed simple Michaelis-Menten kinetics. Km in control cells was 0.98 +/- 0.11 mM (n = 8) and Vmax was 1.86 +/- 0.07 nmol cm-2 min-1 (n = 8). In monolayers treated with 200 ng mL-1 of EGF, Km was 1.11 +/- 0.05 mM (n = 5) and Vmax was 2.79 +/- 0.05 nmol cm-2 min-1 (n = 5). In monolayers treated with 50 ng mL-1 insulin, Km was 1.03 +/- 0.08 mM and Vmax was 2.19 +/- 0.06 nmol cm-2 min-1 (n = 5). Kinetic data thus indicates an increase in the number of active transporters, following stimulation. The incrased Gly-Sar uptake was not accompanied by changes in hPepT1 mRNA, nor by measurable changes in cytosolic pH. CONCLUSIONS: Short-term stimulation with EGF and insulin caused an increase in hPepT1-mediated uptake of Gly-Sar in Caco-2 cell monolayers, which could not be accounted for by changes in hPepT1 mRNA or proton-motive driving force.


Subject(s)
Carrier Proteins/metabolism , Dipeptides/pharmacokinetics , Epidermal Growth Factor/pharmacology , Symporters , Brefeldin A/pharmacology , Caco-2 Cells/drug effects , Colchicine/pharmacology , Dose-Response Relationship, Drug , Glucose/pharmacokinetics , Humans , Hydrogen-Ion Concentration , Insulin/pharmacology , Leucine/pharmacokinetics , Peptide Transporter 1 , Polymerase Chain Reaction , RNA, Messenger/analysis , RNA, Neoplasm/analysis
13.
J Control Release ; 76(1-2): 129-38, 2001 Sep 11.
Article in English | MEDLINE | ID: mdl-11532319

ABSTRACT

The human intestinal di/tri-peptide carrier, hPepT1, has been suggested as a drug delivery target via increasing the intestinal transport of low permeability compounds by designing peptidomimetic prodrugs. Model ester prodrugs using the stabilized dipeptides D-Glu-Ala and D-Asp-Ala as pro-moieties for benzyl alcohol have been shown to maintain affinity for hPepT1. The primary aim of the present study was to investigate if modifications of the benzyl alcohol model drug influence the corresponding D-Glu-Ala and D-Asp-Ala model prodrugs' affinity for hPepT1 in Caco-2 cells. A second aim was to investigate the transepithelial transport and hydrolysis parameters for D-Asp(BnO)-Ala and D-Glu(BnO)-Ala across Caco-2 cell monolayers. In the present study, all investigated D-Asp-Ala and D-Glu-Ala model prodrugs retained various degrees of affinity for hPepT1 in Caco-2 cells. These affinities are used to establish a QSAR of our benzyl alcohol modified model prodrugs, aided at elucidating the observed differences in model prodrug affinity for hPepT1; additionally, these data suggest that the hydrophobicity of the side-chain model drug is the major determinant in the compounds affinity for hPepT1. Transepithelial transport studies performed using Caco-2 cells of D-Asp(BnO)-Ala and D-Glu(BnO)-Ala showed that the K(m) for transepithelial transport was not significantly different for the two compounds. The maximal transport rate of the carrier-mediated flux component does not differ between the two model prodrugs either. The transepithelial transport of D-Asp(BnO)-Ala and D-Glu(BnO)-Ala follows simple kinetics, and the release of benzyl alcohol is pH-dependent, but unaffected by 1 mM of the esterase inhibitor Paraoxon in 80% human plasma and Caco-2 cell homogenate.


Subject(s)
Carrier Proteins/metabolism , Dipeptides/metabolism , Prodrugs/metabolism , Symporters , Caco-2 Cells , Humans , Hydrogen-Ion Concentration , Peptide Transporter 1 , Quantitative Structure-Activity Relationship
14.
Acta Derm Venereol ; 81(2): 125-9, 2001 May.
Article in English | MEDLINE | ID: mdl-11501650

ABSTRACT

Conflicting results regarding the association of a polymorphism at codon 72 of the p53 tumour suppressor gene and susceptibility to develop human papilloma virus (HPV)-associated cervical cancer have been published over the last year, implicating differences in ethnic background, sample origin, sample size and/or detection assay. The material for this study was collected in the identical geographical region as for 2 previous reports with contradictory results regarding the association of codon 72 genotype with squamous cell cancer (SCC). We have used an alternative detection assay, based on pyrosequencing technology, that interrogates the variable position by the accuracy of DNA polymerase. In addition to cervical clinical specimens from SCC, HPV16- and HPV18-infected adenocarcinoma cases as well as cervical intraepithelial neoplasia (CIN) were investigated. No significant association was found between p53 codon 72 genotype and the risk to develop adenocarcinoma, SCC or CIN in the Swedish population.


Subject(s)
Adenocarcinoma/virology , Carcinoma, Squamous Cell/virology , Genes, p53/genetics , Papillomaviridae/genetics , Papillomavirus Infections/complications , Uterine Cervical Dysplasia/virology , Uterine Cervical Neoplasms/virology , Adenocarcinoma/genetics , Carcinoma, Squamous Cell/genetics , Codon/genetics , Female , Humans , Papillomavirus Infections/genetics , Polymorphism, Genetic , Sequence Analysis, DNA/methods , Uterine Cervical Neoplasms/genetics , Uterine Cervical Dysplasia/genetics
15.
Am J Physiol Gastrointest Liver Physiol ; 281(1): G191-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11408272

ABSTRACT

The human intestinal cell line Caco-2 was used as a model system to study the effects of epidermal growth factor (EGF) on peptide transport. EGF decreased apical-to-basolateral fluxes of [(14)C]glycylsarcosine ([(14)C]Gly-Sar) up to 50.2 +/- 3.6% (n = 6) of control values. Kinetic analysis of the fluxes showed that maximal flux (V(max)) of transepithelial transport decreased from 3.00 +/- 0.17 nmol x cm(-2) x min(-1) in control cells to 0.50 +/- 0.07 nmol x cm(-2) x min(-1) in cells treated with 5 ng/ml EGF (n = 6, P < 0.01). The apparent Michaelis-Menten constant (K(m)) was 2.71 +/- 0.31 mM (n = 6) in control cells and 1.89 +/- 0.28 mM (n = 6, not significantly different from control) in EGF-treated cells. Similarly, apical uptake of [(14)C]Gly-Sar decreased in cells treated with EGF, with an ED(50) value of 0.36 +/- 0.06 ng/ml (n = 6) EGF and a maximal inhibition of 80 +/- 0.02% (n = 6). V(max) decreased from 2.61 +/- 0.4 to 1.06 +/- 0.1 nmol x cm(-2) x min(-1) (n = 3, P < 0.05), whereas K(m) remained constant. Basolateral Gly-Sar uptake showed no changes in V(max) or K(m) after EGF treatment (n = 3). RT-PCR showed a decrease in hPepT1 mRNA (using glucose-6-phosphate dehydrogenase mRNA as control) in cells treated with EGF. Western blotting indicated a decrease in hPepT1 protein in cell lysates. We conclude that EGF treatment decreases Gly-Sar transport in Caco-2 cells by decreasing the number of peptide transporter molecules in the apical membrane.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Dipeptides/pharmacokinetics , Epidermal Growth Factor/pharmacology , Intestinal Mucosa/metabolism , Symporters , Biological Transport/drug effects , Biological Transport/physiology , Caco-2 Cells , Carbon Radioisotopes , Carrier Proteins/analysis , Dose-Response Relationship, Drug , Gene Expression/physiology , Humans , Immunohistochemistry , Intestinal Mucosa/cytology , Microscopy, Confocal , Peptide Transporter 1 , RNA, Messenger/analysis
16.
J Control Release ; 73(1): 21-30, 2001 May 18.
Article in English | MEDLINE | ID: mdl-11337056

ABSTRACT

The human intestinal di/tri-peptide carrier, hPepT1, has been suggested as a target for increasing intestinal transport of low permeability compounds by creating prodrugs designed for the transporter. Model ester prodrugs using the stabilized dipeptides D-Glu-Ala and D-Asp-Ala as pro-moieties for benzyl alcohol have been shown to have affinity for hPepT1. Furthermore, in aqueous solution at pH 5.5 to 10, the release of the model drug seems to be controlled by a specific base-catalyzed hydrolysis, indicating that the compounds may remain relatively stable in the upper small intestinal lumen with a pH of approximately 6.0, but still release the model drug at the intercellular and blood pH of approximately 7.4. Even though benzyl alcohol is not a low molecular weight drug molecule, these results indicate that the dipeptide prodrug principle is a promising drug delivery concept. However, the physico-chemical properties such as electronegativity, solubility, and log P of the drug molecule may also have an influence on the potential of these kinds of prodrugs. The purpose of the present study is to investigate whether the model drug electronegativity, estimated as Taft substitution parameter (sigma*) may influence the acid, water or base catalyzed model drug release rates, when released from series of D-Glu-Ala and D-Asp-Ala pro-moieties. Release rates were investigated in both aqueous solutions with varying pH, ionic strength, and buffer concentrations as well as in in vitro biological media. The release rates of all the investigated model drug molecules followed first-order kinetics and were dependent on buffer concentration, pH, ionic strength, and model drug electronegativity. The electronegativity of the model drug influenced acid, water and base catalyzed release from D-Asp-Ala and D-Glu-Ala pro-moieties. The model drug was generally released faster from D-Asp-Ala- than from the D-Glu-Ala pro-moieties. In biological media the release rate was also dependent on the electronegativity of the model drug. These results demonstrate that the model drug electronegativity, estimated as Taft (sigma*) values, has a significant influence on the release rate of the model drug.


Subject(s)
Carrier Proteins/administration & dosage , Prodrugs , Symporters , Algorithms , Animals , Buffers , Carrier Proteins/chemistry , Carrier Proteins/pharmacokinetics , Chromatography, High Pressure Liquid , Drug Carriers , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Intestines/chemistry , Kinetics , Peptide Transporter 1 , Plasma/chemistry , Solubility , Solutions , Swine
17.
Gene ; 268(1-2): 173-82, 2001 May 02.
Article in English | MEDLINE | ID: mdl-11368913

ABSTRACT

The synovial sarcoma translocation t(X;18)(p11.2; q11.2) results in the fusion of the SYT gene on chromosome 18 to exon 5 of either SSX1 or SSX2 genes on chromosome X. We recently reported that the SSX4 gene is also involved in such a translocation. In the present investigation we cloned and sequenced the full-length cDNA of SYT/SSX1, SYT/SSX2 and SYT/SSX4 from synovial sarcoma tissues. We isolated a novel fusion transcript type variant involving the fusion of SYT with exon 6 of the SSX4 gene (SYT/SSX4v). The SYT/SSX4 and SYT/SSX2 open reading frame also differed from previously reported SYT/SSX sequences by an in-frame addition of 93bp exon located in the junction between exon 7 and 8 of the SYT. This exon is identical to that reported for the murine SYT but has not been previously found in the human transcript. Two SYT transcripts, with and without the 93 bp exon, were co-expressed in mouse NIH3T3 cells, human malignant cells and human testis tissue, but not in human normal fibroblasts. Stable transfection of an SYT/SSX4 expression vector into human and murine cell lines correlated with a down-regulation of SYT transcripts. This was also observed in a synovial sarcoma tumor expressing SYT/SSX4. This suggests that the SYT/SSX fusion gene may regulate SYT expression from the normal allele and as such alter the normal function of SYT.


Subject(s)
Oncogene Proteins, Fusion/genetics , Proteins/genetics , RNA Splicing , Sarcoma, Synovial/genetics , Amino Acid Sequence , Animals , Base Sequence , Biomarkers, Tumor/genetics , Cell Line, Transformed , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cloning, Molecular , Down-Regulation , Exons , Humans , Male , Mice , Molecular Sequence Data , Oncogene Proteins, Fusion/metabolism , Proteins/metabolism , Proto-Oncogene Proteins , Repressor Proteins , Sequence Alignment , Testis/physiology , Transcription, Genetic
18.
Biochim Biophys Acta ; 1518(3): 237-48, 2001 Apr 16.
Article in English | MEDLINE | ID: mdl-11311935

ABSTRACT

Recently three orphan G-protein coupled receptors, RAIG1, GPRC5B and GPRC5C, with homology to members of family C (metabotropic glutamate receptor-like) have been identified. Using the protein sequences of these receptors as queries we identified overlapping expressed sequence tags which were predicted to encode an additional subtype. The full length coding regions of mouse mGprc5d and human GPRC5D were cloned and shown to contain predicted open reading frames of 300 and 345 amino acids, respectively. GPRC5D has seven putative transmembrane segments and is expressed in the cell membrane. The four human receptor subtypes, which we assign to group 5 of family C GPCRs, show 31-42% amino acid sequence identity to each other and 20-25% sequence identity to the transmembrane domains of metabotropic glutamate receptor subtypes 2 and 3 and other family C members. In contrast to the remaining family C members, the group 5 receptors have short amino terminal domains of some 30-50 amino acids. GPRC5D was shown to be clustered with RAIG1 on chromosome 12p13.3 and like RAIG1 and GPRC5B to consist of three exons, the first exon being the largest containing all seven transmembrane segments. GPRC5D mRNA is widely expressed in the peripheral system but all four receptors show distinct expression patterns. Interestingly, mRNA levels of all four group 5 receptors were found in medium to high levels in the kidney, pancreas and prostate and in low to medium levels in the colon and the small intestine, whereas other organs only express a subset of the genes. In an attempt to delineate the signal transduction pathway(s) of the orphan receptors, a series of chimeric receptors containing the amino terminal domain of the calcium sensing receptor or metabotropic glutamate receptor subtype 1, and the seven transmembrane domain of the orphan receptors were constructed and tested in binding and functional assays.


Subject(s)
GTP-Binding Proteins/metabolism , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled , Receptors, Metabotropic Glutamate/genetics , Recombinant Fusion Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Membrane/metabolism , Cloning, Molecular , Gene Library , Green Fluorescent Proteins , Humans , Kidney/metabolism , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Male , Mice , Molecular Sequence Data , Pancreas/metabolism , Prostate/metabolism , Quisqualic Acid , Receptors, Cell Surface/chemistry , Receptors, Metabotropic Glutamate/chemistry , Recombinant Fusion Proteins/chemistry , Sequence Alignment , Sequence Homology , Signal Transduction , Transfection , Tretinoin , Tumor Cells, Cultured
19.
J Christ Nurs ; 18(1): 10-3, 2001.
Article in English | MEDLINE | ID: mdl-11915081
20.
Cancer Res ; 60(18): 5278-83, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-11016658

ABSTRACT

We investigated the functional impact of p53 on insulin-like growth factor I receptor (IGF-IR) expression in malignant cells. Using the BL-41tsp53-2 cell line, a transfectant carrying temperature-sensitive (ts) p53 and endogenous mutant p53 (codon 248), we demonstrated a drastic down-regulation of plasma membrane-bound IGF-IRs on induction of wild-type p53. However, a similar response was obtained by treatment of BL-41tsp53-2 cells expressing mutant ts p53 with a p53 antisense oligonucleotide. Thus, even if the negative effect of wild-type p53 predominates under a competitive condition, these data indicate that mutant p53 may be important for up-regulation of IGF-IR. To further elucidate this issue, three melanoma cell lines (BE, SK-MEL-5, and SK-MEL-28) that overexpressed p53 were investigated. The BE cell line has a "hot spot" mutation (codon 248) and expresses only codon 248-mutant p53. SK-MEL-28 has a point mutation at codon 145. SK-MEL-5 cells did not exhibit any p53 mutations, but the absence of p21Waf1 expression suggested functionally aberrant p53. Our data suggest that interaction with Mdm-2 may underlie p53 inactivation in these cells. Using p53 antisense oligonucleotides, we demonstrated a substantial down-regulation of cell surface expression of IGF-IR proteins in all melanoma cell lines after 24 h. This was paralleled by decreased tyrosine phosphorylation of IGF-IR and growth arrest, and, subsequently, massive cell death was observed (this was also seen in BL-41tsp53-2 cells with mutant conformation of ts p53). Taken together, our results suggest that up-regulation of IGF-IR as a result of expression of aberrant p53 may be important for the growth and survival of malignant cells.


Subject(s)
Acetylcysteine/analogs & derivatives , Receptor, IGF Type 1/biosynthesis , Tumor Suppressor Protein p53/physiology , Acetylcysteine/pharmacology , Cell Division/drug effects , Cell Survival/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Humans , Melanoma/metabolism , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...