Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Physiol ; 597(2): 561-582, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30382595

RESUMEN

KEY POINTS: Gain-of-function mutations in the highly selective Ca2+ channel ORAI1 cause tubular aggregate myopathy (TAM) characterized by muscular pain, weakness and cramping. TAM-associated mutations in ORAI1 first and third transmembrane domain facilitate channel opening by STIM1, causing constitutive Ca2+ influx and increasing the currents evoked by Ca2+ store depletion. Mutation V107M additionally decreases the channel selectivity for Ca2+ ions and its inhibition by acidic pH, while mutation T184M does not alter the channel sensitivity to pH or to reactive oxygen species. The ORAI blocker GSK-7975A prevents the constitutive activity of TAM-associated channels and might be used in therapy for patients suffering from TAM. ABSTRACT: Skeletal muscle differentiation relies on store-operated Ca2+ entry (SOCE) mediated by STIM proteins linking the depletion of endoplasmic/sarcoplasmic reticulum Ca2+ stores to the activation of membrane Ca2+ -permeable ORAI channels. Gain-of-function mutations in STIM1 or ORAI1 isoforms cause tubular aggregate myopathy (TAM), a skeletal muscle disorder with muscular pain, weakness and cramping. Here, we characterize two overactive ORAI1 mutants from patients with TAM: V107M and T184M, located in the first and third transmembrane domain of the channel. When ectopically expressed in HEK-293T cells or human primary myoblasts, the mutated channels increased basal and store-operated Ca2+ entry. The constitutive activity of V107M, L138F, T184M and P245L mutants was prevented by low concentrations of GSK-7975A while the G98S mutant was resistant to inhibition. Electrophysiological recordings confirmed ORAI1-V107M constitutive activity and revealed larger STIM1-gated V107M- and T184M-mediated currents with conserved fast and slow Ca2+ -dependent inactivation. Mutation V107M altered the channel selectivity for Ca2+ ions and conferred resistance to acidic inhibition. Ca2+ imaging and molecular dynamics simulations showed a preserved sensitivity of T184M to the negative regulation by reactive oxygen species. Both mutants were able to mediate SOCE in Stim1-/- /Stim2-/- mouse embryonic fibroblasts expressing the binding-deficient STIM1-F394H mutant, indicating a higher sensitivity for STIM1-mediated gating, with ORAI1-T184M gain-of-function being strictly dependent on STIM1. These findings provide new insights into the permeation and regulatory properties of ORAI1 mutants that might translate into therapies against diseases with gain-of-function mutations in ORAI1.


Asunto(s)
Activación del Canal Iónico , Miopatías Estructurales Congénitas/genética , Proteína ORAI1/genética , Animales , Benzamidas/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Señalización del Calcio , Fibroblastos/fisiología , Mutación con Ganancia de Función , Células HEK293 , Humanos , Ratones Noqueados , Mioblastos/fisiología , Miopatías Estructurales Congénitas/fisiopatología , Proteína ORAI1/antagonistas & inhibidores , Proteína ORAI1/química , Proteína ORAI1/fisiología , Dominios Proteicos , Pirazoles/farmacología , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 2/genética
2.
Science ; 291(5509): 1755-9, 2001 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-11230685

RESUMEN

The ability of intestinal mucosa to absorb dietary ferric iron is attributed to the presence of a brush-border membrane reductase activity that displays adaptive responses to iron status. We have isolated a complementary DNA, Dcytb (for duodenal cytochrome b), which encoded a putative plasma membrane di-heme protein in mouse duodenal mucosa. Dcytb shared between 45 and 50% similarity to the cytochrome b561 family of plasma membrane reductases, was highly expressed in the brush-border membrane of duodenal enterocytes, and induced ferric reductase activity when expressed in Xenopus oocytes and cultured cells. Duodenal expression levels of Dcytb messenger RNA and protein were regulated by changes in physiological modulators of iron absorption. Thus, Dcytb provides an important element in the iron absorption pathway.


Asunto(s)
Grupo Citocromo b/metabolismo , Duodeno/metabolismo , Compuestos Férricos/metabolismo , Absorción Intestinal , Mucosa Intestinal/metabolismo , Hierro de la Dieta/metabolismo , Oxidorreductasas/metabolismo , Transfección , Secuencia de Aminoácidos , Anemia/enzimología , Animales , Línea Celular , Clonación Molecular , Grupo Citocromo b/química , Grupo Citocromo b/genética , ADN Complementario , Duodeno/enzimología , Enterocitos/enzimología , Enterocitos/metabolismo , Inducción Enzimática , Hipoxia , Mucosa Intestinal/enzimología , Hierro de la Dieta/administración & dosificación , Masculino , Ratones , Microvellosidades/enzimología , Microvellosidades/metabolismo , Datos de Secuencia Molecular , Nitroazul de Tetrazolio/metabolismo , Oocitos , Oxidación-Reducción , Oxidorreductasas/química , Oxidorreductasas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Sales de Tetrazolio/metabolismo , Tiazoles/metabolismo , Regulación hacia Arriba , Xenopus
3.
Neuron ; 16(3): 675-86, 1996 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8785064

RESUMEN

Three glutamate transporters have been identified in rat, including astroglial transporters GLAST and GLT-1 and a neuronal transporter EAAC1. Here we demonstrate that inhibition of the synthesis of each glutamate transporter subtype using chronic antisense oligonucleotide administration, in vitro and in vivo, selectively and specifically reduced the protein expression and function of glutamate transporters. The loss of glial glutamate transporters GLAST or GLT-1 produced elevated extracellular glutamate levels, neurodegeneration characteristic of excitotoxicity, and a progressive paralysis. The loss of the neuronal glutamate transporter EAAC1 did not elevate extracellular glutamate in the striatum but did produce mild neurotoxicity and resulted in epilepsy. These studies suggest that glial glutamate transporters provide the majority of functional glutamate transport and are essential for maintaining low extracellular glutamate and for preventing chronic glutamate neurotoxicity.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Astrocitos/metabolismo , Glutamatos/metabolismo , Oligonucleótidos Antisentido/metabolismo , Médula Espinal/metabolismo , Sistema de Transporte de Aminoácidos X-AG , Animales , Células Cultivadas , Expresión Génica , Hipocampo/metabolismo , Inmunohistoquímica , Masculino , Microscopía Electrónica , Ratas , Ratas Sprague-Dawley
4.
J Inherit Metab Dis ; 31(2): 178-87, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18415698

RESUMEN

In the late 1990s, the identification of transporters and transporter-associated genes progressed substantially due to the development of new cloning approaches such as expression cloning and, subsequently, to the implementation of the human genome project. Since then, the role of many transporter genes in human diseases has been elucidated. In this overview, we focus on inherited disorders of epithelial transporters. In particular, we review genetic defects of the genes encoding glucose transporters (SLC2 and SLC5 families) and amino acid transporters (SLC1, SLC3, SLC6 and SLC7 families).


Asunto(s)
Trastornos Innatos del Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/genética , Errores Innatos del Metabolismo de los Carbohidratos/genética , Células Epiteliales/metabolismo , Proteínas de Transporte de Monosacáridos/genética , Trastornos Innatos del Transporte de Aminoácidos/metabolismo , Sistemas de Transporte de Aminoácidos/metabolismo , Aminoácidos/metabolismo , Animales , Errores Innatos del Metabolismo de los Carbohidratos/metabolismo , Predisposición Genética a la Enfermedad , Glucosa/metabolismo , Humanos , Proteínas de Transporte de Monosacáridos/metabolismo , Fenotipo , Proteínas de Transporte de Sodio-Glucosa/genética , Proteínas de Transporte de Sodio-Glucosa/metabolismo
5.
Nat Neurosci ; 2(5): 427-33, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10321246

RESUMEN

The mechanism by which Cu2+/Zn2+ superoxide dismutase (SOD1) mutants lead to motor neuron degeneration in familial amyotrophic lateral sclerosis (FALS) is unknown. We show that oxidative reactions triggered by hydrogen peroxide and catalyzed by A4V and I113T mutant but not wild-type SOD1 inactivated the glutamate transporter human GLT1. Chelation of the copper ion of the prosthetic group of A4V prevented GLT1 inhibition. GLT1 was a selective target of oxidation mediated by SOD1 mutants, and its reactivity was confined to the intracellular carboxyl-terminal domain. The antioxidant Mn(III)TBAP rescued GLT1 from inhibition. Because inactivation of GLT1 results in neuronal degeneration, we propose that toxic properties of SOD1 mutants lead to neuronal death via an excitotoxic mechanism in SOD1-linked FALS.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Esclerosis Amiotrófica Lateral/genética , Neuroglía/metabolismo , Superóxido Dismutasa/genética , Sistema de Transporte de Aminoácidos X-AG , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Transporte Biológico/fisiología , Humanos , Mutación , Oocitos/metabolismo , Superóxido Dismutasa-1 , Xenopus
6.
J Clin Invest ; 98(11): 2580-7, 1996 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-8958221

RESUMEN

Absorption of urea in the renal inner medullary collecting duct (IMCD) contributes to hypertonicity in the medullary interstitium which, in turn, provides the osmotic driving force for water reabsorption. This mechanism is regulated by vasopressin via a cAMP-dependent pathway and activation of a specialized urea transporter located in the apical membrane. We report here the cloning of a novel urea transporter, designated UT1, from the rat inner medulla which is functionally and structurally distinct from the previously reported kidney urea transporter UT2. UT1 expressed in Xenopus oocytes mediated passive transport of urea that was inhibited by phloretin and urea analogs but, in contrast to UT2, was strongly stimulated by cAMP agonists. Sequence comparison revealed that the coding region of UT1 cDNA contains the entire 397 amino acid residue coding region of UT2 and an additional 1,596 basepair-stretch at the 5' end. This stretch encodes a novel 532 amino acid residue NH2-terminal domain that has 67% sequence identity with UT2. Thus, UT1 consists of two internally homologous portions that have most likely arisen by gene duplication. Studies of the rat genomic DNA further indicated that UT1 and UT2 are derived from a single gene by alternative splicing. Based on Northern analysis and in situ hybridization, UT1 is expressed exclusively in the IMCD, particularly in its terminal portion. Taken together, our data show that UT1 corresponds to the previously characterized vasopressin-regulated urea transporter in the apical membrane of the terminal IMCD which plays a critical role in renal water conservation.


Asunto(s)
Proteínas Portadoras/biosíntesis , Proteínas Portadoras/química , Médula Renal/metabolismo , Túbulos Renales Colectores/metabolismo , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/química , Proteínas de Transporte de Membrana , Transcripción Genética , Vasopresinas/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas Portadoras/fisiología , Clonación Molecular , Secuencia Conservada , Cisteína , Sondas de ADN , Exones , Biblioteca de Genes , Hibridación in Situ , Médula Renal/fisiología , Túbulos Renales Colectores/fisiología , Glicoproteínas de Membrana/fisiología , Datos de Secuencia Molecular , Especificidad de Órganos , Estructura Secundaria de Proteína , ARN Mensajero/biosíntesis , Ratas , Homología de Secuencia de Aminoácido , Urea/metabolismo , Transportadores de Urea
7.
J Clin Invest ; 93(1): 397-404, 1994 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8282810

RESUMEN

The major reabsorptive mechanism for D-glucose in the kidney is known to involve a low affinity high capacity Na+/glucose cotransporter, which is located in the early proximal convoluted tubule segment S1, and which has a Na+ to glucose coupling ratio of 1:1. Here we provide the first molecular evidence for this renal D-glucose reabsorptive mechanism. We report the characterization of a previously cloned human kidney cDNA that codes for a protein with 59% identity to the high affinity Na+/glucose cotransporter (SGLT1). Using expression studies with Xenopus laevis oocytes we demonstrate that this protein (termed SGLT2) mediates saturable Na(+)-dependent and phlorizin-sensitive transport of D-glucose and alpha-methyl-D-glucopyranoside (alpha MeGlc) with Km values of 1.6 mM for alpha MeGlc and approximately 250 to 300 mM for Na+, consistent with low affinity Na+/glucose cotransport. In contrast to SGLT1, SGLT2 does not transport D-galactose. By comparing the initial rate of [14C]-alpha MeGlc uptake with the Na(+)-influx calculated from alpha MeGlc-evoked inward currents, we show that the Na+ to glucose coupling ratio of SGLT2 is 1:1. Using combined in situ hybridization and immunocytochemistry with tubule segment specific marker antibodies, we demonstrate an extremely high level of SGLT2 message in proximal tubule S1 segments. This level of expression was also evident on Northern blots and likely confers the high capacity of this glucose transport system. We conclude that SGLT2 has properties characteristic of the renal low affinity high capacity Na+/glucose cotransporter as previously reported for perfused tubule preparations and brush border membrane vesicles. Knowledge of the structural and functional properties of this major renal Na+/glucose reabsorptive mechanism will advance our understanding of the pathophysiology of renal diseases such as familial renal glycosuria and diabetic renal disorders.


Asunto(s)
Glucosa/metabolismo , Túbulos Renales/metabolismo , Riñón/metabolismo , Proteínas de Transporte de Monosacáridos/metabolismo , Animales , Transporte Biológico , Línea Celular , Chlorocebus aethiops , Femenino , Transportador de Glucosa de Tipo 2 , Humanos , Hibridación in Situ , Cinética , Metilglucósidos/metabolismo , Proteínas de Transporte de Monosacáridos/biosíntesis , Oocitos/metabolismo , Oocitos/fisiología , ARN Complementario/metabolismo , Transfección , Xenopus laevis
8.
J Clin Invest ; 103(8): 1159-68, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10207168

RESUMEN

Kidney proximal tubule cells take up Krebs cycle intermediates for metabolic purposes and for secretion of organic anions through dicarboxylate/organic anion exchange. Alteration in reabsorption of citrate is closely related to renal stone formation. The presence of distinct types of sodium-coupled dicarboxylate transporters has been postulated on either side of the polarized epithelial membrane in the kidney proximal tubule. Using a PCR-based approach, we isolated a novel member of the sodium-dependent dicarboxylate/sulfate transporter called SDCT2. SDCT2 is a 600-amino acid residue protein that has 47-48% amino acid identity to SDCT1 and NaDC-1, previously identified in kidney and intestine. Northern analysis gave a single band of 3.3 kb for SDCT2 in kidney, liver, and brain. In situ hybridization revealed that SDCT2 is prominently expressed in kidney proximal tubule S3 segments and in perivenous hepatocytes, consistent with the sites of high-affinity dicarboxylate transport identified based on vesicle studies. A signal was also detected in the meningeal layers of the brain. SDCT2 expressed in Xenopus oocytes mediated sodium-dependent transport of di- and tricarboxylates with substrate preference for succinate rather than citrate, but excluding monocarboxylates. SDCT2, unlike SDCT1, displayed a unique pH dependence for succinate transport (optimal pH 7.5-8.5) and showed a high affinity for dimethylsuccinate, two features characteristic of basolateral transport. These data help to interpret the mechanisms of renal citrate transport, their alteration in pathophysiological conditions, and their role in the elimination of organic anions and therapeutic drugs.


Asunto(s)
Proteínas Portadoras/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas Portadoras/fisiología , ADN Complementario , Transportadores de Ácidos Dicarboxílicos , Humanos , Datos de Secuencia Molecular , Ratas , Homología de Secuencia de Aminoácido , Sodio , Distribución Tisular
9.
J Clin Invest ; 99(7): 1506-15, 1997 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-9119994

RESUMEN

Urea transport in the kidney plays an important role in urinary concentration and nitrogen balance. Recently, three types of urea transporters have been cloned, UT1 and UT2 from rat and rabbit kidney and HUT11 from human bone marrow. To elucidate the physiological role of the latter urea transporter, we have isolated the rat homologue (UT3) of HUT11 and studied its distribution of expression and functional characteristics. UT3 cDNA encodes a 384 amino acid residue protein, which has 80% identity to the human HUT11 and 62% identity to rat UT2. Functional expression in Xenopus oocytes induced a large (approximately 50-fold) increase in the uptake of urea compared with water-injected oocytes. The uptake was inhibited by phloretin (0.75 mM) and pCMBS (0.5 mM) (55 and 32% inhibition, respectively). Northern analysis gave a single band of 3.8 kb in kidney inner and outer medulla, testis, brain, bone marrow, spleen, thymus, and lung. In situ hybridization of rat kidney revealed that UT3 mRNA is expressed in the inner stripe of the outer medulla, inner medulla, the papillary surface epithelium, and the transitional urinary epithelium of urinary tracts. Co-staining experiments using antibody against von Willebrand factor showed that UT3 mRNA in the inner stripe of the outer medulla is expressed in descending vasa recta. These data suggest that UT3 in kidney is involved in counter current exchange between ascending and descending vasa recta, to enhance the cortico-papillary osmolality gradient. In situ hybridization of testis revealed that UT3 is located in Sertoli cells of seminiferous tubules. The signal was only detected in Sertoli cells associated with the early stages of spermatocyte development, suggesting that urea may play a role in spermatogenesis.


Asunto(s)
Proteínas Portadoras/genética , Riñón/química , Glicoproteínas de Membrana/genética , Proteínas de Transporte de Membrana , Testículo/química , Urea/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/análisis , Proteínas Portadoras/fisiología , Clonación Molecular , Masculino , Glicoproteínas de Membrana/análisis , Glicoproteínas de Membrana/fisiología , Datos de Secuencia Molecular , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Transportadores de Urea
10.
J Clin Invest ; 96(3): 1556-63, 1995 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-7657826

RESUMEN

In mammals, urea is the predominant end-product of nitrogen metabolism and plays a central role in the urinary-concentrating mechanism. Urea accumulation in the renal medulla is critical to the ability of the kidney to concentrate urine to an osmolality greater than systemic plasma. Regulation of urea excretion and accumulation in the renal medulla depends on the functional state of specialized phloretin-sensitive urea transporters. To study these transporters and their regulation of expression we isolated a cDNA which encodes the rat homologue (rUT2) of rabbit UT2 (You, G., C.P. Smith, Y. Kanai, W.-S. Lee, M. Stelzner, and M.A. Hediger, et al. Nature (Lond.). 1993. 365:844-847). Rat UT2 has 88% amino acid sequence identity to rabbit UT2 and 64% identity to the recently cloned human erythrocyte urea transporter, HUT11 (Olives, B., P. Neav, P. Bailly, M.A. Hediger, G. Rousselet, J.P. Cartron, and P. Ripoch J. Biol. Chem. 1994. 269:31649-31652). Analysis of rat kidney mRNA revealed two transcripts of size 2.9 and 4.0 kb which had spatially distinct distributions. Northern analysis and in situ hybridization showed that the 4.0-kb transcript was primarily responsive to changes in the protein content of the diet whereas the 2.9-kb transcript was responsive to changes in the hydration state of the animal. These studies reveal that the expression levels of the two rUT2 transcripts are modulated by different pathways to allow fluid and nitrogen balance to be regulated independently. Our data provide important insights into the regulation of the renal urea transporter UT2 and provide a basis on which to refine our understanding of the urinary concentrating mechanism and its regulation.


Asunto(s)
Proteínas Portadoras/biosíntesis , Proteínas en la Dieta , Regulación de la Expresión Génica , Riñón/fisiología , Glicoproteínas de Membrana/biosíntesis , Proteínas de Transporte de Membrana , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/química , Proteínas Portadoras/fisiología , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Clonación Molecular , ADN Complementario/metabolismo , Diuresis , Femenino , Hibridación in Situ , Riñón/citología , Médula Renal/metabolismo , Masculino , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/fisiología , Modelos Biológicos , Modelos Estructurales , Datos de Secuencia Molecular , Oocitos/fisiología , Estructura Secundaria de Proteína , Conejos , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Transcripción Genética , Urea/metabolismo , Xenopus laevis , Transportadores de Urea
11.
J Clin Invest ; 102(11): 2011-8, 1998 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-9835627

RESUMEN

Intestinal epithelial cells express hPepT1, an apical transporter responsible for the uptake of a broad array of small peptides. As these could conceivably include n-formyl peptides, we examined whether hPepT1 could transport the model n-formylated peptide fMLP and, if so, whether such cellular uptake of fMLP influenced neutrophil-epithelial interactions. fMLP uptake into oocytes was enhanced by hPepT1 expression. In addition, fMLP competitively inhibited uptake of a known hPepT1 substrate (glycylsarcosine) in hPepT1 expressing oocytes. hPepT1 peptide uptake was further examined in a polarized human intestinal epithelial cell line (Caco2-BBE) known to express this transporter. Epithelial monolayers internalized apical fMLP in a fashion that was competitively inhibited by other hPepT1 recognized solutes, but not by related solutes that were not transported by hPepT1. Fluorescence analyses of intracellular pH revealed that fMLP uptake was accompanied by cytosolic acidification, consistent with the known function of hPepT1 as a peptide H+ cotransporter. Lumenal fMLP resulted in directed movement of neutrophils across epithelial monolayers. Solutes that inhibit hPepT1-mediated fMLP transport decreased neutrophil transmigration by approximately 50%. Conversely, conditions that enhanced the rate of hPepT1-mediated fMLP uptake (cytosolic acidification) enhanced neutrophil-transepithelial migration by approximately 70%. We conclude that hPepT1 transports fMLP and uptake of these peptide influences neutrophil-epithelial interactions. These data (a) emphasize the importance of hPepT1 in mediating intestinal inflammation, (b) raise the possibility that modulating hPepT1 activity could influence states of intestinal inflammation, and (c) provide the first evidence of a link between active transepithelial transport and neutrophil-epithelial interactions.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Portadoras/fisiología , Factores Quimiotácticos/metabolismo , Células Epiteliales/efectos de los fármacos , Escherichia coli/metabolismo , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Simportadores , Animales , Calcio/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Neoplasias del Colon/patología , Dipéptidos/metabolismo , Células Epiteliales/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Transporte Iónico/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/fisiología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Transportador de Péptidos 1 , Células Tumorales Cultivadas , Xenopus laevis
12.
J Clin Invest ; 91(5): 1959-63, 1993 May.
Artículo en Inglés | MEDLINE | ID: mdl-8486766

RESUMEN

We have recently cloned, sequenced, and characterized a rat kidney cDNA (D2) that stimulates cystine as well as dibasic and neutral amino acid transport. In order to evaluate the role of this protein in human inherited diseases such as cystinuria, we have isolated a human D2 clone (D2H) by low stringency screening of a human kidney cDNA library using the radiolabeled D2 insert as a probe. The D2H cDNA is 2284 nucleotides long and encodes a 663 amino acid protein that is 80% identical to the rat D2 amino acid sequence and 86% to that of the rabbit homologue rBAT. Microinjection of in vitro transcribed D2H cRNA into Xenopus oocytes induced uptake of cystine as well as dibasic and neutral amino acids in a pattern similar to that of rat D2 and rabbit rBAT. Both neutral and dibasic amino acids inhibited the D2H-induced uptake of cystine. Northern blot analysis demonstrated that D2H, like D2 and rBAT, is expressed strongly in the kidney and intestine. Southern blot analysis of genomic DNA from a panel of mouse-human somatic cell hybrids showed that the human gene for D2H resides on chromosome 2.


Asunto(s)
Aminoácidos Diaminos/metabolismo , Aminoácidos/metabolismo , Proteínas Portadoras/genética , Cromosomas Humanos Par 2 , Cistina/metabolismo , ADN/genética , Riñón/metabolismo , Secuencia de Aminoácidos , Sistemas de Transporte de Aminoácidos , Animales , Transporte Biológico , Proteínas Portadoras/metabolismo , Mapeo Cromosómico , Clonación Molecular/métodos , Cricetinae , Femenino , Biblioteca de Genes , Humanos , Células Híbridas , Cinética , Datos de Secuencia Molecular , Oocitos/metabolismo , Conejos , Ratas , Homología de Secuencia de Aminoácido , Transcripción Genética , Xenopus
13.
J Clin Invest ; 93(2): 578-85, 1994 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8113395

RESUMEN

The effect of insulinopenic diabetes on the expression of glucose transporters in the small intestine was investigated. Enterocytes were sequentially isolated from jejunum and ileum of normal fed rats, streptozotocin-diabetic rats, and diabetic rats treated with insulin. Facilitative glucose transporter (GLUT) 2, GLUT5, and sodium-dependent glucose transporter 1 protein content was increased from 1.5- to 6-fold in enterocytes isolated from diabetic animals in both jejunum and ileum. Insulin was able to reverse the increase in transporter protein expression seen after induction of diabetes. There was a four- to eightfold increase in the amount of enterocyte glucose transporter mRNA after diabetes with greater changes in sodium-dependent glucose transporter 1 and GLUT2 than in GLUT5 levels. In situ hybridization showed that after the induction of diabetes there was new hybridization in lower villus and crypt enterocytes that was reversed by insulin treatment. Thus, the increase in total hexose transport caused by diabetes is due to a premature expression of hexose transporters by enterocytes along the crypt-villus axis, causing a cumulative increase in enterocyte transporter protein during maturation. These changes are likely to represent an adaptive response by the organism to increase nutrient absorption in a perceived state of tissue starvation. These adaptive changes may lead to exacerbation of hyperglycemia in uncontrolled diabetes.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Íleon/metabolismo , Mucosa Intestinal/metabolismo , Yeyuno/metabolismo , Proteínas de Transporte de Monosacáridos/biosíntesis , Proteínas de Transporte de Monosacáridos/metabolismo , ARN Mensajero/biosíntesis , Secuencia de Aminoácidos , Animales , Western Blotting , Membrana Celular/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Expresión Génica/efectos de los fármacos , Transportador de Glucosa de Tipo 1 , Transportador de Glucosa de Tipo 2 , Transportador de Glucosa de Tipo 5 , Hibridación in Situ , Insulina/farmacología , Insulina/uso terapéutico , Masculino , Datos de Secuencia Molecular , Proteínas de Transporte de Monosacáridos/análisis , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Valores de Referencia
14.
Trends Neurosci ; 16(9): 365-70, 1993 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-7694407

RESUMEN

Removal of glutamate from the synaptic cleft is an essential component of the transmission process at glutamatergic synapses. This requirement is fulfilled by transporters that have a high affinity for glutamate and exhibit a unique coupling to Na+, K+ and OH- ions. Independently, three groups have succeeded in cloning cDNAs encoding high-affinity Na(+)-dependent glutamate transporters. These transporters are structurally distinct from previously characterized neurotransmitter transporters and show sequence identity with prokaryotic glutamate and dicarboxylate transporters. In addition, they exhibit significant differences in their structure, function and tissue distribution. This review compares and contrasts these differences, and incorporates into the existing body of knowledge these new breakthroughs.


Asunto(s)
Glutamatos/fisiología , Glicoproteínas/metabolismo , Sistema de Transporte de Aminoácidos X-AG , Animales , Transporte Biológico/fisiología , Ácido Glutámico , Humanos , Transmisión Sináptica/fisiología
16.
Biochim Biophys Acta ; 1064(2): 360-4, 1991 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-1903656

RESUMEN

The initial stages in the biosynthesis of the cloned Na+/glucose cotransporter were examined by in vitro expression of the protein in the absence and presence of pancreatic microsomes. Glycosylation was detected by endoglycosidase-H shifts in the apparent size of the proteins on SDS-PAGE. In the presence of microsomes, Mr increased from 52,000 to 58,000, and this was reversed by endo-H. This demonstrates that the protein is glycosylated and that there is no large cleavable signal sequence. Using partial transcripts and site-directed mutagenesis, we established that Asn-248 is glycosylated. Glycosylation was not required for the functional expression of the transporter in Xenopus oocytes. In terms of the topology of the protein, these results suggest that Asn-248 is on the external surface of the membrane.


Asunto(s)
Mucosa Intestinal/metabolismo , Proteínas de Transporte de Monosacáridos/biosíntesis , Acetilglucosaminidasa/farmacología , Animales , Secuencia de Bases , Células Cultivadas , Clonación Molecular , ADN/química , Expresión Génica , Glicosilación , Intestinos/efectos de los fármacos , Manosil-Glicoproteína Endo-beta-N-Acetilglucosaminidasa , Datos de Secuencia Molecular , Peso Molecular , Mutación , Oocitos/metabolismo , Conformación Proteica , Conejos , Xenopus
17.
Biochim Biophys Acta ; 1048(1): 100-4, 1990 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-2105101

RESUMEN

Cells derived from the simian kidney, COS-7 cells, were transfected with a eucaryotic expression vector (pEUK-C1) containing the clone for the rabbit intestinal Na+/glucose cotransporter. Expression was monitored after transfection with lipofectin by measuring the initial rate of alpha-methylglucopyranoside (MeGlc) uptake. Cells transfected with vector containing the cDNA for the Na+/glucose cotransporter expressed Na(+)-dependent MeGlc transport. Neither control cells nor cells transfected with vector lacking cloned cDNA expressed the cotransporter. Na(+)-dependent MeGlc uptake into transfected cells was saturable (Km 150 microM), phlorizin-sensitive (Ki 11 microM), and inhibited by sugar analogs (D-glucose greater than MeGlc greater than D-galactose greater than 3-O-methyl-D-glucoside greater than D-allose much greater than L-glucose). Europium was able to mimic Na+ in driving MeGIC uptake. Finally, tunicamycin, an inhibitor of asparagine-linked glycosylation, inhibited the expression of Na(+)-dependent MeGlc transport 80%. We conclude that the rabbit intestinal Na+/glucose cotransporter expressed in COS-7 cell exhibits very similar kinetic properties to that in the native brush border and to that expressed in Xenopus oocytes. In addition, N-linked glycosylation appears to be important for functional expression of this membrane protein.


Asunto(s)
Expresión Génica , Mucosa Intestinal/metabolismo , Proteínas de Transporte de Monosacáridos/genética , Animales , Metabolismo de los Hidratos de Carbono , Línea Celular , Glicosilación , Haplorrinos , Iones , Cinética , Plásmidos , Transfección
18.
Biochim Biophys Acta ; 1235(2): 461-6, 1995 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-7756356

RESUMEN

Mammalian kidney is known to express a transport system specific for small peptides and pharmacologically active aminocephalosporins. This system is energized by a transmembrane electrochemical H+ gradient. Recently, a H(+)-coupled peptide transporter has been cloned from rabbit and human intestine (Fei et al. (1994) Nature 368, 563-566; Liang et al., J. Biol. Chem., in press). Functional studies have established that the renal peptide transport system is similar but not identical to its intestinal counterpart. Therefore, in an attempt to isolate the renal H+/peptide cotransporter cDNA, we screened a human kidney cDNA library with a probe derived from the rabbit intestinal H+/peptide cotransporter cDNA. This has resulted in the isolation of a positive clone with a 2190 bp long open reading frame. The predicted protein consists of 729 amino acids. Hydropathy analysis of the amino acid sequence indicates the presence of twelve putative transmembrane domains. The primary structure of this protein exhibits 50% identity and 70% similarity to the human intestinal H+/peptide cotransporter. Functional expression of the kidney cDNA in HeLa cells results in the induction of a H(+)-coupled transport system specific for small peptides and aminocephalosporins. Reverse transcription-coupled polymerase chain reaction demonstrates that the cloned transporter is expressed in human kidney but not in human intestine. This transporter, henceforth called PEPT 2, represents a new member in the growing family of H(+)-coupled transport systems in the mammalian plasma membrane.


Asunto(s)
Proteínas Portadoras/genética , Clonación Molecular , Riñón/química , Simportadores , Secuencia de Aminoácidos , Secuencia de Bases , Northern Blotting , Proteínas Portadoras/química , ADN Complementario/aislamiento & purificación , Expresión Génica , Células HeLa , Humanos , Intestino Delgado/química , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , Análisis de Secuencia , Transfección
19.
Endocrinology ; 132(5): 2136-40, 1993 May.
Artículo en Inglés | MEDLINE | ID: mdl-8477661

RESUMEN

A complementary DNA clone encoding the type I iodothyronine 5'-deiodinase (5'DI), which converts T4 to T3, has been isolated recently. The 5'DI messenger RNA (mRNA) is most abundant in kidney and liver. To gain insight into the function of 5'DI in the kidney, Northern blot analysis was used to localize the expression of this mRNA. Our results show that 5'DI mRNA was expressed in the cortex and outer medulla, but not inner medulla and papilla, indicating that there are regional differences in its expression. Using in situ hybridization, we demonstrate that the 5'DI hybridization signal is localized predominantly over tubules in the outer stripe of the outer medulla and in medullary rays, suggesting localization to proximal tubules. To identify which tubular cells express 5'DI mRNA, we compared the profile of 5'DI message from in situ hybridization with the immunostaining of adjacent sections with proximal tubular S1, S2, and S3 segment-specific antibodies. Most of the 5'DI antisense complementary RNA hybridized to the same proximal tubular cells with which the S3-specific antibody, anti-ecto-ATPase, reacted. Cells staining with an S1 or S2 segment antibody showed little, if any, 5'DI mRNA. We conclude that the expression of 5'DI mRNA is restricted to the tubular cells of the proximal S3 segment. The S3 segment is also known to express high levels of proteins required for glutathione synthesis consistent with the requirement for a reduced thiol cofactor for iodothyronine deiodination by the 5'DI pathway.


Asunto(s)
Yoduro Peroxidasa/genética , Túbulos Renales Proximales/química , ARN Mensajero/análisis , Animales , Northern Blotting , Inmunohistoquímica , Hibridación in Situ , Corteza Renal/química , Médula Renal/química , Masculino , Sondas ARN , Ratas , Distribución Tisular
20.
FEBS Lett ; 509(2): 309-16, 2001 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-11741608

RESUMEN

The first step in intestinal iron absorption is mediated by the H(+)-coupled Fe(2+) transporter called divalent cation transporter 1/divalent metal ion transporter 1 (DCT1/DMT1) (also known as natural resistance-associated macrophage protein 2). DCT1/DMT1 mRNA levels in the duodenum strongly increase in response to iron depletion. To study the mechanism of iron-dependent DCT1/DMT1 mRNA regulation, we investigated the endogenous expression of DCT1/DMT1 mRNA in various cell types. We found that only the iron responsive element (IRE)-containing form, which corresponds to one of two splice forms of DCT1/DMT1, is responsive to iron treatment and this responsiveness was cell type specific. We also examined the interaction of the putative 3'-UTR IRE with iron responsive binding proteins (IRP1 and IRP2), and found that IRP1 binds to the DCT1/DMT1-IRE with higher affinity compared to IRP2. This differential binding of IRP1 and IRP2 was also reported for the IREs of transferrin receptors, erythroid 5-aminolevulinate synthase and mitochondrial aconitase. We propose that regulation of DCT1/DMT1 mRNA by iron involves post-transcriptional regulation through the binding of IRP1 to the transporter's IRE, as well as other as yet unknown factors.


Asunto(s)
Proteínas de Transporte de Catión/genética , Proteínas de Unión a Hierro , Hierro/metabolismo , Regiones no Traducidas 3' , Transporte Biológico , Células CACO-2 , Proteínas de Transporte de Catión/metabolismo , Cationes Bivalentes/metabolismo , Regulación de la Expresión Génica , Células HeLa , Humanos , Proteína 1 Reguladora de Hierro , Proteína 2 Reguladora de Hierro , Proteínas Reguladoras del Hierro , Proteínas Hierro-Azufre/metabolismo , Conformación de Ácido Nucleico , Unión Proteica , Procesamiento Postranscripcional del ARN , Proteínas de Unión al ARN/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA