Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochim Biophys Acta ; 1832(8): 1149-58, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23579073

RESUMEN

Elevated levels of homocysteine produce detrimental effects in humans but its role in preterm birth is not known. Here we used a mouse model of hyperhomocysteinemia to examine the relevance of homocysteine to preterm birth. The mouse carries a heterozygous deletion of cystathionine ß-synthase (Cbs(+/-)). Gestational period was monitored in wild type and Cbs(+/-) female mice. Mouse uterine and placental tissues, human primary trophoblast cells, and human myometrial and placental cell lines were used to determine the influence of homocysteine on expression of specific genes in vitro. The activity of BKCa channel in the myometrial cell line was monitored using the patch-clamp technique. We found that hyperhomocysteinemia had detrimental effects on pregnancy and induced preterm birth in mice. Homocysteine increased the expression of oxytocin receptor and Cox-2 as well as PGE2 production in uterus and placenta, and initiated premature uterine contraction. A Cox-2 inhibitor reversed these effects. Gpr109a, a receptor for niacin, induced Cox-2 in uterus. Homocysteine upregulated GPR109A and suppressed BKCa channel activity in human myometrial cells. Deletion of Gpr109a in Cbs(+/-) mice reversed premature birth. We conclude that hyperhomocysteinemia causes preterm birth in mice through upregulation of the Gpr109a/Cox-2/PGE2 axis and that pharmacological blockade of Gpr109a may have potential in prevention of preterm birth.


Asunto(s)
Homocisteína/sangre , Hiperhomocisteinemia/fisiopatología , Complicaciones del Embarazo/sangre , Nacimiento Prematuro/sangre , Animales , Línea Celular , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Dinoprostona/genética , Dinoprostona/metabolismo , Femenino , Homocisteína/genética , Homocisteína/metabolismo , Humanos , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Ratones , Ratones Endogámicos C57BL , Contracción Muscular , Miometrio/metabolismo , Miometrio/fisiopatología , Placenta/metabolismo , Placenta/fisiopatología , Embarazo , Complicaciones del Embarazo/genética , Complicaciones del Embarazo/fisiopatología , Nacimiento Prematuro/genética , Nacimiento Prematuro/fisiopatología , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Receptores de Oxitocina/genética , Receptores de Oxitocina/metabolismo , Trofoblastos/metabolismo , Regulación hacia Arriba , Útero/metabolismo , Útero/patología , Útero/fisiopatología
2.
Biochim Biophys Acta ; 1768(11): 2690-7, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17692818

RESUMEN

Recently, we cloned two Na(+)-coupled lactate transporters from mouse kidney, a high-affinity transporter (SMCT1 or slc5a8) and a low-affinity transporter (SMCT2 or slc5a12). Here we report on the cloning and functional characterization of human SMCT2 (SLC5A12) and compare the immunolocalization patterns of slc5a12 and slc5a8 in mouse kidney. The human SMCT2 cDNA codes for a protein consisting of 618 amino acids. When expressed in mammalian cells or Xenopus oocytes, human SMCT2 mediates Na(+) -coupled transport of lactate, pyruvate and nicotinate. The affinities of the transporter for these substrates are lower than those reported for human SMCT1. Several non-steroidal anti-inflammatory drugs inhibit human SMCT2-mediated nicotinate transport, suggesting that NSAIDs interact with the transporter as they do with human SMCT1. Immunofluorescence microscopy of mouse kidney sections with an antibody specific for SMCT2 shows that the transporter is expressed predominantly in the cortex. Similar studies with an anti-SMCT1 antibody demonstrate that SMCT1 is also expressed mostly in the cortex. Dual-labeling of SMCT1 and SMCT2 with 4F2hc (CD98), a marker for basolateral membrane of proximal tubular cells in the S1 and S2 segments of the nephron, shows that both SMCT1 and SMCT2 are expressed in the apical membrane of the tubular cells. These studies also show that while SMCT2 is broadly expressed along the entire length of the proximal tubule (S1/S2/S3 segments), the expression of SMCT1 is mostly limited to the S3 segment. These studies suggest that the low-affinity transporter SMCT2 initiates lactate absorption in the early parts of the proximal tubule followed by the participation of the high-affinity transporter SMCT1 in the latter parts of the proximal tubule.


Asunto(s)
Riñón/química , Transportadores de Ácidos Monocarboxílicos/análisis , Transportadores de Ácidos Monocarboxílicos/genética , Secuencia de Aminoácidos , Animales , Clonación Molecular , ADN Complementario/química , Técnica del Anticuerpo Fluorescente , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Transportadores de Ácidos Monocarboxílicos/fisiología , Simportadores , Xenopus laevis
3.
J Clin Invest ; 107(8): 1035-43, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11306607

RESUMEN

Nitric oxide synthase (NOS) inhibitors have therapeutic potential in the management of numerous conditions in which NO overproduction plays a critical role. Identification of transport systems in the intestine that can mediate the uptake of NOS inhibitors is important to assess the oral bioavailability and therapeutic efficacy of these potential drugs. Here, we have cloned the Na+ - and Cl- -coupled amino acid transport system B(0,+) (ATB(0,+)) from the mouse colon and investigated its ability to transport NOS inhibitors. When expressed in mammalian cells, ATB(0,+) can transport a variety of zwitterionic and cationic amino acids in a Na+ - and Cl- -coupled manner. Each of the NOS inhibitors tested compete with glycine for uptake through this transport system. Furthermore, using a tritiated analog of the NOS inhibitor N(G)-nitro-L-arginine, we showed that Na+ - and Cl- -coupled transport occurs via ATB(0,+). We then studied transport of a wide variety of NOS inhibitors in Xenopus laevis oocytes expressing the cloned ATB(0,+) and found that ATB(0,+) can transport a broad range of zwitterionic or cationic NOS inhibitors. These data represent the first identification of an ion gradient-driven transport system for NOS inhibitors in the intestinal tract.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC , Proteínas Portadoras/metabolismo , Cloruros/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Receptores Virales/metabolismo , Sodio/metabolismo , Animales , Transporte Biológico Activo , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Perfilación de la Expresión Génica , Humanos , Intestino Delgado/metabolismo , Ratones , Antígenos de Histocompatibilidad Menor , Proteínas de Transporte de Neurotransmisores , ARN Mensajero , Receptores Virales/genética , Receptores Virales/fisiología , Xenopus laevis
4.
Int J Pharm ; 336(1): 133-9, 2007 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-17184941

RESUMEN

We have previously demonstrated that dopamine conjugation to glucose allows it to induce therapeutic effects against Parkinson's disease after intravenous administration. In this paper we demonstrate that, unlike dopamine, the prodrug glu-dopamine is a transportable substrate of glucose transporters. Towards this, the effect of glucose-conjugation on the affinity and uptake of dopamine have been assessed in vitro, using human retinal pigment epithelium (HRPE) cells. Glucose transporter-mediated uptake was measured using [(3)H]3-O-methylglucose ([(3)H]3-O-MG) as the tracer. The uptake was found to be rapid and hyperbolically related to its concentrations (K(t)=7.8+/-1.2mM and V(max)=54+/-2 nmol/min mg protein). Inhibition experiments showed that dopamine was able to interact with glucose carriers only when conjugated to glucose (IC(50)=2.6+/-0.6mM). HPLC analysis of HRPE cell extracts showed that both dopamine and the prodrug permeate the cell, but only the uptake of the prodrug is inhibitable by glucose. This confirms that glucose transporters mediate the transport of the prodrug glu-dopamine, but not of dopamine. HRPE cells is therefore proposed as a promising model for in vitro studies involving the glucose transporter-mediated transport of drugs and their conjugates.


Asunto(s)
Dopamina/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Glucosa/metabolismo , Profármacos/metabolismo , 3-O-Metilglucosa/metabolismo , Transporte Biológico , Células Cultivadas , Cromatografía Líquida de Alta Presión , Dopamina/química , Dopamina/farmacocinética , Células Epiteliales/química , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Glucosa/química , Glucosa/farmacocinética , Humanos , Cinética , Estructura Molecular , Epitelio Pigmentado Ocular/citología , Profármacos/farmacocinética
5.
Placenta ; 27(6-7): 550-9, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16129486

RESUMEN

NaS2 is a Na+-coupled transporter for sulfate that belongs to the SLC13 gene family. This transporter was originally cloned from high endothelial venule endothelial cells, but nothing is known about the functional characteristics of this transporter except that it transports sulfate in a Na+-coupled manner. Northern blot analysis indicates that NaS2 is expressed most robustly in placenta. In the present study, we cloned NaS2 from rat placenta and characterized its transport function in detail using the Xenopus laevis oocyte expression system. Rat NaS2 consists of 629 amino acids and is highly similar to human NaS2. In situ hybridization studies with mouse placental sections show that NaS2 transcripts are expressed primarily in trophoblasts of the labyrinth zone. The expression of the transporter is confirmed in primary cultures of trophoblasts isolated from human placenta. When expressed in X. laevis oocytes, rat NaS2 mediates Na+-coupled transport of sulfate. The transport of sulfate is inhibited by oxyanions of selenium, chromium, arsenic, molybdenum, and phosphorous, suggesting that the transporter may mediate the transport of these oxyanions in addition to sulfate. The Kt for sulfate is 153+/-30 microM and the Na+:sulfate stoichiometry is 3:1. The transport process is electrogenic as evidenced from the inhibition of the uptake process by K+-induced depolarization. We conclude that NaS2 is a placenta-specific Na+-coupled, electrogenic, transporter for sulfate expressed in trophoblasts and that it is also responsible for the transport of oxyanions of the micronutrients selenium and chromium.


Asunto(s)
Aniones/metabolismo , Cromo/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Selenio/metabolismo , Sulfatos/metabolismo , Simportadores/metabolismo , Trofoblastos/metabolismo , Secuencia de Aminoácidos , Animales , Biblioteca de Genes , Humanos , Hibridación in Situ , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/metabolismo , Ratas , Homología de Secuencia de Aminoácido , Transportadores de Sulfato , Xenopus laevis
6.
Br J Pharmacol ; 173(23): 3292-3306, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27747870

RESUMEN

BACKGROUND AND PURPOSE: Pancreatic cancer is a solid tumour that is often fatal. Hence, there is an urgent need to identify new drug targets for this disease. Highly proliferating cancer cells have an increased demand for nutrients and, therefore, need to up-regulate selective amino acid transporters. Here, we investigated which amino acid transporters are up-regulated in pancreatic cancer and whether any of these transporters has potential as a drug target for this fatal disease. EXPERIMENTAL APPROACH: The expression of amino acid transporters in pancreatic cancer was analysed using publicly available microarray datasets, and the findings with the transporter SLC6A14 were validated by mRNA and protein analysis. The potential of SLC6A14 as a drug target was evaluated using a pharmacological blocker in vitro and in vivo. KEY RESULTS: SLC6A14 was up-regulated several fold in patient-derived xenografts, primary tumour tissues and pancreatic cancer cells lines compared to normal pancreatic tissue or normal pancreatic epithelial cells. The magnitude of the up-regulation of SLC6A14 was the highest among the amino acid transporters examined. A pharmacological blocker of SLC6A14, α-methyltryptophan, induced amino acid starvation in pancreatic cancer cells and reduced the growth and proliferation of these cells, both in vitro and in vivo. CONCLUSION AND IMPLICATIONS: The salient features of this study are that SLC6A14 is markedly up-regulated in pancreatic cancer and that pharmacological blockade of this transporter interferes with amino acid nutrition and reduces growth and proliferation of pancreatic cancer cells. These findings identify SLC6A14 as a novel druggable target for pancreatic cancer.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/genética , Terapia Molecular Dirigida , Neoplasias Pancreáticas/tratamiento farmacológico , Triptófano/análogos & derivados , Sistemas de Transporte de Aminoácidos , Sistemas de Transporte de Aminoácidos Neutros/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , ARN Mensajero/metabolismo , Triptófano/farmacología , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Biochim Biophys Acta ; 1222(2): 309-14, 1994 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-8031869

RESUMEN

The mechanism of transport of 5-methyltetrahydrofolate, the predominant form of folate in human blood, was investigated in JAR human placental choriocarcinoma cells. Northern analysis revealed the presence of the folate receptor mRNA in these cells. Accumulation of 5-methyltetrahydrofolate in these cells was saturable and inhibited by other folate analogs. The transport of 5-methyltetrahydrofolate into the cytoplasm was decreased by the anion transport inhibitors probenecid and 4,4'-diisothiocyanostilbene 2,2'-disulfonic acid. The cytosolic transport was dependent on a transmembrane H+ gradient because NH4Cl and chloroquine which alkalinize acidic compartments in the cell and protonophores which dissipate transmembrane H+ gradients inhibited the transport. The inhibitors of receptor-mediated endocytosis, monodansylcadaverine and bacitracin, had no effect on the cytosolic transport nor on the accumulation. Bafilomycin A1, a specific inhibitor of the vacuolar type (V-type) H(+)- pump, caused a significant reduction in the cytosolic transport of 5-methyltetrahydrofolate, without affecting the binding of the vitamin to its membrane bound receptor. It is concluded that (a) the JAR cells transport 5-methyltetrahydrofolate via a specific probenecid-sensitive folate transporter, (b) the transporter is driven by a transmembrane H+ gradient, and (c) the H+ gradient involved in this process is generated by a bafilomycin A1-sensitive V-type H(+)-pump present in the plasma membrane. The results strongly suggest that the transport of folate in these cells occurs by potocytosis involving a functional coupling between the folate receptor, the folate transporter and the H+ pump.


Asunto(s)
Antibacterianos/farmacología , Proteínas Portadoras/metabolismo , Ácido Fólico/metabolismo , Macrólidos , Probenecid/farmacología , Bombas de Protones , Receptores de Superficie Celular , Secuencia de Bases , Coriocarcinoma/metabolismo , Femenino , Receptores de Folato Anclados a GPI , Humanos , Datos de Secuencia Molecular , Embarazo , Tetrahidrofolatos/metabolismo , Células Tumorales Cultivadas , Neoplasias Uterinas/metabolismo
8.
Biochim Biophys Acta ; 1369(1): 141-51, 1998 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-9528682

RESUMEN

We investigated the transport pathways available for the uptake of vitamin C in the human placental choriocarcinoma cell line, JAR. These cells were found to possess the capacity to accumulate the vitamin when presented either in the oxidized form (dehydroascorbic acid) or in the reduced form (ascorbate). Dithiothreitol and 5,5'-dithiobis(2-nitrobenzoic acid) were used to maintain vitamin C as ascorbate and dehydroascorbic acid, respectively. The uptake of these two forms of vitamin C in JAR cells was found to occur by different mechanisms. The uptake of the dehydroascorbic acid was Na(+)-independent and was mediated by facilitative glucose transporters as evidenced from the inhibition of the uptake process by glucose. On the other hand, the uptake of ascorbate was Na(+)-dependent and was not sensitive to inhibition by glucose. Substitution of Na+ with other monovalent cations abolished the uptake of ascorbate completely. The uptake process was, however, not influenced by anions. Kinetic analysis indicated the presence of a single saturable transport system for ascorbate with a Michaelis-Menten constant of 22 +/- 1 microM. The dependence of the uptake rare of ascorbate on Na+ concentration exhibited sigmoidal kinetics, suggesting interaction of more than one Na+ ion with the transporter. The Hill coefficient for the Na+ interaction was 2, indicating that the Na(+)-dependent ascorbate transport is electrogenic. The Na(+)-dependent stimulation of ascorbate uptake was primarily due to an increase in the affinity of the transporter for ascorbate in the presence of Na+. It is concluded that the JAR placental trophoblast cell line expresses two different transport systems for vitamin C: one for the reduced form of the vitamin ascorbate; and the other for the oxidized form of the vitamin dehydroascorbic acid.


Asunto(s)
Ácido Ascórbico/metabolismo , Coriocarcinoma/metabolismo , Placenta/patología , Neoplasias Uterinas/metabolismo , Ácido Ascórbico/farmacocinética , Transporte Biológico , Proteínas Portadoras/metabolismo , Coriocarcinoma/patología , Ácido Ditionitrobenzoico/química , Ditiotreitol/química , Femenino , Humanos , Sodio/metabolismo , Células Tumorales Cultivadas , Neoplasias Uterinas/patología
9.
Biochim Biophys Acta ; 1512(2): 299-307, 2001 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-11406107

RESUMEN

The present study was undertaken to elucidate the functional characteristics of choline uptake and deduce the relationship between choline uptake and the expression of organic cation transporters in the rat brain microvessel endothelial cell line RBE4. Confluent RBE4 cells were found to express a high affinity choline uptake system. The system is Na(+)-independent and shows a Michaelis-Menten constant of approx. 20 microM for choline. The choline analogue hemicholinium-3 inhibits choline uptake in these cells with an inhibition constant of approx. 50 microM. The uptake system is also susceptible for inhibition by various organic cations, including 1-methyl-4-phenylpyridinium, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, clonidine, procainamide, and tetramethylammonium. The prototypical organic cation tetraethylammonium shows very little affinity for the choline uptake system in these cells. The inhibition of choline uptake by hemicholinium-3 is competitive. Northern analysis and RT-PCR show that these cells do not express the organic cation transporters OCT2 and OCT3. These cells do express, however, low levels of OCT1, but the functional characteristics of choline uptake in these cells are very different from the known properties of choline uptake via OCT1. The Na(+)-coupled high affinity choline transporter CHT1 is not expressed in these cells as evidenced by RT-PCR. This corroborates the Na(+)-independent nature of choline uptake in these cells. It is concluded that RBE4 cells express an organic cation transporter that is responsible for choline uptake in these cells and that this transporter is not identical to any of the organic cation transporters thus far identified at the molecular level in mammalian cells.


Asunto(s)
Proteínas Portadoras/metabolismo , Circulación Cerebrovascular/fisiología , Colina/metabolismo , Endotelio Vascular/metabolismo , Proteínas de la Membrana/metabolismo , Microcirculación/fisiología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , 1-Metil-4-fenilpiridinio/farmacología , Animales , Transporte Biológico/efectos de los fármacos , Encéfalo/irrigación sanguínea , Tronco Encefálico/metabolismo , Proteínas Portadoras/genética , Línea Celular , Clonidina/farmacología , Endotelio Vascular/citología , Hemicolinio 3/farmacología , Riñón/metabolismo , Cinética , Proteínas de la Membrana/genética , Transportador 1 de Catión Orgánico , Compuestos de Amonio Cuaternario/farmacología , ARN Mensajero/genética , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sodio/metabolismo , Tetraetilamonio/farmacología , Transcripción Genética
10.
Biochim Biophys Acta ; 1467(1): 1-6, 2000 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-10930503

RESUMEN

We report here on the primary structure and functional characteristics of the protein responsible for the system A amino acid transport activity that is known to be expressed in most human tissues. This transporter, designated ATA2 for amino acid transporter A2, was cloned from the human hepatoma cell line HepG2. Human ATA2 (hATA2) consists of 506 amino acids and exhibits a high degree of homology to rat ATA2. hATA2-specific mRNA is ubiquitously expressed in human tissues. When expressed in mammalian cells, hATA2 mediates Na+-dependent transport of alpha-(methylamino)isobutyric acid, a specific model substrate for system A. The transporter is specific for neutral amino acids. It is pH-sensitive and Li+-intolerant. The Na+:amino acid stoichiometry is 1:1.


Asunto(s)
Proteínas Portadoras/fisiología , Secuencia de Aminoácidos , Sistemas de Transporte de Aminoácidos , Transporte Biológico , Radioisótopos de Carbono , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/química , Clonación Molecular , Regulación de la Expresión Génica , Biblioteca de Genes , Humanos , Datos de Secuencia Molecular , ARN Mensajero/análisis , Especificidad por Sustrato , Transfección , Células Tumorales Cultivadas , beta-Alanina/análogos & derivados , beta-Alanina/fisiología
11.
Biochim Biophys Acta ; 1324(2): 296-308, 1997 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-9092716

RESUMEN

The present study was undertaken to investigate the interaction of anionic cephalosporins (cefixime, ceftibuten, and cefdinir) with the renal peptide transporter (PEPT 2) and the intestinal peptide transporter (PEPT 1) using four different experimental model systems. In the first approach, the human colon carcinoma cell line Caco-2 which expresses PEPT 1 and the SHR rat kidney cell line SKPT which expresses PEPT 2 were used. The uptake of the dipeptide Gly-Sar mediated by PEPT 1 or PEPT 2 in these cells was inhibited significantly by the anionic cephalosporins, with the following order of potency: ceftibuten > cefixime > cefdinir. The inhibition was competitive in nature. Even though the order of potency was the same for PEPT 1 and PEPT 2, PEPT 1 exhibited much lesser sensitivity to inhibition than PEPT 2. In the second approach, the cloned human PEPT 1 and PEPT 2 were functionally expressed in HeLa cells following which the cells were used to study the interaction of anionic cephalosporins with PEPT 1 and PEPT 2. Again, Gly-Sar uptake mediated by the human PEPT 1 and PEPT 2 in HeLa cells was found to be inhibited by the anionic cephalosporins with the same order potency as in Caco-2 and SKPT cells. In the third approach, brush border membrane vesicles isolated from rat kidneys were employed. In this approach also it was found that PEPT 2-mediated Gly-Sar uptake was inhibited by cefixime and ceftibuten. In the fourth approach, the human PEPT 1 was expressed in Xenopus laevis oocytes and PEPT 1-mediated transport of ceftibuten was investigated directly by electrophysiological methods. Ceftibuten evoked inward currents in PEPT 1-expressing oocytes but not in water-injected oocytes, showing that the transport of the anionic cephalosporin via PEPT 1 is associated with transfer of positive charge. The ceftibuten-evoked currents were saturable with respect to ceftibuten concentration and were markedly dependent on membrane potential. It is concluded that anionic cephalosporins interact with the peptide transporters expressed in the intestine (PEPT 1) as well as in the kidney (PEPT 2).


Asunto(s)
Proteínas Portadoras/metabolismo , Cefalosporinas/farmacología , Colon/metabolismo , Riñón/metabolismo , Simportadores , Animales , Transporte Biológico/efectos de los fármacos , Células CACO-2/metabolismo , Proteínas Portadoras/genética , Línea Celular , Dipéptidos/metabolismo , Potenciales Evocados , Células HeLa , Humanos , Microvellosidades/metabolismo , Oocitos/fisiología , Transportador de Péptidos 1 , Ratas , Ratas Endogámicas SHR , Ratas Sprague-Dawley , Transfección , Xenopus laevis
12.
Biochim Biophys Acta ; 1461(1): 1-9, 1999 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-10556483

RESUMEN

We report here on the cloning and functional characterization of human Na(+)-dependent vitamin C transporter 1 (SVCT1). The human SVCT1 cDNA, obtained from a Caco2 cell cDNA library, encodes a protein of 598 amino acids with 12 putative transmembrane domains. The SVCT1-specific transcript, 2.4 kb in size, is expressed in kidney, liver, small intestine, thymus and prostate. When expressed heterologously in HRPE cells, SVCT1 mediates the transport of ascorbate, the reduced form of vitamin C, in a Na(+)-dependent manner. The transporter is specific for ascorbate with a K(t) of approximately 75 microM. The relationship between the cDNA-specific uptake rate of ascorbate and Na(+) concentration is sigmoidal with a Na(+):ascorbate stoichiometry of 2:1, indicating that the transport process is electrogenic. In Caco2 cells and in normal human intestine, SVCT1 also exists as a non-functional splice variant with a four amino acid sequence inserted between E-155 and V-156. The splice variant results from the use of a donor site 12 bp downstream of the normal donor site.


Asunto(s)
Transportadores de Anión Orgánico Sodio-Dependiente , Proteínas/química , Cloruro de Sodio/farmacología , Simportadores , Secuencia de Aminoácidos , Ácido Ascórbico/metabolismo , Secuencia de Bases , Transporte Biológico/efectos de los fármacos , Células CACO-2 , Línea Celular , Clonación Molecular , ADN Complementario/biosíntesis , ADN Complementario/química , Biblioteca de Genes , Humanos , Mucosa Intestinal/metabolismo , Cinética , Datos de Secuencia Molecular , Proteínas/análisis , Proteínas/genética , Empalme del ARN , ARN Mensajero/aislamiento & purificación , Transportadores de Sodio Acoplados a la Vitamina C , Especificidad por Sustrato , Transfección
13.
Biochim Biophys Acta ; 1282(2): 274-82, 1996 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-8703983

RESUMEN

The interaction of carnitine with human placental brush-border membrane vesicles was investigated. Carnitine was found to associate with the membrane vesicles in a Na(+)-dependent manner. The time course of this association did not exhibit an overshoot, which is typical of a Na+ gradient-driven transport process. The absolute requirement for Na+ was noticeable whether the association of carnitine with the vesicles was measured with a short time incubation or under equilibrium conditions, indicating Na(+)-dependent binding of carnitine to the human placental brush-border membranes. The binding was saturable and was of a high-affinity type with a dissociation constant of 1.37 +/- 0.03 microM. Anions had little or no influence on the binding process. The binding process was specific for carnitine and its acyl derivatives. Betaine also competed for the binding process, but other structurally related compounds did not. Kinetic analyses revealed that Na+ increased the affinity of the binding process for carnitine and the Na+/carnitine coupling ratio for the binding process was 1. The dissociation constant for the interaction of Na+ with the binding of carnitine was 24 +/- 4 mM. This constitutes the first report on the identification of Na(+)-dependent high-affinity carnitine binding in the plasma membrane of a mammalian cell. Studies with purified rat renal brush-border membrane vesicles demonstrated the presence of Na+ gradient-driven carnitine transport but no Na(+)-dependent carnitine binding in these membrane vesicles. In contrast, purified intestinal brush-border membrane vesicles posses neither Na+ gradient-driven carnitine transport nor Na(+)-dependent carnitine binding.


Asunto(s)
Carnitina/metabolismo , Microvellosidades/metabolismo , Placenta/ultraestructura , Sodio/farmacología , Aniones , Betaína/metabolismo , Unión Competitiva , Transporte Biológico , Cationes , Femenino , Humanos , Intestinos/ultraestructura , Riñón/ultraestructura , Cinética , Liposomas/metabolismo , Cloruro de Potasio/farmacología , Embarazo
14.
Biochim Biophys Acta ; 1512(1): 15-21, 2001 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-11334620

RESUMEN

We investigated the molecular mechanism involved in the adaptive regulation of the amino acid transport system A, a process in which amino acid starvation induces the transport activity. These studies were done with rat C6 glioma cells. System A activity in these cells is mediated exclusively by the system A subtype, amino acid transporter A2 (ATA2). The other two known system A subtypes, ATA1 and ATA3, are not expressed in these cells. Exposure of these cells to an amino acid-free medium induces system A activity. This process consists of an acute phase and a chronic phase. Laser-scanning confocal microscopic immunolocalization of ATA2 reveals that the acute phase is associated with recruitment of preformed ATA2 from an intracellular pool to the plasma membrane. In contrast, the chronic phase is associated with an induction of ata2 gene expression as evidenced from the increase in the steady-state levels of ATA2 mRNA, restoration of the intracellular pool of ATA2 protein, and blockade of the induction by cycloheximide and actinomycin D. The increase in system A activity induced by amino acid starvation is blocked specifically by system A substrates, including the non-metabolizable alpha-(methylamino)isobutyric acid.


Asunto(s)
Proteínas Portadoras/genética , Regulación de la Expresión Génica , Adaptación Fisiológica , Sistemas de Transporte de Aminoácidos , Aminoácidos/biosíntesis , Aminoácidos/deficiencia , Animales , Transporte Biológico , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , ARN Mensajero/análisis , Ratas , Especificidad por Sustrato , Células Tumorales Cultivadas
15.
Biochim Biophys Acta ; 1510(1-2): 10-7, 2001 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-11342143

RESUMEN

We report here on the cloning and functional characterization of the third subtype of amino acid transport system A, designated ATA3 (amino acid transporter A3), from a human liver cell line. This transporter consists of 547 amino acids and is structurally related to the members of the glutamine transporter family. The human ATA3 (hATA3) exhibits 88% identity in amino acid sequence with rat ATA3. The gene coding for hATA3 contains 16 exons and is located on human chromosome 12q13. It is expressed almost exclusively in the liver. hATA3 mediates the transport of neutral amino acids including alpha-(methylamino)isobutyric acid (MeAIB), the model substrate for system A, in a Na(+)-coupled manner and the transport of cationic amino acids in a Na(+)-independent manner. The affinity of hATA3 for cationic amino acids is higher than for neutral amino acids. The transport function of hATA3 is thus similar to that of system y(+)L. The ability of hATA3 to transport cationic amino acids with high affinity is unique among the members of the glutamine transporter family. hATA1 and hATA2, the other two known members of the system A subfamily, show little affinity toward cationic amino acids. hATA3 also differs from hATA1 and hATA2 in exhibiting low affinity for MeAIB. Since liver does not express any of the previously known high-affinity cationic amino acid transporters, ATA3 is likely to provide the major route for the uptake of arginine in this tissue.


Asunto(s)
Aminoácidos Neutros/metabolismo , Proteínas Portadoras/metabolismo , Hígado/metabolismo , beta-Alanina/análogos & derivados , Sistemas de Transporte de Aminoácidos , Arginina/metabolismo , Transporte Biológico , Northern Blotting , Proteínas Portadoras/genética , Línea Celular , Cromosomas Humanos Par 12 , Células Epiteliales/metabolismo , Exones , Glicina/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Intrones , ARN Mensajero/análisis , Especificidad por Sustrato , beta-Alanina/metabolismo
16.
Biochim Biophys Acta ; 1223(1): 71-5, 1994 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-8061055

RESUMEN

The folate receptor (FR), an essential component in the process of folate uptake in various cells, is known to exist in three isoforms, FR-alpha, FR-beta and FR-gamma, with differential tissue expression. Transfer of folate across the human placenta from mother to fetus involves participation of a folate receptor expressed in the syncytiotrophoblast, but the isoform identity of this receptor has not been established. Based on the tissue/cell type from which these isoforms have been cloned, it is currently believed that FR-alpha is the isoform expressed in adult tissues whereas FR-beta is the isoform expressed in fetal tissues including placenta. The present study, undertaken primarily to establish the isoform identity of the FR expressed in the placental syncytiotrophoblast, does not support this currently prevailing nomenclature. Reverse transcription coupled with polymerase chain reaction (RT-PCR) of total/poly(A)+ RNA from placenta, cultured trophoblast cells and JAR choriocarcinoma cells with primer pairs specific for either FR-alpha or FR-beta reveals that while both isoforms are detectable in the whole placental tissue, only FR-alpha is present in the normal trophoblast cells and in the choriocarcinoma cells. Northern analysis with probes designed to distinguish between the mRNA transcripts coding for these two isoforms corroborate the RT-PCR findings. Furthermore, the nucleotide sequences of the PCR products obtained from the trophoblast cells and JAR cells are identical to the nucleotide sequence of the FR-alpha cDNA. These studies establish that it is the FR-alpha isoform, and not the FR-beta isoform, which is selectively expressed in the placental trophoblast cells. FR-beta, which is known to be present in the placenta, most likely arises from the maternal decidua normally associated with this tissue.


Asunto(s)
Proteínas Portadoras/genética , Coriocarcinoma/metabolismo , Receptores de Superficie Celular , Trofoblastos/metabolismo , Neoplasias Uterinas/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Transporte Biológico , Línea Celular , ADN Complementario/aislamiento & purificación , Femenino , Receptores de Folato Anclados a GPI , Expresión Génica , Humanos , Datos de Secuencia Molecular , Embarazo , Células Tumorales Cultivadas
17.
Biochim Biophys Acta ; 1284(1): 109-17, 1996 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-8865821

RESUMEN

The JAR human placental choriocarcinoma cells were found to transport carnitine into the intracellular space by a Na(+)-dependent process. The transport showed no requirement for anions. The Na+-dependent process was saturable and the apparent Michaelis-Menten constant for carnitine was 12.3 +/- 0.5 microM. Na+ activated the transport by increasing the affinity of the transport system for carnitine. The transport system specifically interacted with L-carnitine, D-carnitine, acetyl-DL-carnitine and betaine. 6-N-Trimethyllysine and choline had little or no effect on carnitine transport. Of the total transport measured, transport into the intracellular space represented 90%. Plasma membrane vesicles prepared from JAR cells were found to bind carnitine in a Na(+)-dependent manner. The binding was saturable with an apparent dissociation constant of 0.66 +/- 0.08 microM. The binding process was specific for L-carnitine, D-carnitine, acetyl-DL-carnitine, and betaine. 6-N-Trimethyllysine and choline showed little or no affinity. It is concluded that the JAR cells express a Na(+)-dependent high-affinity system for carnitine transport and that the Na(+)-dependent high-affinity carnitine binding detected in purified JAR cell plasma membrane vesicles is possibly related to the transmembrane transport process.


Asunto(s)
Carnitina/metabolismo , Coriocarcinoma/metabolismo , Placenta/metabolismo , Sodio/metabolismo , Neoplasias Uterinas/metabolismo , Sitios de Unión , Transporte Biológico , Membrana Celular/metabolismo , Femenino , Humanos , Embarazo , Células Tumorales Cultivadas
18.
Biochim Biophys Acta ; 1463(1): 6-14, 2000 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-10631289

RESUMEN

We have isolated a cDNA from a rabbit intestinal cDNA library which, when co-expressed with the heavy chain of the human 4F2 antigen (4F2hc) in mammalian cells, induces system L-like amino acid transport activity. This protein, called LAT2, consists of 535 amino acids and is distinct from LAT1 which also interacts with 4F2hc to induce system L-like amino acid transport activity. LAT2 does not interact with rBAT, a protein with a significant structural similarity to 4F2hc. The 4F2hc/LAT2-mediated transport process differs from the 4F2hc/LAT1-mediated transport in substrate specificity, substrate affinity, tissue distribution, interaction with D-amino acids, and pH-dependence. The 4F2hc/LAT2-associated transport process has a broad specificity towards neutral amino acids with K(t) values in the range of 100-1000 microM, does not interact with D-amino acids to any significant extent, and is stimulated by acidic pH. In contrast, the 4F2hc/LAT1-associated transport process has a narrower specificity towards neutral amino acids, but with comparatively higher affinity (K(t) values in the range of 10-20 microM), interacts with some D-amino acids with high affinity, and is not influenced by pH. LAT2 is expressed primarily in the small intestine and kidney, whereas LAT1 exhibits a much broader tissue distribution.


Asunto(s)
Aminoácidos/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Mucosa Intestinal/metabolismo , Secuencia de Aminoácidos , Sistemas de Transporte de Aminoácidos , Animales , Transporte Biológico Activo , Proteínas Portadoras/química , Clonación Molecular , ADN Complementario/genética , ADN Complementario/aislamiento & purificación , Glutamina/metabolismo , Humanos , Cinética , Datos de Secuencia Molecular , Conejos , Homología de Secuencia de Aminoácido , Distribución Tisular
19.
Endocrinology ; 134(2): 574-81, 1994 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8299556

RESUMEN

The relationship between the transport of the thyroid hormone T3 and the transport of neutral amino acids was investigated in JAR human placental choriocarcinoma cells. The uptake of leucine, mediated by the amino acid transport system L, was inhibited by T3 and T4, and the nature of inhibition was competitive. Uptake of T3 into the cells was predominantly Na+ independent, and so was that of leucine. However, although an acidic extracellular pH stimulated leucine uptake, the uptake of T3 remained unaffected. In addition, leucine failed to inhibit T3 uptake. The aromatic amino acids phenylalanine and tryptophan were found to inhibit the uptake of T3, but these two amino acids were transported into the cells predominantly via system L. The amino acid transport system T, which is specific for aromatic amino acids, was not detectable in these cells. Treatment of the cells with the calmodulin antagonist CGS 9343 B stimulated the uptake of leucine and tryptophan, but inhibited the uptake of T3. Kinetic analysis of T3 uptake revealed the presence of a single saturable system for this hormone in these cells, and the Michaelis-Menten constant for this system was 0.77 +/- 0.06 microM. Metabolic poisons that interfere with the cellular generation of ATP had no effect on the uptake of T3. It is concluded that in placental choriocarcinoma cells, 1) T3 and T4 are high affinity competitive inhibitors of the amino acid transport system L, 2) uptake of T3 occurs via a specific Na(+)-independent, energy-independent, and saturable mechanism that is unrelated to the amino acid transport systems L and T, 3) the aromatic amino acids phenylalanine and tryptophan interact, although weakly, with the T3 uptake system, and 4) calmodulin-dependent processes participate in the regulation of the T3 uptake system.


Asunto(s)
Aminoácidos/metabolismo , Tiroxina/metabolismo , Triyodotironina/metabolismo , Aminoácidos/farmacología , Transporte Biológico/efectos de los fármacos , Línea Celular , Colina/farmacología , Coriocarcinoma , Femenino , Humanos , Concentración de Iones de Hidrógeno , Radioisótopos de Yodo , Cinética , Leucina/metabolismo , Fenilalanina/metabolismo , Embarazo , Sodio/farmacología , Tiroxina/farmacología , Triyodotironina/farmacología , Tritio , Triptófano/metabolismo , Células Tumorales Cultivadas , Neoplasias Uterinas
20.
Invest Ophthalmol Vis Sci ; 38(8): 1578-87, 1997 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9224286

RESUMEN

PURPOSE: To determine the identity of the transport process that is responsible for the uptake of N5-methyltetrahydrofolate, the predominant form of folate in blood, into cultured human retinal pigment epithelial cells. METHODS: Human retinal pigment epithelial cells were cultured on an impermeable plastic support, and the characteristics of the uptake of radiolabeled N5-methyltetrahydrofolate into the cells were investigated. The expression of the folate receptor and the reduced-folate transporter in these cells was evaluated by functional assays and by Northern blot analysis. In addition, the characteristics of N5-methyltetrahydrofolate uptake in these cells were compared with those in folate transport-defective human breast cancer cells that were manipulated to express functionally by transfection the cloned human folate receptor and the human reduced-folate transporter. RESULTS: Transport of N5-methyltetrahydrofolate into these cells occurred by a single saturable process with a Michaelis-Menten constant of 0.13 +/- 0.01 microM. The process was specific for such reduced folates as N5-methyltetrahydrofolate and N5-formyltetrahydrofolate. Nonreduced folate interacted with this transport process only weakly. The transport process was inhibited by anion transport inhibitors. Results of Northern blot analysis indicated the presence of transcripts specific for the reduced-folate transporter in these cells. These cells expressed very small amounts of the folate receptor evidenced from the binding of folate and from the detectable presence of folate receptor-specific transcript, but there was no evidence for participation of the receptor in the observed transport of N5-methyltetrahydrofolate. There was also no evidence in these cells for expression of the folate transporter that prefers nonreduced folate as a substrate. CONCLUSIONS: Transport of N5-methyltetrahydrofolate in human retinal pigment epithelial cells occurs exclusively through the reduced-folate transporter. The folate receptor is expressed at negligible levels in these cells and does not participate in the observed transport. Because the cells were cultured on impermeable supports, making the basolateral membrane inaccessible for transport measurements, it is speculated that the observed findings are related to the transport function of the apical membrane of these polarized cells.


Asunto(s)
Proteínas de la Membrana , Proteínas de Transporte de Membrana , Epitelio Pigmentado Ocular/metabolismo , Receptores de Superficie Celular , Tetrahidrofolatos/farmacocinética , Proteínas Portadoras/metabolismo , Células Cultivadas , Receptores de Folato Anclados a GPI , Humanos , Epitelio Pigmentado Ocular/citología , Proteína Portadora de Folato Reducido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA