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











Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Renal Physiol ; 283(5): F1030-45, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12372779

RESUMO

We studied the cellular phosphatase inhibitors okadaic acid (OKA), calyculin A, and microcystin on the epithelial sodium channel (ENaC) in A6 renal cells. OKA increased the amiloride-sensitive current after approximately 30 min with maximal stimulation at 1-2 h. Fluctuation analysis of cell-attached patches containing a large number of ENaC yielded power spectra with corner frequencies in untreated cells almost two times as large as in cells pretreated for 30 min with OKA, implying an increase in single channel open probability (P(o)) that doubled after OKA. Single channel analysis showed that, in cells pretreated with OKA, P(o) and mean open time approximately doubled. Two other phosphatase inhibitors, calyculin A and microcystin, had similar effects on P(o) and mean open time. An analog of OKA, okadaone, that does not inhibit phosphatases had no effect. Pretreatment with 10 nM OKA, which blocks protein phosphatase 2A (PP2A) but not PP1 in mammalian cells, had no effect even though both phosphatases are present in A6 cells. Several proteins were differentially phosphorylated after OKA, but ENaC subunit phosphorylation did not increase. We conclude that, in A6 cells, there is an OKA-sensitive phosphatase that suppresses ENaC activity by altering the phosphorylation of a regulatory molecule associated with the channel.


Assuntos
Rim/enzimologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosfoproteínas Fosfatases/metabolismo , Canais de Sódio/metabolismo , Animais , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Inibidores Enzimáticos/farmacologia , Canais Epiteliais de Sódio , Ativação do Canal Iônico/fisiologia , Isomerismo , Rim/citologia , Toxinas Marinhas , Microcistinas , Ácido Okadáico/análogos & derivados , Ácido Okadáico/farmacologia , Oxazóis/farmacologia , Técnicas de Patch-Clamp/métodos , Peptídeos Cíclicos/farmacologia , Fosforilação , Proteína Quinase C/metabolismo , Proteína Fosfatase 2 , Canais de Sódio/química
2.
J Biol Chem ; 276(16): 12903-10, 2001 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-11278712

RESUMO

Amiloride-sensitive epithelial Na(+) channels (ENaC) are responsible for trans-epithelial Na(+) transport in the kidney, lung, and colon. The channel consists of three subunits (alpha, beta, gamma) each containing a proline rich region (PPXY) in their carboxyl-terminal end. Mutations in this PPXY domain cause Liddle's syndrome, an autosomal dominant, salt-sensitive hypertension, by preventing the channel's interactions with the ubiquitin ligase Neural precursor cell-expressed developmentally down-regulated protein (Nedd4). It is postulated that this results in defective endocytosis and lysosomal degradation of ENaC leading to an increase in ENaC activity. To show the pathway that degrades ENaC in epithelial cells that express functioning ENaC channels, we used inhibitors of the proteosome and measured sodium channel activity. We found that the inhibitor, MG-132, increases amiloride-sensitive trans-epithelial current in Xenopus distal nephron A6 cells. There also is an increase of total cellular as well as membrane-associated ENaC subunit molecules by Western blotting. MG-132-treated cells also have increased channel density in patch clamp experiments. Inhibitors of lysosomal function did not reproduce these findings. Our results suggest that in native renal cells the proteosomal pathway is an important regulator of ENaC function.


Assuntos
Cisteína Endopeptidases/metabolismo , Complexos Multienzimáticos/metabolismo , Canais de Sódio/metabolismo , Ubiquitinas/metabolismo , Sequência de Aminoácidos , Animais , Especificidade de Anticorpos , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Inibidores de Cisteína Proteinase/farmacologia , Canais Epiteliais de Sódio , Humanos , Cinética , Leupeptinas/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Modelos Biológicos , Dados de Sequência Molecular , Néfrons , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Complexo de Endopeptidases do Proteassoma , Subunidades Proteicas , Canais de Sódio/química , Canais de Sódio/genética , Urotélio/citologia , Urotélio/fisiologia , Xenopus laevis
3.
DNA Seq ; 12(5-6): 425-9, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11913791

RESUMO

The cellular homolog of the oncogene v-src, the proto-oncogene c-src, was cloned from rat testis using a high stringency polymerase chain reaction. Rat c-src cDNA shared identity with chicken and mouse, and Rous sarcoma virus c-src and v-src, respectively. Rat c-Src protein was 98% homologous to both human and mouse c-Src. Interestingly, rat Src contained one extra amino acid compared to the mouse protein. As expected, the rat testis Src lacked the six extra residues common to the neuronal Src identified in human and mouse. Reporting of the cDNA sequence for non-neuronal, rat c-src should facilitate experimentation into cell growth and transformation using rat tissues as models of human disease.


Assuntos
Genes src , Testículo/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Proto-Oncogene Mas , Ratos , Alinhamento de Sequência
4.
J Membr Biol ; 184(3): 313-9, 2001 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11891557

RESUMO

Aldosterone maintains total organism sodium balance in all higher vertebrates. The level of sodium reabsorption is primarily determined by the action of aldosterone on epithelial sodium channels (ENaC) in the distal nephron. Recent work shows that, in an aldosterone-sensitive renal cell line (A6), aldosterone regulates sodium reabsorption by short- and long-term processes. In the short term, aldosterone regulates sodium transport by inducing expression of the small G-protein, K-Ras2A, by stimulating the activity of methyl transferase and S-adenosyl-homocysteine hydrolase to activate Ras by methylation, and, possibly, by subsequent activation by K-Ras2A of phosphatidylinositol phosphate-5-kinase (PIP-5-K) and phosphatidylinositol-3-kinase (PI-3-K), which ultimately activates ENaC. In the long term, aldosterone regulates sodium transport by altering trafficking, assembly, and degradation of ENaC.


Assuntos
Aldosterona/farmacologia , Células Epiteliais/metabolismo , Canais de Sódio/farmacologia , Sódio/farmacocinética , Aldosterona/metabolismo , Aldosterona/fisiologia , Animais , Anuros , Transporte Biológico/fisiologia , Membrana Celular/fisiologia , Condutividade Elétrica , Células Epiteliais/efeitos dos fármacos , Canais Epiteliais de Sódio , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Técnicas de Patch-Clamp/métodos , Canais de Sódio/farmacocinética , ATPase Trocadora de Sódio-Potássio/fisiologia
5.
Am J Physiol Cell Physiol ; 279(2): C429-39, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10913010

RESUMO

Aldosterone increases Na(+) reabsorption by renal epithelial cells: the acute actions (<4 h) appear to be promoted by protein methylation. This paper describes the relationship between protein methylation and aldosterone's action and describes aldosterone-mediated targets for methylation in cultured renal cells (A6). Aldosterone increases protein methylation from 7.90 +/- 0.60 to 20.1 +/- 0.80 methyl ester cpm/microg protein. Aldosterone stimulates protein methylation by increasing methyltransferase activity from 14.0 +/- 0.64 in aldosterone-depleted cells to 31.8 +/- 2.60 methyl ester cpm/microg protein per hour in aldosterone-treated cells. Three known methyltransferase inhibitors reduce the aldosterone-induced increase in methyltransferase activity. One of these inhibitors, the isoprenyl-cysteine methyltransferase-specific inhibitor, S-trans, trans-farnesylthiosalicylic acid, completely blocks aldosterone-induced protein methylation and also aldosterone-induced short-circuit current. Aldosterone induces protein methylation in two molecular weight ranges: near 90 kDa and around 20 kDa. The lower molecular weight range is the weight of small G proteins, and aldosterone does increase both Ras protein 1.6-fold and Ras methylation almost 12-fold. Also, Ras antisense oligonucleotides reduce the activity of Na(+) channels by about fivefold. We conclude that 1) protein methylation is essential for aldosterone-induced increases in Na(+) transport; 2) one target for methylation is p21(ras); and 3) inhibition of Ras expression or Ras methylation inhibits Na(+) channel activity.


Assuntos
Aldosterona/farmacologia , Células Epiteliais/efeitos dos fármacos , Proteínas Metiltransferases/efeitos dos fármacos , Canais de Sódio/efeitos dos fármacos , Proteínas ras/efeitos dos fármacos , Animais , Células Cultivadas , Células Epiteliais/metabolismo , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/metabolismo , Metilação , Proteínas Metiltransferases/metabolismo , Canais de Sódio/metabolismo , Proteínas ras/metabolismo
6.
Am J Physiol ; 276(6): L1046-51, 1999 06.
Artigo em Inglês | MEDLINE | ID: mdl-10362730

RESUMO

Amiloride-sensitive Na+ transport by lung epithelia plays a critical role in maintaining alveolar Na+ and water balance. It has been generally assumed that Na+ transport is mediated by the amiloride-sensitive epithelial Na+ channel (ENaC) because molecular biology studies have confirmed the presence of ENaC subunits alpha, beta, and gamma in lung epithelia. However, the predominant Na+-transporting channel reported from electrophysiological studies by most laboratories is a nonselective, high-conductance channel that is very different from the highly selective, low-conductance ENaC reported in other tissues. In our laboratory, single-channel recordings from apical membrane patches from rat alveolar type II (ATII) cells in primary culture reveal a nonselective cation channel with a conductance of 20.6 +/- 1.1 pS and an Na+-to-K+ selectivity of 0.97 +/- 0.07. This channel is inhibited by submicromolar concentrations of amiloride. Thus there is some question about the relationship between the gene product observed with single-channel methods and the cloned ENaC subunits. We have employed antisense oligonucleotide methods to block the synthesis of individual ENaC subunit proteins (alpha, beta, and gamma) and determined the effect of a reduction in the subunit expression on the density of the nonselective cation channel observed in apical membrane patches on ATII cells. Treatment of ATII cells with antisense oligonucleotides inhibited the production of each subunit protein; however, single-channel recordings showed that only the antisense oligonucleotide targeting the alpha-subunit resulted in a significant decrease in the density of nonselective cation channels. Inhibition of the beta- and gamma-subunit proteins alone or together did not cause any changes in the observed channel density. There were no changes in open probability or other channel characteristics. These results support the hypothesis that the alpha-subunit of ENaC alone or in combination with some protein other than the beta- or gamma-subunit protein is the major component of lung alveolar epithelial cation channels.


Assuntos
Cátions/metabolismo , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/metabolismo , Pulmão/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Canais de Sódio/genética , Animais , Membrana Celular/metabolismo , Células Epiteliais/metabolismo , Canais Epiteliais de Sódio , Canais Iônicos/antagonistas & inibidores , Pulmão/citologia , Isoformas de Proteínas/genética , Ratos , Ratos Sprague-Dawley , Sódio/metabolismo
7.
J Biol Chem ; 274(6): 3842-50, 1999 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-9920939

RESUMO

Aldosterone-induced Na+ reabsorption, in part, is regulated by a critical methyl esterification; however, the signal transduction pathway regulating this methylation remains unclear. The A6 cell line was used as a model epithelia to investigate regulation of aldosterone-induced Na+ transport by S-adenosyl-L-homocysteine hydrolase (SAHHase), the only enzyme in vertebrates known to catabolize S-adenosyl-L-homocysteine (SAH), an end product inhibitor of methyl esterification. Sodium reabsorption was decreased within 2 h by 3-deazaadenosine, a competitive inhibitor of SAHHase, with a half inhibitory concentration between 40 and 50 microM. Aldosterone increased SAH catabolism by activating SAHHase. Increased SAH catabolism was associated with a concomitant increase in S-adenosylmethionine catabolism. Moreover, SAH decreased substrate methylation. Antisense oligonucleotide complementary to SAHHase mRNA decreased SAHHase activity and Na+ current by approximately 50%. Overexpression of SAHHase increased SAHHase activity and dependent substrate methyl esterification. Whereas basal Na+ current was not affected by overexpression of SAHHase, aldosterone-induced current in SAHHase-overexpressing cells was significantly potentiated. These results demonstrate that aldosterone induction of SAHHase activity is necessary for a concomitant relief of the methylation reaction from end product inhibition by SAH and the subsequent increase in Na+ reabsorption. Thus, regulation of SAHHase activity is a control point for aldosterone signal transduction, but SAHHase is not an aldosterone-induced protein.


Assuntos
Aldosterona/metabolismo , Hidrolases/metabolismo , Sódio/metabolismo , Adenosil-Homocisteinase , Anfíbios , Animais , Sequência de Bases , Células Cultivadas , Primers do DNA , Inibidores Enzimáticos/farmacologia , Hidrolases/antagonistas & inibidores , Transporte de Íons , Metilação , Tubercidina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA