51
|
Kikuchi A, Tomoyasu T, Tanaka M, Kanamitsu K, Sasabe H, Maeda T, Odomi M, Tamai I. Peptide Derivation of Poorly Absorbable Drug Allows Intestinal Absorption Via Peptide Transporter. J Pharm Sci 2009; 98:1775-87. [DOI: 10.1002/jps.21551] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
52
|
Foster DR, Yee S, Bleske BE, Carver PL, Shea MJ, Menon SS, Ramachandran C, Welage LS, Amidon GL. Lack of interaction between the peptidomimetic substrates captopril and cephradine. J Clin Pharmacol 2009; 49:360-7. [PMID: 19246733 DOI: 10.1177/0091270008329554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Intestinal peptide transporters, including hPEPT1, facilitate the absorption of cephalosporins and angiotensin-converting enzyme inhibitors, and have been investigated as a means to improve oral drug absorption. Renal peptide transporters including hPEPT2, may also facilitate renal reabsorption of such compounds. In vitro and animal studies suggest that co-administration of peptidomimetic compounds may alter oral pharmacokinetics, although this has not been well studied in humans. The purpose of this study was to determine whether co-administration of the hPEPT substrates captopril and cephradine alters the oral pharmacokinetics of either agent. Nine healthy male volunteers received a single oral 25-mg dose of captopril, a single oral 500-mg dose of cephradine, or concurrent ingestion of captopril and cephradine in a cross-over manner. Venous blood samples were taken and captopril and cephradine pharmacokinetics were determined using noncompartmental analyses. No significant differences were observed in captopril or cephradine pharmacokinetics when administered together as compared to each agent alone (a marginal decrease in C(max) was observed for both captopril and cephradine during co-administration [5-15%]; however, differences were not statistically significant). The results of our study suggest that hPEPT1 and hPEPT2 are unlikely to contribute to clinically important drug interactions in humans.
Collapse
Affiliation(s)
- David R Foster
- Department of Pharmaceutical Sciences, Purde University School of Pharmacy and Pharmaceutical Sciences, Indianapolish, IN, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Pedretti A, De Luca L, Marconi C, Negrisoli G, Aldini G, Vistoli G. Modeling of the intestinal peptide transporter hPepT1 and analysis of its transport capacities by docking and pharmacophore mapping. ChemMedChem 2009; 3:1913-21. [PMID: 18979492 DOI: 10.1002/cmdc.200800184] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
An early pharmacokinetic screen for peptidomimetic drugs should have the ability to predict molecules with high affinity for intestinal transporters, as peptide-like derivatives are seldom absorbed passively. Hence, the first objective of this study was to generate a reliable model for the structure of the hPepT1 protein, which is the main intestinal transporter involved in the absorption of both dietary peptides and peptidomimetics. The modeling was based on the resolved structure of the homologous bacterial lactose permease LacY using a fragmental strategy. The interaction capacities of the hPepT1 model were explored by docking a set of 50 known ligands. Despite the known predilection of hPepT1 for hydrophobic ligands, docking results unveiled the key role of the polar interactions stabilized by charged termini, especially concerning the ammonium head group. The docking results were further verified by developing a pharmacophore model that confirmed the key features required for optimal hPepT1 affinity. The consistency of the docking results and the agreement with the pharmacophore model afford an encouraging validation for the proposed model and suggest that it can be exploited to design peptide-like molecules with an improved affinity for such a transporter.
Collapse
Affiliation(s)
- Alessandro Pedretti
- Istituto di Chimica Farmaceutica e Tossicologica "Pietro Pratesi", Facoltà di Farmacia, Università degli Studi di Milano, Via Mangiagalli 25, 20133 Milano, Italy
| | | | | | | | | | | |
Collapse
|
54
|
Miguel M, Dávalos A, Manso MA, de la Peña G, Lasunción MA, López-Fandiño R. Transepithelial transport across Caco-2 cell monolayers of antihypertensive egg-derived peptides. PepT1-mediated flux of Tyr-Pro-Ile. Mol Nutr Food Res 2008; 52:1507-13. [DOI: 10.1002/mnfr.200700503] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
55
|
Frazier S, Ajiboye K, Olds A, Wyatt T, Luetkemeier ES, Wong EA. Functional Characterization of the Chicken Peptide Transporter 1 (Pept1, Slc15a1) Gene. Anim Biotechnol 2008; 19:201-10. [DOI: 10.1080/10495390802240206] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
56
|
Otake K, Suzuki H, Higashi R, Yabuuchi H, Haga M, Maeda T, Cook TJ, Tamai I. Improved Intestinal Membrane Permeability of Hexose-Quinoline Derivatives via the Hexose Transporter, SGLT1. J Pharm Sci 2008; 97:1821-30. [PMID: 17828732 DOI: 10.1002/jps.21147] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Intestinal membrane permeability is an important factor affecting the bioavailability of drugs. As a strategy to improve membrane permeability, membrane transporters are useful targets since essential nutrients are absorbed efficiently via specific transporters. For example, there are reports that intestinal hexose transporters could be used as a tool to improve permeability; however, there has been no direct evidence that the transporter protein, sodium/glucose cotransporter 1 (SGLT1), is involved in the transport of hexose analogs. Accordingly, we examined directly whether the intestinal membrane permeability of hexose analogs can be improved by utilizing SGLT1. Three hexose-quinoline derivatives were synthesized and their interactions with SGLT1 were evaluated. Among the three derivatives, the glucose-quinoline molecule exhibited an inhibitory effect on D-glucose uptake by both rat intestinal brush-border membrane vesicles (BBMVs) and Xenopus oocytes expressing SGLT1. In addition, significant uptake of the glucose-quinoline derivative by Xenopus oocytes expressing SGLT1 was observed by both an electrophysiological assay and direct measurement of the uptake of the compound, while the galactose-quinoline derivative did not show significant uptake via SGLT1. Thus, it was directly demonstrated that SGLT1 could be used as a tool for the improvement of intestinal membrane permeability of drugs by modification to the glucose analogs.
Collapse
Affiliation(s)
- Katsumasa Otake
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | | | | | | | | | | | | | | |
Collapse
|
57
|
Gilbert ER, Wong EA, Webb KE. Board-invited review: Peptide absorption and utilization: Implications for animal nutrition and health. J Anim Sci 2008; 86:2135-55. [PMID: 18441086 DOI: 10.2527/jas.2007-0826] [Citation(s) in RCA: 218] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Over the last 50 yr, the study of intestinal peptide transport has rapidly evolved into a field with exciting nutritional and biomedical applications. In this review, we describe from a historical and current perspective intestinal peptide transport, the importance of peptides to whole-body nutrition, and the cloning and characterization of the intestinal peptide transporter, PepT1. We focus on the nutritional significance of peptide transport and relate these findings to livestock and poultry. Amino acids are transported into the enterocyte as free AA by a variety of AA transporters that vary in substrate specificity or as di- and tripeptides by the peptide transporter, PepT1. Expression of PepT1 is largely restricted to the small intestine in most species; however, in ruminants, peptide transport and activity is observed in the rumen and omasum. The extent to which peptides are absorbed and utilized is still unclear. In ruminants, peptides make a contribution to the portal-drained visceral flux of total AA and are detected in circulating plasma. Peptides can be utilized by the mammary gland for milk protein synthesis and by a variety of other tissues. We discuss the factors known to regulate expression of PepT1 including development, diet, hormones, diurnal rhythm, and disease. Expression of PepT1 is detected during embryological stages in both birds and mammals and increases with age, a strategic event that allows for the immediate uptake of nutrients after hatch or birth. Both increasing levels of protein in the diet and dietary protein deficiencies are found to upregulate the peptide transporter. We also include in this review a discussion of the use of dietary peptides and potential alternate routes of nutrient delivery to the cell. Our goal is to impart to the reader the nutritional implications of peptide transport and dietary peptides and share discoveries that shed light on various biological processes, including rapid establishment of intestinal function in early neonates and maintenance of intestinal function during fasting, starvation, and disease states.
Collapse
Affiliation(s)
- E R Gilbert
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg 24061-0306, USA
| | | | | |
Collapse
|
58
|
Li F, Maag H, Alfredson T. Prodrugs of nucleoside analogues for improved oral absorption and tissue targeting. J Pharm Sci 2008; 97:1109-34. [PMID: 17696166 DOI: 10.1002/jps.21047] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Nucleoside analogues are widely used for the treatment of antiviral infections and anticancer chemotherapy. However, many nucleoside analogues suffer from poor oral bioavailability due to their high polarity and low intestinal permeability. In order to improve oral absorption of these polar drugs, prodrugs have been employed to increase lipophilicity by chemical modification of the parent. Alternatively, prodrugs targeting transporters present in the intestine have been exploited to facilitate the transport of the nucleoside analogues. Valacyclovir and valganciclovir are two successful valine ester prodrugs transported by the PepT1 transporter. Recently, research efforts have focused on design of prodrugs for tissue specific delivery to improve efficacy and safety. This review presents advances of prodrug approaches for improved oral absorption of nucleoside analogues and recent developments in tissue targeting.
Collapse
Affiliation(s)
- Fujun Li
- Department of Pharmaceutics, Roche Palo Alto LLC, 3431 Hillview Avenue, Palo Alto, CA 94304, USA.
| | | | | |
Collapse
|
59
|
Ravna AW, Sager G, Dahl SG, Sylte I. Membrane Transporters: Structure, Function and Targets for Drug Design. TOPICS IN MEDICINAL CHEMISTRY 2008. [DOI: 10.1007/7355_2008_023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
60
|
Perkins EJ, Abraham T. Pharmacokinetics, metabolism, and excretion of the intestinal peptide transporter 1 (SLC15A1)-targeted prodrug (1S,2S,5R,6S)-2-[(2'S)-(2-amino)propionyl]aminobicyclo[3.1.0.]hexen-2,6-dicarboxylic acid (LY544344) in rats and dogs: assessment of first-pass bioactivation and dose linearity. Drug Metab Dispos 2007; 35:1903-9. [PMID: 17646281 DOI: 10.1124/dmd.107.016154] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The peptidyl prodrug (1S,2S,5R,6S)-2-[(2'S)-(2-Amino)propionyl]a-minobicyclo[3.1.0.]hexen-2,6-dicarboxylic acid, also known as LY544344, was discovered to improve the oral bioavailability of the parent drug (+)-2-aminobicyclo[3.1.0]hexane-2,6-dicarboxylic acid (LY354740), a potent group II metabotropic glutamate receptor agonist. This prodrug has been shown to deliver high plasma concentrations of the active drug via intestinal peptide transporter 1 (SLC15A1) (PepT1)-mediated intestinal transport and presystemic hydrolysis in preclinical species. The current data describe the pharmacokinetic behavior of LY544344 and LY354740, with a specific focus on the first-pass activation processes and dose linearity in rats and dogs. The PepT1 transporter makes an attractive prodrug target because of its high capacity and relatively broad substrate specificity. This was demonstrated by the wide dose proportionality observed in both species (up to 1000 mg/kg in rats and 140 mg/kg in dogs). After oral administration of LY544344, absorption and bioactivation were extensive and rapid, with greater than 97% of prodrug hydrolysis occurring before its appearance in the hepatic portal vein. Systemic activation was likewise extensive, with 100% conversion of a 7-mg/kg intravenous dose in dogs. Radiolabeled studies confirmed that hydrolysis to LY354740 was the only metabolic pathway and that the excretion pattern of the active drug was not altered by administration of the prodrug. These results demonstrate the nearly ideal prodrug properties of LY544344 and further validate the utility of the peptide transporter-directed approach to prodrug design.
Collapse
|
61
|
Biegel A, Gebauer S, Hartrodt B, Knütter I, Neubert K, Brandsch M, Thondorf I. Recognition of 2-aminothiazole-4-acetic acid derivatives by the peptide transporters PEPT1 and PEPT2. Eur J Pharm Sci 2007; 32:69-76. [PMID: 17644326 DOI: 10.1016/j.ejps.2007.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2006] [Revised: 04/10/2007] [Accepted: 06/04/2007] [Indexed: 10/23/2022]
Abstract
The H(+)/peptide cotransporters PEPT1 and PEPT2 have gained considerable interest in pharmaceutical sciences as routes for drug delivery. It is, therefore, of interest to develop uncommon artificial substrates for the two carriers. This study was initiated to investigate the binding affinity of 2-aminothiazole-4-acetic acid (ATAA) conjugates with amino acids to PEPT1 and PEPT2. The 2-aminothiazole-4-acetic acid derivatives have been synthesised and tested for their affinity to PEPT1 and PEPT2. The K(i) values were compared with in silico predicted values from CoMSIA models. C-terminal ATAA-Xaa conjugates proved to be low to medium inhibitors of the [(14)C]Gly-Sar uptake at both carrier systems whereas N-terminal Xaa-ATAA conjugates exhibited medium to high affinity. A promising candidate for further functionalisation is Val-ATAA which shows extraordinary high affinity to PEPT1.
Collapse
Affiliation(s)
- Annegret Biegel
- Institute of Biochemistry and Biotechnology, Faculty of Sciences I, Martin-Luther-University Halle-Wittenberg, D-06120 Halle, Germany
| | | | | | | | | | | | | |
Collapse
|
62
|
Thorn K, Andersen R, Christensen J, Jakobsen P, Nielsen CU, Steffansen B, Begtrup M. Design, Synthesis, and Evaluation of Tripeptidic Promoieties Targeting the Intestinal Peptide Transporter hPEPT1. ChemMedChem 2007; 2:479-87. [PMID: 17407174 DOI: 10.1002/cmdc.200600200] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The human intestinal proton coupled di/tri-peptide transporter hPEPT1 promotes the oral bioavailability of several drug compounds. The strategy behind the present work is that by linking a suitable di- or tripeptidic promoiety to a drug substance, by a hydrolysable ester bond, it may give rise to a prodrug that targets hPEPT1. 29 tripeptides were designed based on known structural requirements for substrates binding hPEPT1. Serine, homoserine, or threonine was incorporated in the tripeptide as hydroxy group donors in order for them to be linked to carboxylic drug substances. Optimisation of the promoiety included a study of 14 unnatural tripeptides whose diversity was expressed by VolSurf descriptors. A total of 29 tripeptides was synthesised by solid phase peptide synthesis and a standard Fmoc protocol. The affinity of the tripeptides to hPEPT1 was determined by measuring the inhibition of [(14)C]Gly-Sar in mature Caco-2 cell monolayers which resulted in K(i) values ranging from 0.22 to 25 mM or above. Translocation through the intestinal membrane, mediated by hPEPT1, was measured by recording the membrane potential relative to that induced by the known substrate Gly-Sar. The change in membrane potential is caused by influx of protons due to the co-transport of substrates and protons by hPEPT1 and is, as such, an indication of translocation. A K(i) value of 0.30 mM combined with efficient translocation indicated that H-Phe-Ser-Ala-OH is a suitable lead promoiety for targeted hPEPT1 prodrug design.
Collapse
Affiliation(s)
- Karina Thorn
- Department of Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | | | | | | | | | | | | |
Collapse
|
63
|
Anderson CMH, Thwaites DT. Regulation of intestinal hPepT1 (SLC15A1) activity by phosphodiesterase inhibitors is via inhibition of NHE3 (SLC9A3). BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2007; 1768:1822-9. [PMID: 17498647 PMCID: PMC2428106 DOI: 10.1016/j.bbamem.2007.04.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2007] [Revised: 03/19/2007] [Accepted: 04/06/2007] [Indexed: 11/26/2022]
Abstract
The H+-coupled transporter hPepT1 (SLC15A1) mediates the transport of di/tripeptides and many orally-active drugs across the brush-border membrane of the small intestinal epithelium. Incubation of Caco-2 cell monolayers (15 min) with the dietary phosphodiesterase inhibitors caffeine and theophylline inhibited Gly–Sar uptake across the apical membrane. Pentoxifylline, a phosphodiesterase inhibitor given orally to treat intermittent claudication, also decreased Gly–Sar uptake through a reduction in capacity (Vmax) without any effect on affinity (Km). The reduction in dipeptide transport was dependent upon both extracellular Na+ and apical pH but was not observed in the presence of the selective Na+/H+ exchanger NHE3 (SLC9A3) inhibitor S1611. Measurement of intracellular pH confirmed that caffeine was not directly inhibiting hPepT1 but rather having an indirect effect through inhibition of NHE3 activity. NHE3 maintains the H+-electrochemical gradient which, in turn, acts as the driving force for H+-coupled solute transport. Uptake of β-alanine, a substrate for the H+-coupled amino acid transporter hPAT1 (SLC36A1), was also inhibited by caffeine. The regulation of NHE3 by non-nutrient components of diet or orally-delivered drugs may alter the function of any solute carrier dependent upon the H+-electrochemical gradient and may, therefore, be a site for both nutrient–drug and drug–drug interactions in the small intestine.
Collapse
Affiliation(s)
- Catriona M H Anderson
- Epithelial Research Group, Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, University of Newcastle upon Tyne, Newcastle upon Tyne, NE2 4HH, UK.
| | | |
Collapse
|
64
|
Porter CJH, Trevaskis NL, Charman WN. Lipids and lipid-based formulations: optimizing the oral delivery of lipophilic drugs. Nat Rev Drug Discov 2007; 6:231-48. [PMID: 17330072 DOI: 10.1038/nrd2197] [Citation(s) in RCA: 1276] [Impact Index Per Article: 70.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Highly potent, but poorly water-soluble, drug candidates are common outcomes of contemporary drug discovery programmes and present a number of challenges to drug development - most notably, the issue of reduced systemic exposure after oral administration. However, it is increasingly apparent that formulations containing natural and/or synthetic lipids present a viable means for enhancing the oral bioavailability of some poorly water-soluble, highly lipophilic drugs. This Review details the mechanisms by which lipids and lipidic excipients affect the oral absorption of lipophilic drugs and provides a perspective on the possible future applications of lipid-based delivery systems. Particular emphasis has been placed on the capacity of lipids to enhance drug solubilization in the intestinal milieu, recruit intestinal lymphatic drug transport (and thereby reduce first-pass drug metabolism) and alter enterocyte-based drug transport and disposition.
Collapse
Affiliation(s)
- Christopher J H Porter
- Department of Pharmaceutics, Victorian College of Pharmacy, Monash University, Parkville campus, 381 Royal Parade, Parkville, Victoria 3052, Australia.
| | | | | |
Collapse
|
65
|
Hang CH, Shi JX, Sun BW, Li JS. Apoptosis and functional changes of dipeptide transporter (PepT1) in the rat small intestine after traumatic brain injury. J Surg Res 2006; 137:53-60. [PMID: 17081567 DOI: 10.1016/j.jss.2006.06.026] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2005] [Revised: 06/01/2006] [Accepted: 06/22/2006] [Indexed: 11/26/2022]
Abstract
BACKGROUND Traumatic brain injury (TBI) can induce significant alterations of intestinal mucosal structure and barrier function. However, it has not been investigated whether, and to what degree, apoptosis and alterations of absorptive function in the intestinal mucosal epithelium occur after TBI. MATERIAL AND METHODS Male Wistar rats were randomly divided into seven groups (five rats each group) including normal group, control group with sham operation, and TBI groups at hours 3, 12, 24, and 72, and on day 7. Parietal brain contusion was adopted using weight-dropping method. Intestinal mucosal structure was examined using histomorphmetric study and electron microscopy, and apoptosis was detected by TUNEL method. An everted sleeve of intestine was securely incubated in Kreb's solution with radioactive dipeptide ((3)H-Gly-Sar, 10 microCi/mL) to measure the uptake and transport of PepT1 of small intestinal epithelial cells. RESULTS The villous height, crypt depth and surface area were significantly decreased at 24 h after TBI, and further declined to the degree of mucosal atrophy on day 7 after TBI. Apoptotic changes of condensed nuclei in epithelial cells and fractured, distorted, and sparse microvilli were found by electron microscopy. The number of apoptotic cells in the mucosal epithelium was significantly increased since 3 h after TBI, peaked at 72 h post-injury, then declined at 7 days, but was still higher than that of control. There was a highly negative relation between the apoptotic index and the villous height, the crypt depth, and villous surface area. Compared with that of normal and control rats, the transport and uptake of dipeptide was significantly increased at 3 h post-injury (P < 0.01), peaked at 12 h and declined a bit at 24 h post-injury, and returned to the level of normal and control rats at 72 h and 7 days. CONCLUSIONS It is highly suggested that intestinal mucosa apoptosis plays an important role in the pathogenesis of acute gut damage after TBI. Intestinal PepT1 expression could be up-regulated after traumatic brain injury, and maintained the normal level under the condition of serious intestinal damage. Up-regulation of PepT1 may adaptively improve absorption of di- and tripeptides, independent of changes in the mucosal surface area.
Collapse
Affiliation(s)
- Chun-Hua Hang
- Department of Neurosurgery, Jinling Hospital, Clinical School of Medicine, Nanjing University, Nanjing, China.
| | | | | | | |
Collapse
|
66
|
Charrier L, Merlin D. The oligopeptide transporter hPepT1: gateway to the innate immune response. J Transl Med 2006; 86:538-46. [PMID: 16652110 DOI: 10.1038/labinvest.3700423] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Bacterial products that are normally present in the lumen of the colon, such as N-formylated peptides and muramyl-dipeptide, are important for inducing the development of mucosal inflammation. The intestinal dipeptide transporter, hPepT1, which is expressed in inflamed but not in noninflamed colonic epithelial cells, mediates the transport of these bacterial products into the cytosol of colonic epithelial cells. The small bacterial peptides subsequently induce an inflammatory response, including the induction of MHC class I molecules expression and cytokines secretion, via the activation of nucleotide-binding site and leucine-rich repeat (NBS-LRR) proteins, for example NOD2, and activation of NF-kappaB. Subsequent secretion of chemoattractants by colonic epithelial cells induces the movement of neutrophils through the underlying matrix, as well as across the epithelium. These bacterial products can also reach the lamina propria through the paracellular pathway and across the basolateral membrane of epithelial cells. As a consequence, small formylated peptides can interact directly with immune cells through specific membrane receptors. Since immune cells, including macrophages, also express hPepT1, they can transport small bacterial peptides into the cytosol where these may interact with the NBS-LRR family of intracellular receptors. As in intestinal epithelial cells, the presence of these small bacterial peptides in immune cells may trigger immune response activation.
Collapse
Affiliation(s)
- Laetitia Charrier
- Department of Medicine, Division of Digestive Diseases, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA
| | | |
Collapse
|
67
|
Andersen R, Jørgensen FS, Olsen L, Våbenø J, Thorn K, Nielsen CU, Steffansen B. Development of a QSAR Model for Binding of Tripeptides and Tripeptidomimetics to the Human Intestinal Di-/Tripeptide Transporter hPEPT1. Pharm Res 2006; 23:483-92. [PMID: 16489544 DOI: 10.1007/s11095-006-9462-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2005] [Accepted: 11/11/2005] [Indexed: 11/29/2022]
Abstract
PURPOSE The aim of this study was to develop a three-dimensional quantitative structure-activity relationship (QSAR) model for binding of tripeptides and tripeptidomimetics to hPEPT1 based on a series of 25 diverse tripeptides. METHODS VolSurf descriptors were generated and correlated with binding affinities by multivariate data analysis. The affinities for hPEPT1 of the tripeptides and tripeptidomimetics were determined experimentally by use of Caco-2 cell monolayers. RESULTS The Ki-values of the 25 tripeptides and tripeptidomimetics ranged from 0.15 to 25 mM and the structural diversity of the compounds was described by VolSurf descriptors. A QSAR model that correlated the VolSurf descriptors of the tripeptides with their experimental binding affinity for hPEPT1 was established. CONCLUSION Structural information on tripeptide properties influencing the binding to hPEPT1 was extracted from the QSAR model. This information may contribute to the drug design process of tripeptides and tripeptidomimetics where hPEPT1 is targeted as an absorptive transporter for improvement of intestinal absorption. To our knowledge, this is the first time a correlation between VolSurf descriptors and binding affinities for hPEPT1 has been reported.
Collapse
Affiliation(s)
- Rikke Andersen
- Molecular Biopharmaceutics, The Danish University of Pharmaceutical Sciences, 2-Universitetsparken, DK-2100, Copenhagen, Denmark.
| | | | | | | | | | | | | |
Collapse
|
68
|
Rodriguez L, Batlle A, Di Venosa G, MacRobert AJ, Battah S, Daniel H, Casas A. Study of the mechanisms of uptake of 5-aminolevulinic acid derivatives by PEPT1 and PEPT2 transporters as a tool to improve photodynamic therapy of tumours. Int J Biochem Cell Biol 2006; 38:1530-9. [PMID: 16632403 DOI: 10.1016/j.biocel.2006.03.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2006] [Revised: 03/02/2006] [Accepted: 03/03/2006] [Indexed: 11/22/2022]
Abstract
Endogenous porphyrin accumulation after administration of 5-aminolevulinic acid is employed in photodynamic therapy of tumours. Due to its low membrane permeability, esterified 5-aminolevulinic acid derivatives less hydrophilic than the parental compound are under investigation. Knowledge of the mechanisms of 5-aminolevulinic acid derivatives uptake into target cells is essential to understand and improve photodynamic therapy and useful in the design of new derivatives with better affinity and with higher selectivity for tumour cells in specific tissues. The aim of this work was to assess the interaction of 5-aminolevulinic acid derivatives with the intestinal PEPT1 and renal transporter PEPT2 expressed in Pichia pastoris yeasts. We found that Undecanoyl, Hexyl, Methyl and 2-(hydroxymethyl)tetrahydropyranyl 5-aminolevulinic acid esters and the dendron 3m-ALA inhibited (14)C-5-aminolevulinic acid uptake by PEPT2. However, only the Undecanoyl ester inhibited 5-aminolevulinic acid uptake by PEPT1. We have also found through a new developed colorimetric method, that Hexyl and 2-(hydroxymethyl)tetrahydropyranyl 5-aminolevulinic acid esters display more affinity than 5-aminolevulinic acid for PEPT2 whereas none of the compounds surpass 5-aminolevulinic acid affinity for PEPT1. In addition, the Undecanoyl ester binds with high affinity to the membranes of PEPT2 and PEPT1-expressing yeasts and to the control yeasts. The main finding of this work was that some derivatives have the potential to improve 5-aminolevulinic acid-based photodynamic therapy by increased efficiency of transport into cells expressing PEPT2 such as kidney, mammary gland, brain or lung whereas in tissues expressing exclusively PEPT1 the parent 5-aminolevulinic acid remains the compound of choice.
Collapse
Affiliation(s)
- Lorena Rodriguez
- Centro de Investigaciones sobre Porfirinas y Porfirias, CONICET and Hospital de Clínicas José de San Martín, University of Buenos Aires, Córdoba 2351 1er subsuelo, Ciudad de Buenos Aires, CP 1120AAF, Argentina
| | | | | | | | | | | | | |
Collapse
|
69
|
Takano M, Yumoto R, Murakami T. Expression and function of efflux drug transporters in the intestine. Pharmacol Ther 2006; 109:137-61. [PMID: 16209890 DOI: 10.1016/j.pharmthera.2005.06.005] [Citation(s) in RCA: 235] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2005] [Accepted: 06/21/2005] [Indexed: 02/08/2023]
Abstract
A variety of drug transporters expressed in the body control the fate of drugs by affecting absorption, distribution, and elimination processes. In the small intestine, transporters mediate the influx and efflux of endogenous or exogenous substances. In clinical pharmacotherapy, ATP-dependent efflux transporters (ATP-binding cassette [ABC] transporters) expressed on the apical membrane of the intestinal epithelial cells determine oral bioavailability, intestinal efflux clearance, and the site of drug-drug interaction of certain drugs. The expression and functional activity of efflux transporters exhibit marked interindividual variation and are relatively easily modulated by factors such as therapeutic drugs and daily foods and beverages. In this article, we will summarize the recent findings regarding the intestinal efflux transporters, especially P-glycoprotein (P-gp or human multidrug resistance gene [MDR] 1), multidrug resistance-associated protein 2 (MRP2), and breast cancer resistance protein (BCRP).
Collapse
Affiliation(s)
- Mikihisa Takano
- Department of Pharmaceutics and Therapeutics, Programs for Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan.
| | | | | |
Collapse
|
70
|
Våbenø J, Nielsen CU, Steffansen B, Lejon T, Sylte I, Jørgensen FS, Luthman K. Conformational restrictions in ligand binding to the human intestinal di-/tripeptide transporter: implications for design of hPEPT1 targeted prodrugs. Bioorg Med Chem 2005; 13:1977-88. [PMID: 15727852 DOI: 10.1016/j.bmc.2005.01.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2004] [Revised: 01/05/2005] [Accepted: 01/12/2005] [Indexed: 11/16/2022]
Abstract
The aim of the present study was to develop a computational method aiding the design of dipeptidomimetic pro-moieties targeting the human intestinal di-/tripeptide transporter hPEPT1. First, the conformation in which substrates bind to hPEPT1 (the bioactive conformation) was identified by conformational analysis and 2D dihedral driving analysis of 15 hPEPT1 substrates, which suggested that psi(1) approximately 165 degrees , omega(1) approximately 180 degrees , and phi(2) approximately 280 degrees were descriptive of the bioactive conformation. Subsequently, the conformational energy required to change the peptide backbone conformation (DeltaE(bbone)) from the global energy minimum conformation to the identified bioactive conformation was calculated for 20 hPEPT1 targeted model prodrugs with known K(i) values. Quantitatively, an inverse linear relationship (r(2)=0.81, q(2)=0.80) was obtained between DeltaE(bbone) and log1/K(i), showing that DeltaE(bbone) contributes significantly to the experimentally observed affinity for hPEPT1 ligands. Qualitatively, the results revealed that compounds classified as high affinity ligands (K(i)<0.5 mM) all have a calculated DeltaE(bbone)<1 kcal/mol, whereas medium and low-affinity compounds (0.5 mM<K(i)<15 mM) have DeltaE(bbone) values in the range 1-3 kcal/mol. The findings also shed new light on the basis for the experimentally observed stereoselectivity of hPEPT1.
Collapse
Affiliation(s)
- Jon Våbenø
- Department of Medicinal Chemistry, Institute of Pharmacy, University of Tromsø, N-9037 Tromsø, Norway
| | | | | | | | | | | | | |
Collapse
|
71
|
Hubatsch I, Lazorova L, Vahlne A, Artursson P. Orally active antiviral tripeptide glycyl-prolyl-glycinamide is activated by CD26 (dipeptidyl peptidase IV) before transport across the intestinal epithelium. Antimicrob Agents Chemother 2005; 49:1087-92. [PMID: 15728907 PMCID: PMC549227 DOI: 10.1128/aac.49.3.1087-1092.2005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The tripeptide amide glycyl-prolyl-glycinamide (GPG-amide) is a new antiretroviral drug candidate, but its absorption mechanism is unknown. In this investigation, the transport and metabolism of GPG-amide were studied in a model of the human intestinal epithelium, Caco-2 cell monolayers. The results show that when the tripeptide amide came into contact with the apical enterocyte membrane, it was degraded by CD26 (dipeptidyl peptidase IV) to glycylproline and the antiretrovirally active metabolite glycinamide. Glycinamide retained antiretroviral activity in vitro after transport through the Caco-2 cell monolayers. The transport of glycinamide across Caco-2 cell monolayers occurred via passive diffusion with an apparent permeability coefficient of about 2 x 10(-6) cm s(-1), which suggests that it is absorbed by the oral route in sufficient amounts to be considered for oral administration. In conclusion, the tripeptide GPG-amide acts as a prodrug that is activated by CD26 to release the orally active antiretroviral compound glycinamide.
Collapse
Affiliation(s)
- Ina Hubatsch
- Department of Pharmacy, Biomedical Center, Uppsala University, S-75123 Uppsala, Sweden
| | | | | | | |
Collapse
|
72
|
Katsura T, Inui KI. Intestinal absorption of drugs mediated by drug transporters: mechanisms and regulation. Drug Metab Pharmacokinet 2005; 18:1-15. [PMID: 15618714 DOI: 10.2133/dmpk.18.1] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The absorption of drugs from the gastrointestinal tract is one of the important determinants for oral bioavailability. Development of in vitro experimental techniques such as isolated membrane vesicles and cell culture systems has allowed us to elucidate the transport mechanisms of various drugs across the plasma membrane. Recent introduction of molecular biological techniques resulted in the successful identification of drug transporters responsible for the intestinal absorption of a wide variety of drugs. Each transporter exhibits its own substrate specificity, though it usually shows broad substrate specificity. In this review, we first summarize the recent advances in the characterization of drug transporters in the small intestine, classified into peptide transporters, organic cation transporters and organic anion transporters. In particular, peptide transporter (PEPT1) is the best-characterized drug transporter in the small intestine, and therefore its utilization to improve the oral absorption of poorly absorbed drugs is briefly described. In addition, regulation of the activity and expression levels of drug transporters seems to be an important aspect, because alterations in the functional characteristics and/or expression levels of drug transporters in the small intestine could be responsible for the intra- and interindividual variability of oral bioavailability of drugs. As an example, regulation of the activity and expression of PEPT1 is summarized.
Collapse
Affiliation(s)
- Toshiya Katsura
- Department of Pharmacy, Kyoto University Hospital, Faculty of Medicine, Kyoto University, Japan
| | | |
Collapse
|
73
|
Nielsen CU, Brodin B, Jørgensen FS, Frokjaer S, Steffansen B. Human peptide transporters: therapeutic applications. Expert Opin Ther Pat 2005. [DOI: 10.1517/13543776.12.9.1329] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
74
|
Klang JE, Burnworth LA, Pan YX, Webb KE, Wong EA. Functional characterization of a cloned pig intestinal peptide transporter (pPepT1). J Anim Sci 2005; 83:172-81. [PMID: 15583057 DOI: 10.2527/2005.831172x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Absorption of dietary protein can be mediated through the uptake of AA as free AA or small peptides. A H(+)-coupled, peptide transport protein, PepT1, is responsible for the absorption of small peptides arising from digestion of dietary proteins in the small intestine. The magnitude of peptide absorption and the nutritional significance of PepT1 are unknown for many food-producing animals; thus, the objective of this study was to clone and determine the functional characteristics of the pig PepT1 (pPepT1). Two cDNA-encoding pPepT1 were isolated, which contain alternative polyadenylation sites. The predicted pPepT1 is a 708-AA protein, which shows 82.8, 85.7, and 64.7% AA identity to human, sheep, and chicken PepT1, respectively. On northern blots, two pPepT1 mRNA of approximately 2.9 and 3.5 kb were detected in the duodenum, jejunum, and ileum of the small intestine and are presumed to result from alternative polyadenylation. Uptake of [(3)H]-Gly-Sar was measured in Chinese hamster ovary cells transiently transfected with a pPepT1 expression vector to study the functional expression of pPepT1. Peptide transport was H(+)-dependent, with an optimal pH of 6.0 to 6.5. The ability of pPepT1 to transport various peptides was assayed by calculating the concentration of unlabeled peptide that inhibited 50% of [(3)H]-Gly-Sar uptake (IC(50)) in transfected cells. Eleven dipeptides and two tripeptides had IC(50) values that ranged from 0.004 to 0.53 mM. Three peptides, Lys-Lys, Arg-Lys, and Lys-Trp-Lys, had IC(50) values greater than 1. 38 mM and seem to be poor substrates for pPepT1. For all three tetrapeptides examined, uptake of Gly-Sar was too small to measure, even at a concentration of 10 mM tetrapeptide; therefore, IC(50) values could not be calculated. These results demonstrate that pPepT1 can transport a variety of dipeptides and tripeptides but not tetrapeptides.
Collapse
Affiliation(s)
- J E Klang
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg 24061-0306, USA
| | | | | | | | | |
Collapse
|
75
|
Dietrich D, Hammes U, Thor K, Suter-Grotemeyer M, Flückiger R, Slusarenko AJ, Ward JM, Rentsch D. AtPTR1, a plasma membrane peptide transporter expressed during seed germination and in vascular tissue of Arabidopsis. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2004; 40:488-99. [PMID: 15500465 DOI: 10.1111/j.1365-313x.2004.02224.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
For the efficient translocation of organic nitrogen, small peptides of two to three amino acids are posited as an important alternative to amino acids. A new transporter mediating the uptake of di- and tripeptides was isolated from Arabidopsis thaliana by heterologous complementation of a peptide transport-deficient Saccharomyces cerevisiae mutant. AtPTR1 mediated growth of S. cerevisiae cells on different di- and tripeptides and caused sensitivity to the phytotoxin phaseolotoxin. The spectrum of substrates recognized by AtPTR1 was determined in Xenopus laevis oocytes injected with AtPTR1 cRNA under voltage clamp conditions. AtPTR1 not only recognized a broad spectrum of di- and tripeptides, but also substrates lacking a peptide bond. However, amino acids, omega-amino fatty acids or peptides with more than three amino acid residues did not interact with AtPTR1. At pH 5.5 AtPTR1 had an apparent lower affinity (K(0.5) = 416 microm) for Ala-Asp compared with Ala-Ala (K(0.5) = 54 microm) and Ala-Lys (K(0.5) = 112 microm). Transient expression of AtPTR1/GFP fusion proteins in tobacco protoplasts showed that AtPTR1 is localized at the plasma membrane. In addition, transgenic plants expressing the beta-glucuronidase (uidA) gene under control of the AtPTR1 promoter demonstrated expression in the vascular tissue throughout the plant, indicative of a role in long-distance transport of di- and tripeptides.
Collapse
Affiliation(s)
- Daniela Dietrich
- Molecular Plant Physiology, Institute of Plant Sciences, University of Berne, 3013 Berne, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Schmelzer CEH, Schöps R, Ulbrich-Hofmann R, Neubert RHH, Raith K. Mass spectrometric characterization of peptides derived by peptic cleavage of bovine β-casein. J Chromatogr A 2004; 1055:87-92. [PMID: 15560483 DOI: 10.1016/j.chroma.2004.09.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
This study investigated the digestion of the milk protein beta-casein with pepsin under gastro-analogous conditions. Peptide sequences were identified using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry with post-source decay as well as liquid chromatography-tandem mass spectrometry by means of database searching. The new software tool, Mascot Distiller, improved the identification rate remarkably. In the case of small peptides, such as di- and tri-peptides, which are promising candidates for intestinal absorption and possible biological effects, identification was possible only after spectrum simulation and manual matching. A list of 41 identified peptides having 2-36 amino acids is given, and unexpected cleavage sites for pepsin are reported. Sequence coverage was 75%.
Collapse
Affiliation(s)
- Christian E H Schmelzer
- Institute of Pharmaceutics and Biopharmaceutics, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Strasse 4, 06120 Halle (Saale), Germany
| | | | | | | | | |
Collapse
|
77
|
Knütter I, Hartrodt B, Theis S, Foltz M, Rastetter M, Daniel H, Neubert K, Brandsch M. Analysis of the transport properties of side chain modified dipeptides at the mammalian peptide transporter PEPT1. Eur J Pharm Sci 2004; 21:61-7. [PMID: 14706812 DOI: 10.1016/s0928-0987(03)00141-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
This study was initiated to examine systematically the effect of side chain modifications at dipeptides on their transport via PEPT1. We synthesized a series of Xaa(R)-Ala and Ala-Xaa(R) dipeptides with the functional groups of the side chains modified by structurally different blocking groups R. Recognition and transport of these derivatives by PEPT1 was measured in Caco-2 cells, in transgenic Pichia pastoris cells and in Xenopus laevis oocytes expressing PEPT1. The dipeptide derivatives displayed K(i) values between 0.002 and 4 mM. Electrophysiological analyses showed that the Ala-Xaa(R) derivatives were transported by PEPT1. In contrast, most Xaa(R)-Ala derivatives--although recognized--did not show significant transport rates. Substitution of a terminal phenyl residue in the side chain blocking group by a p-nitrophenyl residue enhanced the affinity of several dipeptide derivatives for interaction with PEPT1. However, none of these compounds showed electrogenic transport in oocytes. With a K(i) value of 0.002 mM, Lys[Z(NO(2))]-Val displayed the highest affinity to PEPT1 ever reported. We conclude that the transport of side chain modified dipeptides into enterocytes depends (a) on the position of the modified trifunctional amino acid in the dipeptide, (b) the distance between its alpha-carbon and the side chain blocking group and (c) the hydrophobic character of the side chain modification.
Collapse
Affiliation(s)
- Ilka Knütter
- Institute of Biochemistry, Department of Biochemistry/Biotechnology, and Membrane Transport Group, Biozentrum, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Strasse 3, D-06120 Halle, Germany
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Steffansen B, Nielsen CU, Brodin B, Eriksson AH, Andersen R, Frokjaer S. Intestinal solute carriers: an overview of trends and strategies for improving oral drug absorption. Eur J Pharm Sci 2004; 21:3-16. [PMID: 14706808 DOI: 10.1016/j.ejps.2003.10.010] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
A large amount of absorptive intestinal membrane transporters play an important part in absorption and distribution of several nutrients, drugs and prodrugs. The present paper gives a general overview on intestinal solute carriers as well as on trends and strategies for targeting drugs and/or prodrugs to these carriers in order to increasing oral bioavailability and distribution. A number of absorptive intestinal transporters are described in terms of gene and protein classification, driving forces, substrate specificities and cellular localization. When targeting absorptive large capacity membrane transporters in the small intestine in order to increase oral bioavailabilities of drug or prodrug, the major influence on in vivo pharmacokinetics is suggested to be dose-dependent increase in bioavailability as well as prolonged blood circulation due to large capacity facilitated absorption, and renal re-absorption, respectively. In contrast, when targeting low-capacity transporters such as vitamin transporters, dose independent saturable absorption kinetics are suggested. We thus believe that targeting drug substrates for absorptive intestinal membrane transporters could be a feasible strategy for optimizing drug bioavailability and distribution.
Collapse
Affiliation(s)
- Bente Steffansen
- Department of Pharmaceutics, The Danish University of Pharmaceutical Sciences, 2 Universitetsparken, DK-2100 Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
79
|
Neumann J, Bruch M, Gebauer S, Brandsch M. Transport of the phosphonodipeptide alafosfalin by the H+/peptide cotransporters PEPT1 and PEPT2 in intestinal and renal epithelial cells. ACTA ACUST UNITED AC 2004; 271:2012-7. [PMID: 15128310 DOI: 10.1111/j.1432-1033.2004.04114.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The interaction of the antibacterial phosphonodipeptide alafosfalin with mammalian H(+)/peptide cotransporters was studied in Caco-2 cells, expressing the low-affinity intestinal type peptide transporter 1 (PEPT1), and SKPT cells, expressing the high-affinity renal type peptide transporter 2 (PEPT2). Alafosfalin strongly inhibited the uptake of [(14)C]glycylsarcosine with K(i) values of 0.19 +/- 0.01 mm and 0.07 +/- 0.01 mm for PEPT1 and PEPT2, respectively. Saturation kinetic studies revealed that in both cell types alafosfalin affected only the affinity constant (K(t)) but not the maximal velocity (V(max)) of glycylsarcosine (Gly-Sar) uptake. The inhibition constants and the competitive nature of inhibition were confirmed in Dixon-type experiments. Caco-2 cells and SKPT cells were also cultured on permeable filters: apical uptake and transepithelial apical to basolateral flux of [(14)C]Gly-Sar across Caco-2 cell monolayers were reduced by alafosfalin (3 mm) by 73%. In SKPT cells, uptake of [(14)C]Gly-Sar but not flux was inhibited by 61%. We found no evidence for an inhibition of the basolateral to apical uptake or flux of [(14)C]Gly-Sar by alafosfalin. Alafosfalin (3 mm) did not affect the apical to basolateral [(14)C]mannitol flux. Determined in an Ussing-type experiment with Caco-2 cells cultured in Snapwells trade mark, alafosfalin increased the short-circuit current through Caco-2 cell monolayers. We conclude that alafosfalin interacts with both H(+)/peptide symporters and that alafosfalin is actively transported across the intestinal epithelium in a H(+)-symport, explaining its oral availability. The results also demonstrate that dipeptides where the C-terminal carboxyl group is substituted by a phosphonic function represent high-affinity substrates for mammalian H(+)/peptide cotransporters.
Collapse
Affiliation(s)
- Jana Neumann
- Membrane Transport Group, Biozentrum, Martin-Luther-University Halle-Wittenburg, Halle, Germany
| | | | | | | |
Collapse
|
80
|
Våbenø J, Lejon T, Nielsen CU, Steffansen B, Chen W, Ouyang H, Borchardt RT, Luthman K. Phe-Gly Dipeptidomimetics Designed for the Di-/Tripeptide Transporters PEPT1 and PEPT2: Synthesis and Biological Investigations. J Med Chem 2004; 47:1060-9. [PMID: 14761208 DOI: 10.1021/jm031022+] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A series of five Phe-Gly dipeptidomimetics containing different amide bond replacements have been synthesized in a facile way from the readily available unsaturated ketoester 1, and their affinities for the di-/tripeptide transporters hPEPT1 (Caco-2 cells) and rPEPT2 (SKPT cells) were tested. The compounds contained the amide bond isosteres ketomethylene (2a), (R)- and (S)-hydroxyethylidene (3a and 4a), and (R)- and (S)-hydroxyethylene (5a and 6a) to provide information on the conformational and stereochemical requirements for hPEPT1 and rPEPT2 affinity. The affinity studies showed that for rPEPT2 there is no significant difference in affinity between the ketomethylene isostere 2a and the natural substrate Phe-Gly (K(i) values of 18.8 and 14.6 microM, respectively). Also the affinities for hPEPT1 are in the same range (K(i) values of 0.40 and 0.20 mM, respectively). This corroborates earlier findings that the amide bond as such is not essential for binding to PEPTX, but the results also reveal possible differences in the binding of ketomethylene isosteres to hPEPT1 and rPEPT2. The trans-hydroxyethylidene and hydroxyethylene isosteres proved to be poor substrates for PEPTX. These results provide new information about the importance of flexibility and of the stereochemistry at the C(4)-position for this class of compounds. Furthermore, the intracellular uptake of 2a-4a in Caco-2 cells was investigated, showing a 3-fold reduction of the uptake of 2a in the presence of the competetive inhibitor Gly-Pro, indicating contribution from an active transport component. No active uptake of 3a and 4a was observed. Transepithelial transport studies also indicated active transport of 2a across Caco-2 monolayers.
Collapse
Affiliation(s)
- Jon Våbenø
- Department of Medicinal Chemistry, Institute of Pharmacy, University of Tromsø, N-9037 Tromsø, Norway
| | | | | | | | | | | | | | | |
Collapse
|
81
|
Brandsch M, Knütter I, Leibach FH. The intestinal H+/peptide symporter PEPT1: structure–affinity relationships. Eur J Pharm Sci 2004; 21:53-60. [PMID: 14706811 DOI: 10.1016/s0928-0987(03)00142-8] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Peptide transporter 1, PEPT1, of the mammalian enterocyte is presently under intense investigation in many laboratories because of its nutritional importance in the absorption of protein hydrolysis products and because more recent studies have shown that many drugs and prodrugs gain entry into the systemic circulation via PEPT1. Until the exact structural features of the substrate binding site of PEPT1 become available, for example by X-ray crystallography, determination of affinities followed by proof of actual membrane translocation will have to suffice when testing for possible new substrates for PEPT1. Affinity constants reflect the strength of their interaction with the binding site of the transporter. A review of the literature shows a wide range of affinity constants between 2 microM and 30 mM. We consider affinity constants for substrates or inhibitors of PEPT1 lower than 0.5 mM as high affinity, between 0.5 and 5.0 mM as medium affinity and above 5 mM as low affinity. Values above 15 mM we consider with great caution. In this mini-review we discuss affinities and structural determinants which affect affinities of a variety of substrates for PEPT1.
Collapse
Affiliation(s)
- Matthias Brandsch
- Membrane Transport Group, Biozentrum of Martin-Luther-University Halle-Wittenberg, Weinbergweg 22, D-06120 Halle, Germany.
| | | | | |
Collapse
|
82
|
Sun BW, Zhao XC, Wang GJ, Li N, Li JS. Changes of biological functions of dipeptide transporter (PepT1) and hormonal regulation in severe scald rats. World J Gastroenterol 2003; 9:2782-5. [PMID: 14669333 PMCID: PMC4612052 DOI: 10.3748/wjg.v9.i12.2782] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM: To determine the regulatory effects of recombinant human growth hormone (rhGH) on dipeptide transport (PepT1) in normal and severe scald rats.
METHODS: Male Sprague-Dawley rats with 30% total body surface area (TBSA)IIIdegree scald were employed as the model. In this study rhGH was used at the dose of 2 IU.kg-1d-1. An everted sleeve of intestine 4 cm long obtained from mid-jejunum was securely incubated in Kreb’s solution with radioactive dipeptide (3H-glycylsarcosine, 3H-Gly-Sar, 10 μCi/ml) at 37 °C for 15 min to measure the effects of uptake and transport of PepT1 of small intestinal epithelial cells in normal and severe scald rats.
RESULTS: Abundant blood supply to intestine and mesentery was observed in normal and scald rats administered rhGH, while less supply of blood to intestine and mesentery was observed in rats without rhGH. Compared with controls, the transport of dipeptide in normal rats with injection of rhGH was not significantly increased (P = 0.1926), while the uptake was significantly increased (P = 0.0253). The effects of transport and uptake of PepT1 in scald rats with injection of rhGH were significantly increased (P = 0.0082, 0.0391).
CONCLUSION: Blood supply to intestine and mesentery of rats was increased following injection of rhGH. The effects of uptake and transport of dipeptide transporters in small intestinal epithelial cells of rats with severe scald were markedly up-regulated by rhGH.
Collapse
Affiliation(s)
- Bing-Wei Sun
- Department of General Surgery, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu Province, China.
| | | | | | | | | |
Collapse
|
83
|
Bernardo PH, Brasch N, Chai CLL, Waring P. A novel redox mechanism for the glutathione-dependent reversible uptake of a fungal toxin in cells. J Biol Chem 2003; 278:46549-55. [PMID: 12947114 DOI: 10.1074/jbc.m304825200] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The fungal metabolite gliotoxin is characterized by an internal disulfide bridge and can exist in either disulfide or dithiol forms. Gliotoxin and other members of the epipolythiodioxopiperazine class of toxins have immunosuppressive properties and have been implicated in human and animal mycotoxicoses. The bridged disulfide moiety is thought to be generally essential for biological activity. Here we show that only the natural (oxidized) form of gliotoxin is actively concentrated in a cell line in a glutathione-dependent manner. Intracellular levels of the toxin can be up to 1500-fold greater than the applied concentration, and toxin in the cells exists almost exclusively in the reduced form. A simple model of toxin entry followed by reduction to the cell-impermeant dithiol explains active uptake, cell density dependence of EC50 values and predicts a value for the maximum concentration of toxin at limiting cell density in agreement with the experiment. Oxidation of the intracellular toxin results in rapid efflux from the cell that also occurs when glutathione levels fall following induction of apoptotic cell death by the toxin. This mechanism allows for minimal production of the toxin while enabling maximal intracellular concentration and thus maximal efficacy of killing in a competitor organism initially present at low cell density. The toxin effluxes from the apoptotic cell exclusively in the oxidized form and can further enter and kill neighboring cells, thus acting in a pseudocatalytic way.
Collapse
Affiliation(s)
- Paul H Bernardo
- Centre for the Study of Bioactive Molecules, Department of Chemistry, The Faculties, Australian National University, Canberra 0200, Australia
| | | | | | | |
Collapse
|
84
|
Nozawa T, Toyobuku H, Kobayashi D, Kuruma K, Tsuji A, Tamai I. Enhanced Intestinal Absorption of Drugs by Activation of Peptide Transporter PEPT1 Using Proton‐Releasing Polymer. J Pharm Sci 2003; 92:2208-16. [PMID: 14603506 DOI: 10.1002/jps.10491] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Utilization of carrier-mediated transport systems in the gastrointestinal tract to increase the bioavailability of drugs is of great interest. In the present study, an increased supply of the driving force for peptide transporter PEPT1 by utilizing a proton-releasing polymer, Eudragit L100-55, was employed to increase the intestinal transport activity. Intestinal absorption of zwitterionic cefadroxil and dianionic cefixime was studied in rats by using in situ ileal closed loops and by in vivo oral administration of the drugs concomitantly with Eudragit L100-55. The results showed that Eudragit L100-55 decreased the pH in the intestinal loops, and increased the disappearance of both cefadroxil and cefixime from the loops. In rats, the plasma concentration after oral administration was increased significantly by coadministration of Eudragit L100-55, whereas a proton-nonreleasing analogous polymer, Eudragit RSPO, did not have any effect. Furthermore, the increased absorption of cefixime caused by Eudragit L100-55 was blocked by simultaneous administration of cefadroxil, a PEPT1 substrate/inhibitor, in a concentration-dependent manner. These results demonstrate that improvement of intestinal absorption of peptide-mimetics via a peptide transporter is possible by optimizing the transporter activity through coadministration of a proton-releasing polymer that supplies the driving force for the transporter.
Collapse
Affiliation(s)
- Takashi Nozawa
- Faculty of Pharmaceutical Sciences, Department of Molecular Biopharmaceutics, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | | | | | | | | | | |
Collapse
|
85
|
Moreno-Vargas AJ, Jiménez-Barbero J, Robina I. Hetaryleneaminopolyols and hetarylenecarbopeptoids: a new type of glyco- and peptidomimetics. Syntheses and studies on solution conformation and dynamics. J Org Chem 2003; 68:4138-50. [PMID: 12762712 DOI: 10.1021/jo026631o] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Ready access to a new class of oligomers has been demonstrated by the synthesis of hetaryleneaminopolyols and hetarylenecarbopeptoids using 3-hydroxymethyl-5-(4-amino-4-deoxy-d-arabinotetritol-1-yl)-2-methylfuran and 5-(4-amino-4-deoxy-d-arabinotetritol-1-yl)-2-methyl-3-furoic acid as novel scaffolds. The conformational behavior of peptidomimetics 22, 23, 25, 26, and 36 have been analyzed by NMR spectroscopy and extensive molecular dynamics simulations. MD simulations using the GB/SA continuum solvent model for water and the MM3 force field provide a population distribution of conformers which satisfactorily agrees with the experimental NMR data for the torsional degrees of freedom of the molecule.
Collapse
Affiliation(s)
- Antonio J Moreno-Vargas
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 553, Spain
| | | | | |
Collapse
|
86
|
Thomsen AE, Friedrichsen GM, Sørensen AH, Andersen R, Nielsen CU, Brodin B, Begtrup M, Frokjaer S, Steffansen B. Prodrugs of purine and pyrimidine analogues for the intestinal di/tri-peptide transporter PepT1: affinity for hPepT1 in Caco-2 cells, drug release in aqueous media and in vitro metabolism. J Control Release 2003; 86:279-92. [PMID: 12526824 DOI: 10.1016/s0168-3659(02)00413-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A general drug delivery approach for increasing oral bioavailability of purine and pyrimidine analogues such as acyclovir may be to link these compounds reversibly to stabilized dipeptide pro-moieties with affinity for the human intestinal di/tri-peptide transporter, hPepT1. In the present study, novel L-Glu-Sar and D-Glu-Ala ester prodrugs of acyclovir and 1-(2-hydroxyethyl)-linked thymine were synthesized and their affinities for hPepT1 in Caco-2 cells were determined. Furthermore, the degradation of the prodrugs was investigated in various aqueous and biological media and compared to the corresponding hydrolysis of the prodrug valaciclovir. Affinity studies showed that the L-Glu-Sar prodrugs had high affinity for hPepT1 (K(i) approximately 0.2-0.3 mM), whereas the D-Glu-Ala prodrugs had poor affinity (K(i) approximately 50 mM). The pH-rate profiles of the prodrugs D-Glu[1-(2-hydroxyethyl)thymine]-Ala and L-Glu[acyclovir]-Sar showed specific base catalyzed degradation at pH above 4.5 and 5.5, respectively. This implicates that the degradation rates at pH approximately 7.4 (t(1/2) approximately 3.5 and 5.5 h) are approximately 25 times faster than at upper small intestinal pH approximately 6.0. In 10% porcine intestinal homogenate and 80% human plasma the half-lives of the L-Glu-Sar prodrugs were approximately between 45 and 90 min indicating a limited enzyme catalyzed degradation. In contrast, valaciclovir underwent extensive enzyme catalyzed hydrolysis in 10% porcine intestinal homogenate (t(1/2) approximately 1 min). In conclusion, L-Glu-Sar may potentially function as pro-moiety for purine and pyrimidine analogues, where release of parent compound primarily is controlled by a specific base catalyzed hydrolysis. Acyclovir is quantitatively released at the relevant pH 7.4, whereas the 1-(2-hydroxyethyl)-linked thymine is released instead of the parent compound thymine.
Collapse
Affiliation(s)
- Anne Engelbrecht Thomsen
- Department of Pharmaceutics, The Royal Danish School of Pharmacy, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|