Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Amino Acids ; 44(2): 373-81, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22711289

RESUMEN

Mechanism and substrate specificity of the proton-coupled amino acid transporter 2 (PAT2, SLC36A2) have been studied so far only in heterologous expression systems such as HeLa cells and Xenopus laevis oocytes. In this study, we describe the identification of the first cell line that expresses PAT2. We cultured 3T3-L1 cells for up to 2 weeks and differentiated the cells into adipocytes in supplemented media containing 2 µM rosiglitazone. During the 14 day differentiation period the uptake of the prototype PAT2 substrate L-[(3)H]proline increased ~5-fold. The macro- and microscopically apparent differentiation of 3T3-L1 cells coincided with their H(+) gradient-stimulated uptake of L-[(3)H]proline. Uptake was rapid, independent of a Na(+) gradient but stimulated by an inwardly directed H(+) gradient with maximal uptake occurring at pH 6.0. L-Proline uptake was found to be mediated by a transport system with a Michaelis constant (K(t)) of 130 ± 10 µM and a maximal transport velocity of 4.9 ± 0.2 nmol × 5 min(-1 )mg of protein(-1). Glycine, L-alanine, and L-tryptophan strongly inhibited L-proline uptake indicating that these amino acids also interact with the transport system. It is concluded that 3T3-L1 adipocytes express the H(+)-amino acid cotransport system PAT2.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Diferenciación Celular , Prolina/metabolismo , Simportadores/metabolismo , Células 3T3-L1 , Adipocitos/química , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Transporte Biológico , Cinética , Ratones , Simportadores/genética
2.
Anal Biochem ; 425(1): 88-90, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22425542

RESUMEN

In this note, we present a detailed procedure for highly effective and reproducible 3T3-L1 cell differentiation. Due to their potential to differentiate from fibroblasts to adipocytes, 3T3-L1 cells are widely used for studying adipogenesis and the biochemistry of adipocytes. However, using different kits and protocols published so far, we were not able to obtain full differentiation of the currently available American Type Culture Collection (ATCC) 3T3-L1 cell lots. Using rosiglitazone (2 µM) as an additional prodifferentiative agent, we achieved apparently complete differentiation of 3T3-L1 cells within 10 to 12 days that persisted for at least up to cell culture passage 10.


Asunto(s)
Células 3T3-L1/citología , Adipocitos/citología , Diferenciación Celular , Células 3T3-L1/metabolismo , Adipocitos/metabolismo , Animales , Fibroblastos/metabolismo , Ratones , Rosiglitazona , Tiazolidinedionas/farmacología
3.
Chembiochem ; 12(8): 1270-9, 2011 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-21538757

RESUMEN

In glycation reactions, the side chains of protein-bound nucleophilic amino acids such as lysine and arginine are post-translationally modified to a variety of derivatives also known as Maillard reaction products (MRPs). Considerable amounts of MRPs are taken up in food. Here we have studied the interactions of free and dipeptide-bound MRPs with intestinal transport systems. Free and dipeptide-bound derivatives of N(6)-(1-fructosyl)lysine (FL), N(6)-(carboxymethyl)lysine (CML), N(6)-(1-carboxyethyl)lysine (CEL), formyline, argpyrimidine, and methylglyoxal-derived hydroimidazolone 1 (MG-H1) were synthesized. The inhibition of L-[(3)H]lysine and [(14) C]glycylsarcosine uptakes was measured in Caco-2 cells which express the H(+)/peptide transporter PEPT1 and lysine transport system(s). Glycated amino acids always displayed lower affinities than their unmodified analogues towards the L-[(3)H]lysine transporter(s). In contrast, all glycated dipeptides except Ala-FL were medium- to high-affinity inhibitors of [(14)C]Gly-Sar uptake. The transepithelial flux of the derivatives across Caco-2 cell monolayers was determined. Free amino acids and intact peptides derived from CML and CEL were translocated to very small extents. Application of peptide-bound MRPs, however, led to elevation (up to 80-fold) of the net flux and intracellular accumulation of glycated amino acids, which were hydrolyzed from the dipeptides inside the cells. We conclude 1) that free MRPs are not substrates for the intestinal lysine transporter(s), and 2) that dietary MRPs are absorbed into intestinal cells in the form of dipeptides, most likely by the peptide transporter PEPT1. After hydrolysis, hydrophobic glycated amino acids such as pyrraline, formyline, maltosine, and argpyrimidine undergo basolateral efflux, most likely by simple diffusion down their concentration gradients.


Asunto(s)
Aminoácidos/química , Aminoácidos/metabolismo , Epitelio/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Aminoácidos/biosíntesis , Transporte Biológico , Células CACO-2 , Glicosilación , Humanos , Mucosa Intestinal/metabolismo , Ligandos , Espectroscopía de Resonancia Magnética , Reacción de Maillard , Modelos Moleculares , Péptidos/química , Péptidos/metabolismo
4.
Bioorg Med Chem ; 19(21): 6409-18, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21955456

RESUMEN

The proton-coupled amino acid transporter hPAT1 has recently gained much interest due to its ability to transport small drugs thereby allowing their oral administration. A three-dimensional quantitative structure-activity relationship (3D QSAR) study has been performed on its natural and synthetic substrates employing comparative molecular similarity indices analysis (CoMSIA) to investigate the structural requirements for substrates and to derive a predictive model that may be used for the design of new prodrugs. The cross-validated CoMSIA models have been derived from a training set of 40 compounds and the predictive ability of the resulting models has been evaluated against a test set of 10 compounds. Despite the relatively narrow range of binding affinities (K(i) values) reliable statistical models with good predictive power have been obtained. The best CoMSIA model in terms of a proper balance of all statistical terms and the overall contribution of individual properties has been obtained by considering steric, hydrophobic, hydrogen bond donor and acceptor descriptors (q(cv)(2)=0.683, r(2)=0.958 and r(PRED)(2)=0.666). The 3D QSAR model provides insight in the interactions between substrates and hPAT1 on the molecular level and allows the prediction of affinity constants of new compounds. A pharmacophore model has been generated from the training set by means of the MOE (molecular operating environment) program. This model has been used as a query for virtual screening to retrieve potential new substrates from the small-molecule, 'lead-like' databases of MOE. The affinities of the compounds were predicted and 11 compounds were identified as possible high-affinity substrates. Two selected compounds strongly inhibited the hPAT mediated l-[(3)H]proline uptake into Caco-2 cells constitutively expressing the transport protein.


Asunto(s)
Sistemas de Transporte de Aminoácidos/química , Sistemas de Transporte de Aminoácidos/metabolismo , Modelos Químicos , Simportadores/química , Simportadores/metabolismo , Sistemas de Transporte de Aminoácidos/antagonistas & inhibidores , Células CACO-2 , Endocitosis , Humanos , Cinética , Modelos Moleculares , Conformación Molecular , Relación Estructura-Actividad Cuantitativa , Especificidad por Sustrato , Simportadores/antagonistas & inhibidores
5.
Nano Lett ; 10(1): 219-23, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20017497

RESUMEN

The great potential for medical applications of inorganic nanoparticles in living organisms is severely restricted by the concern that nanoparticles can harmfully interact with biological systems, such as lipid membranes or cell proteins. To enable an uptake of such nanoparticles by cells without harming their membranes, platinum nanoparticles were synthesized within cavities of hollow protein nanospheres (apoferritin). In vitro, the protein-platinum nanoparticles show good catalytic efficiency and long-term stability. Subsequently the particles were tested after ferritin-receptor-mediated incorporation in human intestinal Caco-2 cells. Upon externally induced stress, for example, with hydrogen peroxide, the oxygen species in the cells decreased and the viability of the cells increased.


Asunto(s)
Apoferritinas/química , Nanopartículas del Metal/química , Nanotecnología/métodos , Platino (Metal)/química , Antioxidantes/química , Células CACO-2 , Supervivencia Celular , Células Epiteliales/citología , Humanos , Peróxido de Hidrógeno/química , Microscopía Electrónica de Transmisión/métodos , Nanopartículas/química , Nanotecnología/instrumentación , Oxígeno/química , Proteínas/química , Especies Reactivas de Oxígeno
6.
Biochim Biophys Acta ; 1778(4): 1042-50, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18230330

RESUMEN

The proton-coupled amino acid transporter 1 (PAT1) represents a major route by which small neutral amino acids are absorbed after intestinal protein digestion. The system also serves as a novel route for oral drug delivery. Having shown that H+ affects affinity constants but not maximal velocity of transport, we investigated which histidine residues are obligatory for PAT1 function. Three histidine residues are conserved among the H+-coupled amino acid transporters PAT1 to 4 from different animal species. We individually mutated each of these histidine residues and compared the catalytic function of the mutants with that of the wild type transporter after expression in HRPE cells. His-55 was found to be essential for the catalytic activity of hPAT1 because the corresponding mutants H55A, H55N and H55E had no detectable l-proline transport activity. His-93 and His-135 are less important for transport function since H93N and H135N mutations did not impair transport function. The loss of transport function of His-55 mutants was not due to alterations in protein expression as shown both by cell surface biotinylation immunoblot analyses and by confocal microscopy. We conclude that His-55 might be responsible for binding and translocation of H+ in the course of cellular amino acid uptake by PAT1.


Asunto(s)
Sistemas de Transporte de Aminoácidos/química , Sistemas de Transporte de Aminoácidos/metabolismo , Histidina/genética , Simportadores/química , Simportadores/metabolismo , Secuencia de Aminoácidos , Transporte Biológico , Células Cultivadas , Secuencia Conservada , Análisis Mutacional de ADN , ADN Complementario , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Prolina/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Especificidad por Sustrato
7.
Drug Metab Dispos ; 37(1): 143-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18824524

RESUMEN

Sartans are very effective drugs for treatment of hypertension, heart failure, and other cardiovascular disorders. They antagonize the effects of angiotensin II at the AT(1) receptor and display p.o. bioavailability rates of 13 to 80%. Because some sartans sterically resemble dipeptide derivatives, we investigated whether they are transported by peptide transporters. We first assessed the effects of sartans on [(14)C]glycylsarcosine uptake into Caco-2 cells expressing H(+)/peptide transporter (PEPT) 1 and into SKPT cells expressing PEPT2. Losartan, irbesartan, valsartan, and eprosartan inhibited [glycine-1-(14)C]glycylsarcosine ([(14)C]Gly-Sar) uptake into Caco-2 cells in a competitive manner with K(i) values of 24, 230, 390, and >1000 microM. Losartan and valsartan also strongly inhibited the total transepithelial flux of [(14)C]Gly-Sar across Caco-2 cell monolayers. In SKPT cells, [(14)C]Gly-Sar uptake was inhibited with K(i) values of 2.2 microM (losartan), 65 microM (irbesartan), 260 microM (valsartan), and 490 microM (eprosartan). We determined by the two-electrode voltage-clamp technique whether the compounds elicited transport currents by PEPT1 or PEPT2 when expressed in Xenopus laevis oocytes. No currents were observed for any of the sartans, but the compounds strongly and reversibly inhibited peptide-induced currents. Uptake of valsartan, losartan, and cefadroxil was quantified in HeLa cells after heterologous expression of human PEPT1 (hPEPT1). In contrast to cefadroxil, no PEPT1-specific uptake of valsartan and losartan was found. We conclude that the sartans tested in this study display high-affinity interaction with PEPTs but are not transported themselves. However, they strongly inhibit hPEPT1-mediated uptake of dipeptides and cefadroxil.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Simportadores/metabolismo , Animales , Células CACO-2 , Cromatografía Líquida de Alta Presión , Células HeLa , Humanos , Transportador de Péptidos 1 , Unión Proteica , Ratas , Espectrofotometría Ultravioleta , Xenopus laevis
8.
J Pharmacol Exp Ther ; 327(2): 432-41, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18713951

RESUMEN

Angiotensin-converting enzyme (ACE) inhibitors are often regarded as substrates for the H+/peptide transporters (PEPT)1 and PEPT2. Even though the conclusions drawn from published data are quite inconsistent, in most review articles PEPT1 is claimed to mediate the intestinal absorption of ACE inhibitors and thus to determine their oral availability. We systematically investigated the interaction of a series of ACE inhibitors with PEPT1 and PEPT2. First, we studied the effect of 14 ACE inhibitors including new drugs on the uptake of the dipeptide [14C]glycylsarcosine into human intestinal Caco-2 cells constitutively expressing PEPT1 and rat renal SKPT cells expressing PEPT2. In a second approach, the interaction of ACE inhibitors with heterologously expressed human PEPT1 and PEPT2 was determined. In both assay systems, zofenopril and fosinopril were found to have very high affinity for binding to peptide transporters. Medium to low affinity for transporter interaction was found for benazepril, quinapril, trandolapril, spirapril, cilazapril, ramipril, moexipril, quinaprilat, and perindopril. For enalapril, lisinopril, and captopril, very weak affinity or lack of interaction was found. Transport currents of PEPT1 and PEPT2 expressed in Xenopus laevis oocytes were recorded by the two-electrode voltage-clamp technique. Statistically significant, but very low currents were only observed for lisinopril, enalapril, quinapril, and benazepril at PEPT1 and for spirapril at PEPT2. For the other ACE inhibitors, electrogenic transport activity was extremely low or not measurable at all. The present results suggest that peptide transporters do not control intestinal absorption and renal reabsorption of ACE inhibitors.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/metabolismo , Simportadores/fisiología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Transporte Biológico , Células CACO-2 , Dipéptidos/metabolismo , Femenino , Humanos , Absorción Intestinal , Riñón/metabolismo , Transportador de Péptidos 1 , Ratas , Xenopus laevis
9.
Eur J Pharm Biopharm ; 70(2): 486-92, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18577448

RESUMEN

To reach its target cells, the antiepileptic drug valproate has to cross both the intestinal epithelial barrier and the blood-brain barrier in intact form as well as in sufficient amounts. This study was performed to characterize the epithelial transport of valproate at intestinal (Caco-2) and at blood-brain barrier (RBE4) cells. At both cell types, uptake of [(3)H]valproate was independent of inwardly directed Na(+), Ca(2+), Mg(2+), K(+) or Cl(-) gradients. Uptake was, however, strongly stimulated by an inwardly directed H(+) gradient. The cells accumulated valproate against a concentration gradient and the uptake rate of valproate was saturable with K(t) values of 0.6 and 0.8mM. At Caco-2 cell monolayers, the total apical-to-basolateral flux of [(3)H]valproate exceeded the basolateral-to-apical flux 14-fold. Various monocarboxylic acids like salicylate, benzoate, acetate, propionate, butyrate, hexanoate, diclofenac and ibuprofen inhibited [(3)H]valproate uptake at both cell types. Lactate and pyruvate inhibited valproate uptake at RBE4 cells but not at Caco-2 cells. We conclude that valproate is accumulated in intestinal cells against a concentration gradient by the activity of a specific H(+)-dependent DIDS-insensitive transport system for monocarboxylates not identical with monocarboxylate transporter 1 (MCT1). The passage of valproate across the blood-brain barrier is very likely mediated by MCT1.


Asunto(s)
Anticonvulsivantes/farmacocinética , Barrera Hematoencefálica , Encéfalo/irrigación sanguínea , Células Endoteliales/metabolismo , Ácido Valproico/farmacocinética , Transporte Biológico , Células CACO-2 , Humanos , Concentración de Iones de Hidrógeno , Transportadores de Ácidos Monocarboxílicos/fisiología , Especificidad por Sustrato , Simportadores/fisiología
10.
J Pharm Pharmacol ; 60(5): 543-85, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18416933

RESUMEN

Peptide transport is currently a prominent topic in membrane research. The transport proteins involved are under intense investigation because of their physiological importance in protein absorption and also because peptide transporters are possible vehicles for drug delivery. Moreover, in many tissues peptide carriers transduce peptidic signals across membranes that are relevant in information processing. The focus of this review is on the pharmaceutical relevance of the human peptide transporters PEPT1 and PEPT2. In addition to their physiological substrates, both carriers transport many beta-lactam antibiotics, valaciclovir and other drugs and prodrugs because of their sterical resemblance to di- and tripeptides. The primary structure, tissue distribution and substrate specificity of PEPT1 and PEPT2 have been well characterized. However, there is a dearth of knowledge on the substrate binding sites and the three-dimensional structure of these proteins. Until this pivotal information becomes available by X-ray crystallography, the development of new drug substrates relies on classical transport studies combined with molecular modelling. In more than thirty years of research, data on the interaction of well over 700 di- and tripeptides, amino acid and peptide derivatives, drugs and prodrugs with peptide transporters have been gathered. The aim of this review is to put the reports on peptide transporter-mediated drug uptake into perspective. We also review the current knowledge on pharmacogenomics and clinical relevance of human peptide transporters. Finally, the reader's attention is drawn to other known or proposed human peptide-transporting proteins.


Asunto(s)
Proteínas de Transporte de Membrana/metabolismo , Preparaciones Farmacéuticas/metabolismo , Animales , Animales Modificados Genéticamente , Humanos , Mucosa Intestinal/metabolismo , Riñón/metabolismo , Proteínas de Transporte de Membrana/genética , Farmacogenética , Polimorfismo Genético , Profármacos/metabolismo , Especificidad por Sustrato , Distribución Tisular
11.
FEBS J ; 274(22): 5905-14, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17944948

RESUMEN

In this study we described the design, rational synthesis and functional characterization of a novel radiolabeled hydrolysis-resistant high-affinity substrate for H(+)/peptide cotransporters. L-4,4'-Biphenylalanyl-L-Proline (Bip-Pro) was synthesized according to standard procedures in peptide chemistry. The interaction of Bip-Pro with H(+)/peptide cotransporters was determined in intestinal Caco-2 cells constitutively expressing human H(+)/peptide cotransporter 1 (PEPT1) and in renal SKPT cells constitutively expressing rat H(+)/peptide cotransporter 2 (PEPT2). Bip-Pro inhibited the [(14)C]Gly-Sar uptake via PEPT1 and PEPT2 with exceptional high affinity (K(i) = 24 microm and 3.4 microm, respectively) in a competitive manner. By employing the two-electrode voltage clamp technique in Xenopus laevis oocytes expressing PEPT1 or PEPT2 it was found that Bip-Pro was transported by both peptide transporters although to a much lower extent than the reference substrate, Gly-Gln. Bip-Pro remained intact to > 98% for at least 8 h when incubated with intact cell monolayers. Bip-[(3)H]Pro uptake into SKPT cells was linear for up to 30 min and pH dependent with a maximum at extracellular pH 6.0. Uptake was strongly inhibited, not only by unlabeled Bip-Pro but also by known peptide transporter substrates such as dipeptides, cefadroxil, Ala-4-nitroanilide and delta-aminolevulinic acid, but not by glycine. Bip-Pro uptake in SKPT cells was saturable with a Michaelis-Menten constant (K(t)) of 7.6 microm and a maximal velocity (V(max)) of 1.1 nmol x 30 min(-1) x mg of protein(-1). Hence, the uptake of Bip-Pro by PEPT2 is a high-affinity, low-capacity process in comparison to the uptake of Gly-Sar. We conclude that Bip-[(3)H]Pro is a valuable substrate for both mechanistic and structural studies of H(+)/peptide transporter proteins.


Asunto(s)
Proteínas Portadoras/metabolismo , Hidrógeno/metabolismo , Péptidos/metabolismo , Radioisótopos , Animales , Línea Celular , Cromatografía Líquida de Alta Presión , Femenino , Cinética , Técnicas de Placa-Clamp , Ratas , Xenopus laevis
12.
Eur J Pharm Sci ; 32(4-5): 291-5, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17869491

RESUMEN

After transport across several epithelial barriers including the blood-brain barrier, clonidine interacts with alpha(2)-adrenergic receptors and imidazoline binding sites in the brain. We hypothesized that neuronal cells take up clonidine thereby removing the drug from the extracellular fluid compartment. Uptake of [(3)H]clonidine into SH-SY5Y neuroblastoma cells was linear for up to 1 min, unaffected by inside directed Na(+) or Cl(-) gradients but strongly inhibited by an outside pH of 6.0. The cells accumulated [(3)H]clonidine 50-70-fold uphill against a concentration gradient. Unlabeled clonidine, guanabenz, imipramine, diphenhydramine, maprotiline, quinine and the endogenous monoamine phenylethylamine (2 mM) strongly inhibited the [(3)H]clonidine uptake by 60-95%. Tetraethylammonium, choline and N-methyl-4-phenylpyridinium had no effect. The accumulation at pH 7.5 was saturable with an apparent Michaelis-Menten constant (K(t)) of 0.7 mM. We conclude that SH-SY5Y cells not only bind clonidine to extracellular receptors but also take up the drug rapidly by a specific and concentrative mechanism.


Asunto(s)
Clonidina/metabolismo , Neuronas/metabolismo , Transporte Biológico/efectos de los fármacos , Células Cultivadas , Clonidina/farmacocinética , Humanos , Concentración de Iones de Hidrógeno , Cinética , Neuronas/citología , Neuronas/efectos de los fármacos , Factores de Tiempo , Tritio/metabolismo , Tritio/farmacocinética
13.
Eur J Pharm Sci ; 32(1): 69-76, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17644326

RESUMEN

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.


Asunto(s)
Cefalosporinas/metabolismo , Simportadores/metabolismo , Aminoácidos/química , Aminoácidos/metabolismo , Aminoácidos/farmacocinética , Animales , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacocinética , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Células CACO-2 , Ceftibuteno , Células Cultivadas , Cefalosporinas/química , Cefalosporinas/farmacocinética , Dipéptidos/química , Dipéptidos/metabolismo , Dipéptidos/farmacocinética , Humanos , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Cinética , Modelos Moleculares , Estructura Molecular , Oligopéptidos/química , Oligopéptidos/metabolismo , Oligopéptidos/farmacocinética , Transportador de Péptidos 1 , Unión Proteica , Ratas , Simportadores/antagonistas & inhibidores , Simportadores/fisiología
15.
J Med Chem ; 49(14): 4286-96, 2006 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-16821788

RESUMEN

The renal type H(+)/peptide cotransporter PEPT2 has a substantial influence on the in vivo disposition of dipeptides and tripeptides as well as peptide-like drugs within the body, particularly in kidney, lung, and the brain. The comparative molecular similarity indices analysis (CoMSIA) method was applied to identify those regions in the substrate structures that are responsible for recognition and for differences in affinity. We have developed a comprehensive 3D quantitative structure-activity relationship (3D-QSAR) model based on 83 compounds that is able to explain and predict the binding affinities of new PEPT2 substrates. This 3D-QSAR model possesses a high predictive power (q(2) = 0.755; r(2) = 0.893). An additional 3D-QSAR model based on the same compounds was generated and correlated with affinity data of the intestinal H(+)/peptide cotransporter PEPT1. By comparing the CoMSIA contour plots, differences in selectivity between the intestinal and the renal type peptide carrier become evident.


Asunto(s)
Antibacterianos/química , Oligopéptidos/química , Relación Estructura-Actividad Cuantitativa , Simportadores/química , beta-Lactamas/química , Cefalosporinas/química , Dipéptidos/química , Modelos Moleculares , Conformación Molecular , Penicilinas/química , Transportador de Péptidos 1 , Unión Proteica
16.
FASEB J ; 19(11): 1468-73, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16126914

RESUMEN

The proton-coupled amino acid transporter PAT1, cloned recently from brain and intestine, mediates the uphill transport of l- and d-proline, l-alanine, glycine, taurine, d-serine, GABA, and many other related compounds and drugs. Here we describe the novel finding that l-tryptophan and its derivatives tryptamine, 5-hydroxy-l-tryptophan, serotonin, and indole-3-propionic acid strongly inhibit H+-dependent l-[3H]proline uptake into Caco-2 cells with inhibition constants (K(i)) of 0.9 to 6.1 mM. Uptake of l-[3H]tryptophan into Caco-2 cells on the other hand was not inhibited by l-proline. Whereas PAT1 substrates produced significant changes in a membrane potential assay for electrogenic transport in Caco-2 cells, l-tryptophan, tryptamine, and 5-hydroxy-l-tryptophan failed to alter membrane voltage. When PAT1 was expressed in Xenopus laevis oocytes and analyzed by the two-electrode voltage clamp technique, glycine elicited high inward currents that were dependent on membrane potential but no currents were observed with l-tryptophan, tryptamine, 5-hydroxy-l-tryptophan, or serotonin. Although not transported electrogenically by PAT1, l-tryptophan and its derivatives inhibited glycine-evoked currents dose-dependently. We conclude that serotonin, l-tryptophan, and tryptamine bind to PAT1 with potencies similar to the prototype substrates, inhibit transport function but are not transported by this carrier protein. They may be considered as the carriers' naturally occurring inhibitors that may alter the transport function of PAT1.


Asunto(s)
Proteínas de Transporte de Membrana/efectos de los fármacos , Serotonina/farmacología , Triptaminas/farmacología , Triptófano/farmacología , 5-Hidroxitriptófano/farmacología , Animales , Transporte Biológico/efectos de los fármacos , Células CACO-2 , Glicina/farmacología , Humanos , Potenciales de la Membrana/efectos de los fármacos , Proteínas de Transporte de Membrana/fisiología , Oocitos/metabolismo , Prolina/metabolismo , Transportadores de Sulfato , Xenopus laevis
17.
Eur J Pharm Biopharm ; 63(3): 360-4, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16531028

RESUMEN

The recently cloned proton-coupled amino acid transporter 1 (PAT1) not only accepts several amino acids as substrates but also pharmaceutically relevant L-proline or GABA derivatives such as cis-4-hydroxy-L-proline, L-azetidine-2-carboxylic acid (LACA), 3-amino-1-propanesulfonic acid, nipecotic acid, and the antituberculotic agent D-cycloserine. Because human intestine expresses hPAT1 at the brush border membrane, the transporter may serve as a new oral drug delivery route. Using the human intestinal cell line Caco-2, we have investigated the influence of an inwardly directed proton gradient on the kinetic parameters of L-proline uptake. H+ altered only the apparent affinity of L-proline transport and not the maximal transport velocity. Similarly, treatment of the cells with diethylpyrocarbonate (DEPC), known to chemically modify histidyl residues and block their function, affected only the Kt value of L-proline transport. Both increasing pH and DEPC treatment strongly increased the inhibition constants (Ki) of several drugs at hPAT1. It is concluded that H+ stimulates hPAT1 primarily by increasing the substrate affinity with no detectable influence on the maximal transport velocity of the transporter.


Asunto(s)
Sistemas de Transporte de Aminoácidos/fisiología , Simportadores/fisiología , Transporte Biológico , Células CACO-2 , Catálisis , Humanos , Concentración de Iones de Hidrógeno , Prolina/metabolismo
18.
J Med Chem ; 48(13): 4410-9, 2005 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-15974593

RESUMEN

The utilization of the membrane transport protein PEPT1 as a drug delivery system is a promising strategy to enhance the oral bioavailability of drugs. Since very little is known about the substrate binding site of PEPT1, computational methods are a meaningful tool to gain a more detailed insight into the structural requirements for substrates. Three-dimensional quantitative structure-activity relationship (3D-QSAR) studies using the comparative molecular similarity indices analysis (CoMSIA) method were performed on a training set of 98 compounds. Affinity constants of beta-lactam antibiotics and tripeptides were determined at Caco-2 cells. A statistically reliable model of high predictive power was obtained (q(2) = 0.828, r(2) = 0.937). The results derived from CoMSIA were graphically interpreted using different field contribution maps. We identified those regions which are crucial for the interaction between peptidomimetics and PEPT1. The new 3D-QSAR model was used to design a new druglike compound mimicking a dipeptide. The predicted K(i) value was confirmed experimentally.


Asunto(s)
Oligopéptidos/síntesis química , Simportadores/metabolismo , beta-Lactamas/síntesis química , Animales , Línea Celular Tumoral , Dipéptidos/metabolismo , Diseño de Fármacos , Humanos , Mamíferos , Modelos Moleculares , Oligopéptidos/química , Oligopéptidos/farmacocinética , Transportador de Péptidos 1 , Relación Estructura-Actividad Cuantitativa , Especificidad por Sustrato , beta-Lactamas/química , beta-Lactamas/farmacocinética
19.
Biochem Pharmacol ; 70(12): 1851-60, 2005 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-16263091

RESUMEN

This study was performed to investigate which human organic cation transporter, hOCT1, hOCT2 or hOCT3, participates with regard to cation specificity and membrane localization in the intestinal absorption of orally available cationic drugs. Inhibition of N-[methyl-3H]4-phenylpyridinium ([3H]MPP+) uptake by various compounds into Caco-2 cells and into cells (HEK-293 or CHO) that were stably transfected with hOCT1, hOCT2 or hOCT3 was compared. The uptake of [3H]MPP+ into Caco-2 cells was inhibited by atropine, butylscopolamine, clonidine, diphenhydramine, etilefrine, quinine and ranitidine with IC50 values between 6 microM and 4 mM. Transepithelial, apical to basal flux of [3H]MPP+ across Caco-2 cell monolayers was also strongly inhibited by these compounds. The inhibitory potency of the cationic drugs and prototypical organic cations at Caco-2 cells correlated well with the inhibitory potency measured at CHO-hOCT3 cells but much less with that at HEK-hOCT1 and -hOCT2 cells. This is functional evidence for the predominant role of hOCT3. Etilefrine and atropine were specifically transported into CHO cells by hOCT3. In Caco-2 cells, the mRNA of all three hOCT and the proteins hOCT2 and hOCT3 were detected. More importantly, immunocytochemical analyses of human jejunum revealed for the first time that hOCT3 is localized to the brush border membrane whereas hOCT1 immunolabeling was mainly observed at the lateral membranes of the enterocytes.


Asunto(s)
Intestinos/química , Factor 1 de Transcripción de Unión a Octámeros/análisis , Proteínas de Transporte de Catión Orgánico/análisis , 1-Metil-4-fenilpiridinio/farmacocinética , Secuencia de Aminoácidos , Transporte Biológico/efectos de los fármacos , Células CACO-2 , Membrana Celular/química , Humanos , Datos de Secuencia Molecular , Factor 1 de Transcripción de Unión a Octámeros/fisiología , Proteínas de Transporte de Catión Orgánico/fisiología , Transportador 2 de Cátion Orgánico , Transfección
20.
Eur J Pharm Biopharm ; 59(1): 17-24, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15567297

RESUMEN

The activity of the renal peptide transporters PEPT2 and PEPT1 determines-among other factors such as metabolic stability in liver and plasma-the circulatory half-life of penicillins and cephalosporins during therapy. This study was initiated to examine systematically the interaction of beta-lactam antibiotics with PEPT2. Interaction of 31 cephalosporins and penicillins with the carrier protein was characterized by measuring their ability to inhibit the uptake of [(14)C]Gly-Sar into renal SKPT cells. Cefadroxil, cefaclor, cyclacillin, cephradine, cephalexin and moxalactam were recognized by PEPT2 with very high affinity comparable to that of natural dipeptides (K(i)=3-100microM). Ceftibuten, dicloxacillin, amoxicillin, metampicillin, cloxacillin, ampicillin, cefixime, cefamandole, oxacillin and cefmetazole interacted with PEPT2 with medium affinity (K(i)=0.1-5mM). For the other beta-lactam antibiotics studied interaction was very low or not measurable (K(i)>5mM). The affinity constants of beta-lactam antibiotics at rPEPT2 and hPEPT1 are significantly correlated, but the rank orders are not identical. Decisive differences between PEPT1 and PEPT2 recognition of the N-terminal part of the compounds became evident. Moreover, this large data set of affinity constants of beta-lactam antibiotics will be useful for structure-transport (binding) analyses of PEPT2.


Asunto(s)
Antibacterianos/metabolismo , Simportadores/metabolismo , beta-Lactamas/metabolismo , Animales , Antibacterianos/química , Sitios de Unión/fisiología , Células CACO-2 , Relación Dosis-Respuesta a Droga , Humanos , Transportador de Péptidos 1 , Unión Proteica/fisiología , Ratas , beta-Lactamas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA