Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 276(47): 44091-8, 2001 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-11564745

RESUMEN

Epithelial sodium channels (ENaC) are composed of three structurally related subunits (alpha, beta, and gamma). Each subunit has two transmembrane domains termed M1 and M2, and residues conferring cation selectivity have been shown to reside in a pore region immediately preceding the M2 domains of the three subunits. Negatively charged residues are interspersed within the M2 domains, and substitution of individual acidic residues within human alpha-ENaC with arginine essentially eliminated channel activity in oocytes, suggesting that these residues have a role in ion permeation. We examined the roles of M2 residues in contributing to the permeation pore by individually mutating residues within the M2 domain of mouse alphaENaC to cysteine and systematically characterizing functional properties of mutant channels expressed in Xenopus oocytes by two-electrode voltage clamp. The introduction of cysteine residues at selected sites, including negatively charged residues (alphaGlu(595), alphaGlu(598), and alphaAsp(602)) led to a significant reduction of expressed amiloride-sensitive Na(+) currents. Two mutations (alphaE595C and alphaD602C) resulted in K(+)-permeable channels whereas multiple mutations altered Li(+)/Na(+) current ratios. Channels containing alphaD602K or alphaD602A also conducted K(+) whereas more conservative mutations (alphaD602E and alphaD602N) retained wild type selectivity. Cysteine substitution at the site equivalent to alphaAsp(602) within beta mENaC (betaD544C) did not alter either Li(+)/Na(+) or K(+)/Na(+) current ratios, although mutation of the equivalent site within gamma mENaC (gammaD562C) significantly increased the Li(+)/Na(+) current ratio. Mutants containing introduced cysteine residues at alphaGlu(595), alphaGlu(598), alphaAsp(602), or alphaThr(607) did not respond to externally applied sulfhydryl reagent with significant changes in macroscopic currents. Our results suggest that some residues within the M2 domain of alphaENaC contribute to the channel's conduction pore and that, in addition to the pore region, selected sites within M2 (alphaGlu(595) and alphaAsp(602)) may have a role in conferring ion selectivity.


Asunto(s)
Canales de Sodio/metabolismo , Secuencia de Aminoácidos , Animales , Permeabilidad de la Membrana Celular , Canales Epiteliales de Sodio , Humanos , Transporte Iónico , Potenciales de la Membrana , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Conformación Proteica , Homología de Secuencia de Aminoácido , Canales de Sodio/química , Canales de Sodio/genética , Canales de Sodio/fisiología , Xenopus
2.
Am J Physiol Renal Physiol ; 281(2): F213-21, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11457713

RESUMEN

The renal epithelial cell line A6, derived from Xenopus laevis, expresses epithelial Na(+) channels (ENaCs) and serves as a model system to study hormonal regulation and turnover of ENaCs. Our previous studies suggest that the alpha-subunit of Xenopus ENaC (alpha-xENaC) is detectable as 150- and 180-kDa polypeptides, putative immature and mature alpha-subunit heterodimers. The 150- and 180-kDa alpha-xENaC were present in distinct fractions after sedimentation of A6 cell lysate through a sucrose density gradient. Two anti-alpha-xENaC antibodies directed against distinct domains demonstrated that only 180-kDa alpha-xENaC was expressed at the apical cell surface. The half-life of cell surface-expressed alpha-xENaC was 24-30 h, suggesting that once ENaC matures and is expressed at the plasma membrane, its turnover is similar to that reported for mature cystic fibrosis transmembrane conductance regulator. No significant changes in apical surface expression of alpha-xENaC were observed after treatment of A6 cells with aldosterone for 24 h, despite a 5.3-fold increase in short-circuit current. This lack of change in surface expression is consistent with previous observations in A6 cells and suggests that aldosterone regulates ENaC gating and increases channel open probability.


Asunto(s)
Células Epiteliales/metabolismo , Túbulos Renales/metabolismo , Canales de Sodio/metabolismo , Urotelio/metabolismo , Aldosterona/farmacología , Animales , Línea Celular , Polaridad Celular , Centrifugación por Gradiente de Densidad , Células Epiteliales/efectos de los fármacos , Canales Epiteliales de Sodio , Immunoblotting , Túbulos Renales/citología , Túbulos Renales/efectos de los fármacos , Pruebas de Precipitina , Subunidades de Proteína , Factores de Tiempo , Urotelio/citología , Urotelio/efectos de los fármacos , Xenopus laevis
3.
J Biol Chem ; 276(2): 1326-34, 2001 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-11022046

RESUMEN

Epithelial sodium channels (ENaC) have a crucial role in the regulation of extracellular fluid volume and blood pressure. To study the structure of the pore region of ENaC, the susceptibility of introduced cysteine residues to sulfhydryl-reactive methanethiosulfonate derivatives ((2-aminoethyl)methanethiosulfonate hydrobromide (MTSEA) and [(2-(trimethylammonium)ethyl]methanethiosulfonate bromide (MTSET)) and to Cd(2+) was determined. Selected mutants within the amino-terminal portion (alphaVal(569)-alphaTrp(582)) of the pore region responded to MTSEA, MTSET, or Cd(2+) with stimulation or inhibition of whole cell Na(+) current. The reactive residues were not contiguous but were separated by 2-3 residues where substituted cysteine residues did not respond to the reagents and line one face of an alpha-helix. The activation of alphaS580Cbetagamma mENaC by MTSET was associated with a large increase in channel open probability. Within the carboxyl-terminal portion (alphaSer(583)-alphaSer(592)) of the pore region, only one mutation (alphaS583C) conferred a rapid, nearly complete block by MTSEA, MTSET, and Cd(2+), whereas several other mutant channels were partially blocked by MTSEA or Cd(2+) but not by MTSET. Our data suggest that the outer pore of ENaC is formed by an alpha-helix, followed by an extended region that forms a selectivity filter. Furthermore, our data suggest that the pore region participates in ENaC gating.


Asunto(s)
Activación del Canal Iónico/fisiología , Potenciales de la Membrana/fisiología , Canales de Sodio/química , Canales de Sodio/fisiología , Amilorida/farmacología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Cisteína , Canales Epiteliales de Sodio , Femenino , Humanos , Potenciales de la Membrana/efectos de los fármacos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Oocitos/fisiología , Técnicas de Placa-Clamp , Conformación Proteica , Estructura Secundaria de Proteína , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Xenopus laevis
4.
J Biol Chem ; 275(12): 8572-81, 2000 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-10722696

RESUMEN

The epithelial sodium channel (ENaC) is composed of three homologous subunits termed alpha, beta, and gamma. Previous studies suggest that selected residues within a hydrophobic region immediately preceding the second membrane-spanning domain of each subunit contribute to the conducting pore of ENaC. We probed the pore of mouse ENaC by systematically mutating all 24 amino acids within this putative pore region of the alpha-subunit to cysteine and co-expressing these mutants with wild type beta- and gamma-subunits of mouse ENaC in Xenopus laevis oocytes. Functional characteristics of these mutants were examined by two-electrode voltage clamp and single channel recording techniques. Two distinct domains were identified based on the functional changes associated with point mutations. An amino-terminal domain (alpha-Val(569)-alpha-Gly(579)) showed minimal changes in cation selectivity or amiloride sensitivity following cysteine substitution. In contrast, cysteine substitutions within the carboxyl-terminal domain (alpha-Ser(580)-alpha-Ser(592)) resulted in significant changes in cation selectivity and moderately altered amiloride sensitivity. The mutant channels containing alphaG587C or alphaS589C were permeable to K(+), and mutation of a GSS tract (positions alpha587-alpha589) to GYG resulted in a moderately K(+)-selective channel. Our results suggest that the C-terminal portion of the pore region within the alpha-subunit contributes to the selectivity filter of ENaC.


Asunto(s)
Canales de Sodio/metabolismo , Amilorida/farmacología , Secuencia de Aminoácidos , Animales , Cationes Monovalentes/metabolismo , Análisis Mutacional de ADN , Relación Dosis-Respuesta a Droga , Conductividad Eléctrica , Canales Epiteliales de Sodio , Litio/metabolismo , Ratones , Modelos Teóricos , Datos de Secuencia Molecular , Oocitos , Técnicas de Placa-Clamp , Mutación Puntual , Potasio/metabolismo , Proteínas Recombinantes/metabolismo , Sodio/metabolismo , Canales de Sodio/genética , Xenopus
5.
Am J Physiol ; 274(1): F91-6, 1998 01.
Artículo en Inglés | MEDLINE | ID: mdl-9458827

RESUMEN

The mammalian urinary bladder exhibits transepithelial Na+ absorption that contributes to Na+ gradients established by the kidney. Electrophysiological studies have demonstrated that electrogenic Na+ absorption across the urinary bladder is mediated in part by amiloride-sensitive Na+ channels situated within the apical membrane of the bladder epithelium. We have used a combination of in situ hybridization, Northern blot analysis, and immunocytochemistry to examine whether the recently cloned epithelial Na+ channel (ENaC) is expressed in the rat urinary bladder. In situ hybridization and Northern blot analyses indicate that alpha-, beta-, and gamma-rat ENaC (rENaC) are expressed in rat urinary bladder epithelial cells. Quantitation of the levels of alpha-, beta-, and gamma-rENaC mRNA expression in rat urinary bladder, relative to beta-actin mRNA expression, indicates that, although comparable levels of alpha- and beta-rENaC subunits are expressed in the urinary bladder of rats maintained on standard chow, the level of gamma-rENaC mRNA expression is 5- to 10-fold lower than alpha- or beta-rENaC mRNA. Immunocytochemistry, using an antibody directed against alpha-rENaC, revealed that ENaCs are predominantly localized to the luminal membrane of the bladder epithelium. Together, these data demonstrate that ENaC is expressed in the mammalian urinary bladder and suggest that amiloride-sensitive Na+ transport across the apical membrane of the mammalian urinary bladder epithelium is mediated primarily by ENaC.


Asunto(s)
Canales de Sodio/biosíntesis , Transcripción Genética , Vejiga Urinaria/metabolismo , Animales , Células Epiteliales/citología , Células Epiteliales/metabolismo , Canales Epiteliales de Sodio , Técnica del Anticuerpo Fluorescente Indirecta , Mamíferos , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Sodio/metabolismo , Canales de Sodio/análisis , Vejiga Urinaria/citología
6.
J Biol Chem ; 272(1): 594-600, 1997 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-8995302

RESUMEN

Mutations in a Cl- channel (cystic fibrosis transmembrane conductance regulator or CFTR) are responsible for the cystic fibrosis (CF) phenotype. Increased Na+ transport rates are observed in CF airway epithelium, and recent studies suggest that this is due to an increase in Na+ channel open probability (Po). The Xenopus renal epithelial cell line, A6, expresses both cAMP-activated 8-picosiemen (pS) Cl- channels and amiloride-sensitive 4-pS Na+ channels, and provides a model system for examining the interactions of CFTR and epithelial Na+ channels. A6 cells express CFTR mRNA, as demonstrated by reverse transcriptase-polymerase chain reaction and partial sequence analysis. A phosphorothioate antisense oligonucleotide, complementary to the 5' end of the open reading frame of Xenopus CFTR, was used to inhibit functional expression of CFTR in A6 cells. Parallel studies utilized the corresponding sense oligonucleotide as a control. CFTR protein expression was markedly reduced in cells incubated with the antisense oligonucleotide. Incubation of A6 cells with the antisense oligonucleotide led to inhibition of forskolin-activated amiloride-insensitive short circuit current (Isc). After a 30-min exposure to 10 microM forskolin, 8-pS Cl- channel activity was detected in only 1 of 31 (3%) cell-attached patches on cells treated with antisense oligonucleotide, compared to 5 of 19 (26%) patches from control cells. A shift in the single-channel current-voltage relationship derived from antisense-treated cells was also consistent with a reduction in Cl- reabsorption. Both amiloride-sensitive Isc and Na+ channel Po were significantly increased in antisense-treated, forskolin-stimulated A6 cells, when compared with forskolin-stimulated controls. These data suggest that the regulation of Na+ channels by CFTR is not limited to respiratory epithelia and to epithelial cells in culture overexpressing CFTR and epithelial Na+ channels.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Fibrosis Quística/fisiopatología , Canales de Sodio/fisiología , Amilorida/farmacología , Animales , Células Cultivadas , Técnica del Anticuerpo Fluorescente Indirecta , Expresión Génica/efectos de los fármacos , Modelos Biológicos , Oligonucleótidos Antisentido/farmacología , Técnicas de Placa-Clamp , Fenotipo , Sodio/fisiología , Bloqueadores de los Canales de Sodio , Xenopus laevis
7.
Neurology ; 46(3): 673-7, 1996 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8618665

RESUMEN

Apolipoprotein E type 4 allele (apoE epsilon4) is associated with Alzheimer's disease (AD) in the late-onset familial form and in sporadic cases, but the age-associated risk in a randomly sampled elderly population is not established. We examined the association of apoE epsilon4 with AD and other dementias (mainly multi-infarct or dementia following stroke) in 1,030 persons aged 71 to 100 years in the population-based Framingham Study cohort. Kaplan-Meier survival analysis revealed that 55% of the apoE epsilon4/epsilon4 homozygotes developed AD by age 80, whereas 27% of apoE epsilon3/epsilon4 heterozygotes developed AD by age 85, and 9% of those without a 4 allele developed AD by age 85 years. In comparison with persons without a 4 allele, the risk ration for AD was 3.7 (95% CI = 1.9 to 7.5) for apoE epsilon3/epsilon4 heterozygotes and 30.1 (95% CI = 10.7 to 84.4) for apoE epsilon4 homozygotes. ApoE epsilon2 (2/2, 2/3, or 2/4 genotypes) was associated with an absence of AD. One-half (n=21) of the 43 AD patients were either homozygous or heterozygous for apoE epsilon4. We found evidence for an association of apoE epsilon4 with other dementia, primarily multi-infarct dementia and stroke. The risk ratio was 2.3 (95% CI = 0.9 to 6.1) for non-AD dementias among persons with apoE epsilon3/epsilon4. Although the apoE epsilon4 allele is a potent risk factor for AD and may be associated with other forms of dementia, most apoE epsilon4 carriers do not develop dementia, and about one-half of AD is not apoE epsilon4 associated. The low positive predictive value of this marker (0.10) suggest that use of apoE genotyping as a screening test for AD is not supported.


Asunto(s)
Apolipoproteínas E/genética , Demencia/genética , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Enfermedad de Alzheimer/epidemiología , Enfermedad de Alzheimer/genética , Apolipoproteína E4 , Secuencia de Bases , Estudios de Cohortes , Femenino , Humanos , Incidencia , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Sondas Moleculares/genética , Datos de Secuencia Molecular , Oportunidad Relativa , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA