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











Base de datos
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.
Mol Aspects Med ; 34(2-3): 299-312, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23506872

RESUMEN

The SLC13 family comprises five genes (SLC13A1, SLC13A2, SLC13A3, SLC13A4, and SLC13A5) encoding structurally related multi-spanning transporters (8-13 transmembrane domains) with orthologues found in prokaryotes and eukaryotes. Mammalian SLC13 members mediate the electrogenic Na(+)-coupled anion cotransport at the plasma membrane of epithelial cells (mainly kidney, small intestine, placenta and liver) or cells of the central nervous system. While the two SLC13 cotransporters NaS1 (SLC13A1) and NaS2 (SLC13A4) transport anions such sulfate, selenate and thiosulfate, the three other SLC13 members, NaDC1 (SLC13A2), NaCT (SLC13A5) and NaDC3 (SLC13A3), transport di- and tri-carboxylate Krebs cycle intermediates such as succinate, citrate and α-ketoglutarate. All these transporters play a variety of physiological and pathophysiological roles in the different organs. Thus, the purpose of this review is to summarize the roles of SLC13 members in human physiology and pathophysiology and what the therapeutic perspectives are. We have also described the most recent advances on the structure, expression, function and regulation of SLC13 transporters.


Asunto(s)
Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/fisiología , Células Epiteliales/metabolismo , Modelos Moleculares , Familia de Multigenes/genética , Conformación Proteica , Simportadores/genética , Simportadores/fisiología , Proteínas de Transporte de Catión/metabolismo , Ácidos Dicarboxílicos/metabolismo , Humanos , Modelos Biológicos , Cotransportador de Sodio-Sulfato , Simportadores/metabolismo , Ácidos Tricarboxílicos/metabolismo
3.
Mol Aspects Med ; 34(2-3): 735-41, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23506906

RESUMEN

Prostate cancer is a major health concern as it has the second highest incidence rate among cancers in men. Despite progress in tumor diagnostics and therapeutic approaches, prognosis for men with advanced disease remains poor. In this review we provide insight into the changes of the intermediary metabolism in normal prostate and prostate cancer. In contrast to normal cells, prostate cancer cells are reprogrammed for optimal energy-efficiency with a functional Krebs cycle and minimal apoptosis rates. A key element in this relationship is the uniquely high zinc level of normal prostate epithelial cells. Zinc is transported by the SLC30 and SLC39 families of zinc transporters. However, in prostate cancer the intracellular zinc content is remarkably reduced and expression levels of certain zinc transporters are altered. Here, we summarize the role of different zinc transporters in the development of prostate cancer.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Ciclo del Ácido Cítrico/fisiología , Metabolismo Energético/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Neoplasias de la Próstata/metabolismo , Zinc/metabolismo , Humanos , Masculino , Modelos Biológicos
4.
Bone ; 47(2): 301-8, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20399919

RESUMEN

Energy-dependent intestinal calcium absorption is important for the maintenance of calcium and bone homeostasis, especially when dietary calcium supply is restricted. The active form of vitamin D, 1,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)], is a crucial regulator of this process and increases the expression of the transient receptor potential vanilloid 6 (Trpv6) calcium channel that mediates calcium transfer across the intestinal apical membrane. Genetic inactivation of Trpv6 in mice (Trpv6(-/-)) showed, however, that TRPV6 is redundant for intestinal calcium absorption when dietary calcium content is normal/high and passive diffusion likely contributes to maintain normal serum calcium levels. On the other hand, Trpv6 inactivation impaired the increase in intestinal calcium transport following calcium restriction, however without resulting in hypocalcemia. A possible explanation is that normocalcemia is maintained at the expense of bone homeostasis, a hypothesis investigated in this study. In this study, we thoroughly analyzed the bone phenotype of Trpv6(-/-) mice receiving a normal (approximately 1%) or low (approximately 0.02%) calcium diet from weaning onwards using micro-computed tomography, histomorphometry and serum parameters. When dietary supply of calcium is normal, Trpv6 inactivation did not affect growth plate morphology, bone mass and remodeling parameters in young adult or aging mice. Restricting dietary calcium had no effect on serum calcium levels and resulted in a comparable reduction in bone mass accrual in Trpv6(+/+) and Trpv6(-/-) mice (-35% and 45% respectively). This decrease in bone mass was associated with a similar increase in bone resorption, whereas serum osteocalcin levels and the amount of unmineralized bone matrix were only significantly increased in Trpv6(-/-) mice. Taken together, our findings indicate that TRPV6 contributes to intestinal calcium transport when dietary calcium supply is limited and in this condition indirectly regulates bone formation and/or mineralization.


Asunto(s)
Huesos/efectos de los fármacos , Huesos/metabolismo , Canales de Calcio/metabolismo , Calcio de la Dieta/farmacología , Calcio/metabolismo , Homeostasis/efectos de los fármacos , Absorción Intestinal/efectos de los fármacos , Canales Catiónicos TRPV/metabolismo , Envejecimiento/efectos de los fármacos , Envejecimiento/patología , Animales , Remodelación Ósea/efectos de los fármacos , Calcio/sangre , Canales de Calcio/deficiencia , Canales de Calcio/genética , Duodeno/efectos de los fármacos , Duodeno/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Activación del Canal Iónico/efectos de los fármacos , Ratones , Tamaño de los Órganos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Fosfatos/sangre , ARN Mensajero/genética , ARN Mensajero/metabolismo , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/genética
5.
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
6.
Ther Umsch ; 61(9): 541-5, 2004 Sep.
Artículo en Alemán | MEDLINE | ID: mdl-15493112

RESUMEN

In humans, uric acid is the final breakdown product of unwanted purine nucleotides. Uric acid is the last stage in purine degradation, because humans lack the enzyme uricase which converts uric acid into allantoin. Uric acid has profound beneficial effects since it scavenges potential harmful radicals in our body. However, in conjunction with genetic or environmental factors, uric acid can cause significant health problems, leading to kidney stones when it builds up in the kidneys and to gout when crystals accumulate in the joints. The levels of uric acid in the blood must be tightly controlled to minimize these detrimental effects. Normally, the body eliminates enough uric acid in the kidney, and in part also through the intestines, to keep its concentration at a healthy level in the blood (approximately 300 microM). In patients with gout or kidney stone disease, however, the body either produces excessive amounts of uric acid or its ability to eliminate uric acid is disturbed in some way. In the kidney, uric acid is reabsorbed via the uric acid transporter URAT1. This transporter is the major mechanism for regulating blood uric acid levels and therefore may prove an interesting target for future drug development.


Asunto(s)
Proteínas Portadoras/fisiología , Gota/fisiopatología , Cálculos Renales/fisiopatología , Transportadores de Anión Orgánico/fisiología , Urato Oxidasa/fisiología , Ácido Úrico/sangre , Animales , Aves , Tasa de Filtración Glomerular/fisiología , Humanos , Proteínas de Transporte de Catión Orgánico , Filogenia , Primates
7.
J Membr Biol ; 197(2): 91-9, 2004 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-15014911

RESUMEN

Iron(II) exacerbates the effects of oxidative stress via the Fenton reaction. A number of human diseases are associated with iron accumulation including ischemia-reperfusion injury, inflammation and certain neurodegenerative diseases. The functional properties and localization in plasma membrane of cells and endosomes suggest an important role for the divalent metal transporter DMT1 (also known as DCT1 and Nramp2) in iron transport and cellular iron homeostasis. Although iron metabolism is strictly controlled and the activity of DMT1 is central in controlling iron homeostasis, no regulatory mechanisms for DMT1 have been so far identified. Our studies show that the activity of DMT1 is modulated by compounds that affect its redox status. We also show that both iron and zinc are transported by DMT1 when expressed in Xenopus laevis oocytes. Radiotracer uptake and electrophysiological measurements revealed that H(2)O(2) and Hg(2+) treatments result in substantial inhibition of DMT1. These findings may have a profound relevance from a physiological and pathophysiological standpoint.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Hierro/metabolismo , Estrés Oxidativo/fisiología , Zinc/metabolismo , Animales , Transporte Biológico Activo/efectos de los fármacos , Proteínas de Transporte de Catión/antagonistas & inhibidores , Electrofisiología , Humanos , Peróxido de Hidrógeno/toxicidad , Cloruro de Mercurio/toxicidad , Oocitos , Oxidación-Reducción , Xenopus
8.
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
9.
J Membr Biol ; 184(2): 113-20, 2001 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-11719848

RESUMEN

The human epithelial channels, CaT1 and CaT2, were expressed in oocytes, and their single-channel characteristics were compared. In the presence of Na+ and K+ as charge carriers in the pipette solutions, channel activities were observed only when the the extracellular sides of the patches were exposed to nominally Ca2+- and Mg2+-free solutions. In patches of both CaT1- and CaT2-expressing oocytes, multiple channel openings were observed, but the current levels were higher in CaT2-expressing oocytes, particularly at more negative voltages. With K+ as a charge carrier in patches of CaT1-expressing oocytes, the channel activity was low at -10 to -60 mV, but increased dramatically at more negative potentials. This voltage dependence was observed in the presence of both Na+ and K+. The channel activity with Na+, however, was higher at all potentials. Differences between the voltage dependencies for the two cations were also observed in CaT2-expressing oocytes, but the channel activities were higher than those in CaT1-expressing oocytes, particularly in the presence of Na+. We also found that low concentrations of extracellular Mg2+ (5-50 microm) elicited a strong inhibitory action on the CaT channels. Activation of the CaT1 and CaT2 channels by hyperpolarization and other factors may promote increased Ca2+ entry that participates in stimulation of intestinal absorption and renal reabsorption and/or other Ca2+ transport mechanisms in epithelial cells.


Asunto(s)
Canales de Calcio/fisiología , Calcio/metabolismo , Sistemas de Transporte de Aminoácidos Básicos , Animales , Canales de Calcio/efectos de los fármacos , Canales de Calcio/genética , Células Epiteliales/fisiología , Humanos , Magnesio/farmacología , Potenciales de la Membrana/fisiología , Oocitos/metabolismo , Potasio/metabolismo , ARN Complementario/genética , Ratas , Sodio/metabolismo , Canales Catiónicos TRPV , Transfección , Xenopus laevis
10.
Brain Res ; 914(1-2): 196-203, 2001 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-11578612

RESUMEN

Recent evidence indicates that second messengers and protein kinases regulate the activity and expression of glutamate transporters. The aim of the present study was to determine if direct activation of protein kinases C or A modulates the activity of the sodium-dependent glutamate transporter EAAC1. EAAC1 modulation was studied in cRNA-injected Xenopus oocytes by measuring [3H]L-glutamate uptake or glutamate-evoked uptake currents. We found that activation of PKA was ineffective, whereas treatment with the PKC agonist phorbol 12-myristate 13-acetate (PMA) caused a significant decrease in EAAC1 transport activity (IC(50)=44.7+/-12 nM). PMA-induced EAAC1 inhibition was PKC-mediated because the inhibition could be blocked by specific PKC inhibitors and incubation with the inactive 4alpha-phorbol-12,13-didecanoate (4alpha-PDD) did not affect EAAC1. Saturation studies of glutamate-evoked uptake currents showed that PMA-mediated inhibition was due to a decrease in I(max) with no change in K(m). PMA simultaneously decreased membrane capacitance (C(m)) and transport-associated current and increased cytosolic accumulation of EAAC1 protein, compared to control. These results suggest that PKC activation inhibits EAAC1 by promoting its retrieval from the plasma membrane. PMA also significantly decreased glutamate uptake in a Madin-Darby canine kidney (MDCK) cell line stably transfected with EAAC1 but enhanced EAAC1-mediated glutamate uptake in the rat C6 glioma cells, consistent with previous observations. Because activation of PKC by phorbol esters leads to opposite effects on EAAC1 activity in different culture models, we conclude that the PKC-mediated regulation of EAAC1 is cell-type specific.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG , Carcinógenos/farmacología , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ácido Glutámico/metabolismo , Proteína Quinasa C/metabolismo , Simportadores , Acetato de Tetradecanoilforbol/farmacología , 1-Metil-3-Isobutilxantina/farmacología , Animales , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Membrana Celular/efectos de los fármacos , Colforsina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Transportador 3 de Aminoácidos Excitadores , Proteínas de Transporte de Glutamato en la Membrana Plasmática , Ácido Glutámico/farmacocinética , Humanos , Indoles/farmacología , Maleimidas/farmacología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Forboles/farmacología , Proteína Quinasa C/efectos de los fármacos , ARN Complementario/farmacología , Tritio/farmacocinética , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo , Xenopus laevis
11.
Genomics ; 76(1-3): 99-109, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11549322

RESUMEN

We report here the genomic structures of the genes encoding human calcium transport proteins CaT1 and CaT2, which belong to a recently identified class of highly selective calcium entry channels. The mRNA for CaT1 was expressed more abundantly than that for CaT2 in three major tissues involved in transcellular calcium transport, namely intestine, kidney, and placenta, as determined by quantitative PCR. The genes encoding CaT1 and CaT2, ECAC2 and ECAC1, respectively, are completely conserved in terms of exon size in the coding regions. They also share similar intron-exon structures with the genes encoding the closely related, nonselective cation channels VR1, VRL-1, OTRPC4 (also known as VR-OAC, Trp12, and VRL-2), and a hypothetical protein, VRL-3. We conclude that ECAC2 and ECAC1, which encode calcium selective channels, share a common ancestral gene with the genes encoding the related nonselective cation channels.


Asunto(s)
Canales de Calcio/química , Secuencia Conservada , Secuencia de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos Básicos , Animales , Canales de Calcio/biosíntesis , Canales de Calcio/genética , Canales de Calcio/fisiología , Clonación Molecular , Secuencia Conservada/genética , Exones/genética , Femenino , Biblioteca de Genes , Humanos , Intestino Delgado/química , Riñón/química , Riñón/metabolismo , Datos de Secuencia Molecular , Oocitos/química , Oocitos/metabolismo , Placenta/química , Embarazo , Canales Catiónicos TRPV , Xenopus laevis/genética
12.
Biochem Biophys Res Commun ; 282(3): 729-34, 2001 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-11401523

RESUMEN

Ca(2+) signaling is important for growth and survival of prostatic carcinoma (PCa) cells. Here we report that the gene for CaT1, a channel protein highly selective for Ca(2+), is expressed at high levels in human PCa and in the LNCaP PCa cell line. CaT1 mRNA levels were elevated in PCa specimens in comparison to benign prostatic hyperplasia (BPH) specimens and positively correlated with Gleason grade in a PCa series. CaT1 mRNA was suppressed by androgen and was induced by a specific androgen receptor antagonist in LNCaP cells, suggesting that the gene is negatively regulated by androgen. These findings are the first to implicate a Ca(2+) channel in PCa progression and suggest that CaT1 may be a novel target for therapy.


Asunto(s)
Canales de Calcio/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Antagonistas de Andrógenos/farmacología , Andrógenos/metabolismo , Anilidas/farmacología , Secuencia de Bases , Señalización del Calcio , Línea Celular , Cartilla de ADN/genética , Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Hibridación in Situ , Masculino , Nitrilos , Próstata/metabolismo , Hiperplasia Prostática/genética , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patología , Neoplasias de la Próstata/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Canales Catiónicos TRPV , Compuestos de Tosilo , Células Tumorales Cultivadas
13.
Gastroenterology ; 120(7): 1666-79, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11375948

RESUMEN

BACKGROUND & AIMS: hPepT1 is an intestinal epithelial apical membrane transporter responsible for uptake of di/tripeptides (including bacterial derived proinflammatory n-formyl peptides). hPepT1 expression normally has a strict axial gradient-highest in the proximal small intestine with no expression in the colon. METHODS: Small intestinal-like cells (Caco2-BBE), and colonic-like cells (HT29-Cl.19A), and colonic mucosa from diseased and control patients were used in the present study. RESULTS: hPepT1 expression occurs aberrantly in the colon with chronic ulcerative colitis (6 patients) and Crohn's disease (4 patients), but not in normal colon (4 patients) or colon with microscopic colitis (4 patients). To model expression of hPepT1 by colonic-like cells in inflamed states, we stably transfected HT29-Cl.19A cells with a modified hPepT1 tagged on the N-terminus with green fluorescence protein. Analysis of transfected cells revealed that: GFP-hPepT1 protein, like the natural protein, is targeted to the apical plasma membrane. In addition, the tagged protein retains the capability of di/tripeptide absorption, and the expression of the tagged protein by HT29-Cl.19A cells permits absorption of N-formyl-methionyl-leucyl-phenylalanine (fMLP), as occurs in hPepT1 expressing Caco2-BBE cells. fMLP uptake by colonic cells expressing GFP-hPepT1 specifically enhances major histocompatibility complex class I surface expression. CONCLUSIONS: These data collectively indicate that, in some states of chronic inflammation, hPepT1 may be anomolously expressed in the colon. Further, transport of fMLP by hPepT1 potentially stimulates expression of key accessory immune molecule, MHC-1.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Portadoras/fisiología , Antígenos de Histocompatibilidad Clase I/análisis , Enfermedades Inflamatorias del Intestino/metabolismo , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Simportadores , Secuencia de Aminoácidos , Transporte Biológico , Células CACO-2 , Proteínas Portadoras/análisis , Colitis/metabolismo , Colon/química , Células HT29 , Humanos , Intestino Delgado/química , Datos de Secuencia Molecular , Transportador de Péptidos 1
14.
J Comp Neurol ; 433(1): 101-14, 2001 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-11283952

RESUMEN

The ventral one-third of the ventricular lining in the hypothalamus is formed by specialized ependymal cells called the tanycytes. These cells may serve a neuroendocrine transport function because of their structural specializations, which include apical microvili on the ventricular surface and long basal processes that terminate on blood vessels or on the glia limitans. Here, we describe the expression of mRNA and protein for the glutamate transporters GLT-1 and GLAST in unique tanycyte populations of the third ventricle in rat brain. Using nonisotopic in situ hybridization, we demonstrate GLAST mRNA labeling in tanycytes of the ventral floor and lateral walls in the tuberal and mammillary recess portions of the third ventricle. This GLAST mRNA labeling had a higher intensity than the labeling intensity observed in regular ependymal cells throughout the ventricular system. Furthermore, we have identified strong GLT-1 mRNA labeling in a population of tanycytes situated in the dorsolateral walls of caudal tuberal and mammillary recess portions. Immunocytochemical staining indicates that both GLT-1 and GLAST protein are expressed in the tanycyte populations as well. These data corroborate previous findings that third ventricle tanycytes are functionally heterogeneous. Furthermore, the GLT-1-expressing tanycytes represent a population of tanycytes that, to date, has not been recognized as functionally distinct. The strong GLAST expression by the ventral tanycytes in the hypophysiotropic area suggests a role of tanycyte-mediated glutamate transport in neuroendocrine activity. The functional role of GLT-1 in dorsal wall tanycytes remains to be explored.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/análisis , Epéndimo/química , Epéndimo/citología , Tercer Ventrículo/química , Tercer Ventrículo/citología , Transportadoras de Casetes de Unión a ATP/genética , Sistema de Transporte de Aminoácidos X-AG , Animales , Transportador 2 de Aminoácidos Excitadores , Proteína Ácida Fibrilar de la Glía/análisis , Inmunohistoquímica , Hibridación in Situ , Masculino , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Receptores de Neurotransmisores/análisis , Receptores de Neurotransmisores/genética
15.
Nature ; 410(6829): 705-9, 2001 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-11287959

RESUMEN

The calcium-release-activated Ca2+channel, ICRAC, is a highly Ca2+-selective ion channel that is activated on depletion of either intracellular Ca2+ levels or intracellular Ca2+ stores. The unique gating of ICRAC has made it a favourite target of investigation for new signal transduction mechanisms; however, without molecular identification of the channel protein, such studies have been inconclusive. Here we show that the protein CaT1 (ref. 4), which has six membrane-spanning domains, exhibits the unique biophysical properties of ICRAC when expressed in mammalian cells. Like ICRAC, expressed CaT1 protein is Ca2+ selective, activated by a reduction in intracellular Ca2+ concentration, and inactivated by higher intracellular concentrations of Ca2+. The channel is indistinguishable from ICRAC in the following features: sequence of selectivity to divalent cations; an anomalous mole fraction effect; whole-cell current kinetics; block by lanthanum; loss of selectivity in the absence of divalent cations; and single-channel conductance to Na+ in divalent-ion-free conditions. CaT1 is activated by both passive and active depletion of calcium stores. We propose that CaT1 comprises all or part of the ICRAC pore.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Animales , Células CHO , Canales de Calcio/genética , Cricetinae , Cricetulus , Electrofisiología , Ratas , Canales Catiónicos TRPV , Transfección , Células Tumorales Cultivadas
16.
Biochem Biophys Res Commun ; 282(5): 1251-6, 2001 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-11302751

RESUMEN

Most patients with autosomal dominant polycystic kidney disease (ADPKD) harbor mutations truncating polycystin-1 (PC1) or polycystin-2 (PC2), products of the PKD1 and PKD2 genes, respectively. A third member of the polycystin family, polycystin-L (PCL), was recently shown to function as a Ca(2+)-modulated nonselective cation channel. More recently, PC2 was also shown to be a nonselective cation channel with comparable properties to PCL, though the membrane targeting of PC2 likely varies with cell types. Here we show that PC2 expressed heterologously in Xenopus oocytes is targeted to intracellular compartments. By contrast, a truncated form of mouse PC2 corresponding to a naturally occurring human mutation R742X is targeted predominantly to the plasma membrane where it mediates K(+), Na(+), and Ca(2+) currents. Unlike PCL, the truncated form does not display Ca(2+)-activated transport activities, possibly due to loss of an EF-hand at the C-terminus. We propose that PC2 forms ion channels utilizing structural components which are preserved in the R742X form of the protein. Implications for epithelial cell signaling are discussed.


Asunto(s)
Sustitución de Aminoácidos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación/genética , Secuencias de Aminoácidos/fisiología , Animales , Calcio/metabolismo , Calcio/farmacocinética , Compartimento Celular/fisiología , Membrana Celular/metabolismo , Células Cultivadas , Líquido Intracelular/metabolismo , Canales Iónicos/fisiología , Potenciales de la Membrana/fisiología , Ratones , Microinyecciones , Técnicas de Placa-Clamp , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Transporte de Proteínas/fisiología , ARN Mensajero/administración & dosificación , ARN Mensajero/metabolismo , Canales Catiónicos TRPP , Xenopus
17.
Biochem Biophys Res Commun ; 282(1): 341-50, 2001 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-11264013

RESUMEN

Mutations in polycystins-1 and -2 (PC1 and PC2) cause autosomal dominant polycystic kidney disease (ADPKD), which is characterized by progressive development of epithelial renal cysts, ultimately leading to renal failure. The functions of these polycystins remain elusive. Here we show that PC2 is a Ca(2+)-permeable cation channel with properties distinct from any known intracellular channels. Its kinetic behavior is characterized by frequent transitions between closed and open states over a wide voltage range. The activity of the PC2 channel is transiently increased by elevating cytosolic Ca(2+). Given the predominant endoplasmic reticulum (ER) location of PC2 and its unresponsiveness to the known modulators of mediating Ca(2+) release from the ER, inositol-trisphosphate (IP(3)) and ryanodine, these results suggest that PC2 represents a novel type of channel with properties distinct from those of the other Ca(2+)-release channels. Our data also show that the PC2 channel can be translocated to the plasma membranes by defined chemical chaperones and proteasome modulators, suggesting that in vivo, it may also function in the plasma membrane under specific conditions. The sensitivity of the PC2 channel to changes of intracellular Ca(2+) concentration is deficient in a mutant found in ADPKD patients. The dysfunction of such mutants may result in defective coupling of PC2 to intracellular Ca(2+) homeostasis associated with the pathogenesis of ADPKD.


Asunto(s)
Canales de Calcio/fisiología , Calcio/metabolismo , Homeostasis/fisiología , Proteínas de la Membrana/fisiología , Enfermedades Renales Poliquísticas/fisiopatología , Animales , Canales de Calcio/genética , Clonación Molecular , ADN Complementario , Humanos , Inmunohistoquímica , Proteínas de la Membrana/genética , Ratones , Canales Catiónicos TRPP , Xenopus
18.
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
19.
Am J Physiol Renal Physiol ; 280(3): F487-94, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11181411

RESUMEN

In the terminal part of the kidney collecting duct, rapid urea reabsorption is essential to maintaining medullary hypertonicity, allowing maximal urinary concentration to occur. This process is mediated by facilitated urea transporters on both apical and basolateral membranes. Our previous studies have identified three rat urea transporters involved in the urinary concentrating mechanism, UT1, UT2 and UT3, herein renamed UrT1-A, UrT1-B, and UrT2, which exhibit distinct spatial distribution in the kidney. Here we report the molecular characterization of an additional urea transporter isoform, UrT1-C, from rat kidney that encodes a 460-amino acid residue protein. UrT1-C has 70 and 62% amino acid identity to rat UrT1-B and UrT2 (UT3), respectively, and 99% identity to a recently reported rat isoform (UT-A3; Karakashian A, Timmer RT, Klein JD, Gunn RB, Sands JM, and Bagnasco SM. J Am Soc Nephrol 10: 230-237, 1999). We report the anatomic distribution of UrT1-C in the rat kidney tubule system as well as a detailed functional characterization. UrT1-C m RNA is primarily expressed in the deep part of the inner medulla. When expressed in Xenopus laevis oocytes, UrT1-C induced a 15-fold stimulation of urea uptake, which was inhibited almost completely by phloretin (0.7 mM) and 60-95% by thiourea analogs (150 mM). The characteristics are consistent with those described in perfusion studies with inner medullary collecting duct (IMCD) segments, but, contrary to UrT1-A, UrT1-C-mediated urea uptake was not stimulated by activation of protein kinase A. Our data show that UrT1-C is a phloretin-inhibitable urea transporter expressed in the terminal collecting duct that likely serves as an exit mechanism for urea at the basolateral membrane of IMCD cells.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Túbulos Renales Colectores/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Transporte de Membrana , Secuencia de Aminoácidos/genética , Animales , Proteínas Portadoras/fisiología , Clonación Molecular , ADN Complementario/genética , Médula Renal , Glicoproteínas de Membrana/fisiología , Datos de Secuencia Molecular , Oocitos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/metabolismo , Ratas , Distribución Tisular , Xenopus laevis , Transportadores de Urea
20.
Am J Physiol Gastrointest Liver Physiol ; 280(2): G209-15, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11208542

RESUMEN

Mechanisms underlying the circadian rhythmicity in intestinal sugar absorption remain unclear. To test whether this rhythmicity is caused by changes in Na(+)-glucose cotransporter 1 (SGLT-1) function, we measured phloridzin-inhibitable sugar fluxes as an index of SGLT-1 activity. Jejunum obtained from rats killed at 6-h intervals during a 12-h light-dark cycle (CT0 is circadian time 0 h, time of light onset) were mounted in Ussing chambers, and 3-O-methylglucose (3-OMG) fluxes were calculated before and after addition of phloridzin. 3-OMG-induced change in short-circuit current and absorptive flux were significantly greater at CT9 than at CT3. This increase was phloridzin inhibitable. Kinetic studies indicated a significant increase in SGLT-1 maximal velocity (V(max)) at CT9. Food intake between CT3 and CT9 was <10% of the daily total, indicating that the increased SGLT-1 activity was anticipatory. Diurnicity of SGLT-1 mRNA was confirmed by Northern blotting. Expression topography analyzed by in situ hybridization revealed more intense labeling along the entire villus axis at CT9 and CT15 compared with CT3 and CT21. We conclude that diurnicity in intestinal sugar absorption is caused by periodicity in SGLT-1 V(max).


Asunto(s)
Ritmo Circadiano , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/fisiología , ARN Mensajero/metabolismo , Animales , Ingestión de Alimentos/efectos de los fármacos , Conductividad Eléctrica , Femenino , Guanosina/análogos & derivados , Guanosina/farmacocinética , Guanosina/farmacología , Técnicas In Vitro , Yeyuno/efectos de los fármacos , Yeyuno/metabolismo , Yeyuno/fisiología , Cinética , Florizina/farmacología , Ratas , Ratas Sprague-Dawley , Transportador 1 de Sodio-Glucosa , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA