Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Renal Physiol ; 290(6): F1285-94, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16682484

RESUMO

Amiloride-sensitive epithelial Na+ channels (ENaC) play a crucial role in Na+ transport and fluid reabsorption in the kidney, lung, and colon. The magnitude of ENaC-mediated Na+ transport in epithelial cells depends on the average open probability of the channels and the number of channels on the apical surface of epithelial cells. The number of channels in the apical membrane, in turn, depends on a balance between the rate of ENaC insertion and the rate of removal from the apical membrane. ENaC is made up of three homologous subunits: alpha, beta, and gamma. The COOH-terminal domain of all three subunits is intracellular and contains a proline-rich motif (PPxY). Mutations or deletion of this PPxY motif in the beta- and gamma-subunits prevent the binding of one isoform of a specific ubiquitin ligase, neural precursor cell-expressed, developmentally downregulated protein (Nedd4-2), to the channel in vitro and in transfected cell systems, thereby impeding ubiquitin conjugation of the channel subunits. Ubiquitin conjugation would seem to imply that ENaC turnover is determined by the ubiquitin-proteasome system, but when Madin-Darby canine kidney cells are transfected with ENaC, ubiquitin conjugation apparently leads to lysosomal degradation. However, in untransfected renal cells (A6) expressing endogenous ENaC, ENaC is indeed degraded by the ubiquitin-proteasome system. Nonetheless, in both transfected and untransfected cells, the rate of ENaC degradation is apparently controlled by Nedd4-2 activity. In this review, we discuss the role of the ubiquitin conjugation and the alternative degradative pathways (lysosomal or proteasomal) in regulating the rate of ENaC turnover in untransfected renal cells and compare this regulation to that of transfected cell systems.


Assuntos
Células Epiteliais/química , Complexo de Endopeptidases do Proteassoma/metabolismo , Canais de Sódio/fisiologia , Ubiquitina/metabolismo , Animais , Transporte Biológico/fisiologia , Linhagem Celular , Cães , Complexos Endossomais de Distribuição Requeridos para Transporte , Células Epiteliais/metabolismo , Canais Epiteliais de Sódio , Humanos , Rim , Proteínas de Membrana/metabolismo , Mutação , Ubiquitina-Proteína Ligases Nedd4 , Prolina , Subunidades Proteicas/química , Subunidades Proteicas/fisiologia , Sódio/metabolismo , Canais de Sódio/química , Canais de Sódio/genética , Relação Estrutura-Atividade , Transfecção , Ubiquitina-Proteína Ligases/metabolismo
2.
Am J Physiol Renal Physiol ; 289(1): F107-16, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15769939

RESUMO

Amiloride-sensitive epithelial sodium channels (ENaC) are responsible for transepithelial Na(+) transport in the kidney, lung, and colon. The channel consists of three subunits (alpha, beta, and gamma). In Madin-Darby canine kidney (MDCK) cells and Xenopus laevis oocytes transfected with all three ENaC subunits, neural precursor cell-expressed developmentally downregulated protein (Nedd4-2) promotes ubiquitin conjugation of ENaC. For native proteins in some cells, ubiquitin conjugation is a signal for their degradation by the ubiquitin-proteasome pathway, whereas in other cell types ubiquitin conjugation is a signal for endocytosis and lysosomal protein degradation. When ENaC are transfected into MDCK cells, ubiquitin conjugation leads to lysosomal degradation. In this paper, we characterize the involvement of the ubiquitin-proteasome proteolytic pathway in the regulation of functional ENaC in untransfected renal A6 cells expressing native ENaC subunits. In contrast to transfected cells, we show that total cellular alpha-, beta-, and gamma-ENaC subunits are polyubiquitinated and that ubiquitin conjugation of subunits increases when the cells are treated with a proteasome inhibitor. We show that Nedd4-2 is associated with alpha- and beta-subunits and is associated with the apical membrane. We also show the Nedd4-2 can regulate the number of functional ENaC subunits in the apical membrane. The results reported here suggest that the ubiquitin-proteasome proteolytic pathway is an important determinant of ENaC function in untransfected renal cells expressing endogenous ENaC.


Assuntos
Rim/metabolismo , Poliubiquitina/fisiologia , Canais de Sódio/metabolismo , Ubiquitina-Proteína Ligases/fisiologia , Animais , Linhagem Celular , Complexos Endossomais de Distribuição Requeridos para Transporte , Células Epiteliais/metabolismo , Canais Epiteliais de Sódio , Rim/citologia , Ubiquitina-Proteína Ligases Nedd4 , Proteínas de Xenopus , Xenopus laevis
3.
J Neurophysiol ; 93(6): 3146-56, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15647401

RESUMO

BK-channels in GH3 cells are activated by arachidonic acid produced by c-PLA2. beta-adrenergic agonists also activate BK channels and were presumed to do so via production of cAMP. We, however, show for the first time in GH3 cells that a beta-adrenergic agonist activates a pertussis-toxin-sensitive G protein that activates c-PLA2. The arachidonic acid produced by c-PLA2 then activates BK channels. We examined BK channels in cell-attached patches and in excised patches from untreated GH3 cells and from GH3 cells exposed to c-PLA2 antisense oligonucleotides. For the cell-attached patch experiments, physiologic pipette and bath solutions were used. For the excised patches, 150 mM KCl was used in both the pipette and bath solutions, and the cytosolic surface contained 1 microM free Ca2+ (buffered with 5 mM K2EGTA). Treatment of GH3 cells with the G protein activator, fluoroaluminate, (AlF4-) produced an increase in the Po of BK channels of 177 +/- 41% (mean +/- SD) in cell-attached patches. Because G proteins are membrane associated, we also added an activator of G proteins, 100 microM GTP-gamma-S, to the cytosolic surface of excised patches. This treatment leads to an increase in Po of 50 +/- 9%. Similar treatment of excised patches with GDP-beta-S had no effect on Po. Isoproterenol (1 microM), an activator of beta-adrenergic receptors and, consequently, some G proteins, increased BK channel activity 229 +/- 37% in cell-attached patches from cultured GH3 cells. Western blot analysis showed that GH3 cells have beta-adrenergic receptor protein and that isoproterenol acts through these receptors because the beta-adrenergic receptor antagonist, propanolol, blocks the action of isoproterenol. To test whether G protein activation of BK channels involves c-PLA2, we studied the effects of GTP-gamma-S on excised patches and isoproterenol on cell attached patches from GH3 cells previously treated with c-PLA2 antisense oligonucleotides or pharmacological inhibitors of c-PLA2. Neither isoproterenol nor GTP-gamma-S had any effect on Po in these patches. Similarly, neither isoproterenol nor GTP-gamma-S had any effect on Po in cultured GH3 cells pretreated with pertussis toxin. Isoproterenol also significantly increased the rate of arachidonic production in GH3 cells. These results show that some receptor-linked, pertussis-toxin-sensitive G protein in GH3 cells can activate c-PLA2 to increase the amount of arachidonic acid present and ultimately increase BK-channel activity.


Assuntos
Proteínas de Ligação ao GTP/fisiologia , Fosfolipases A/metabolismo , Hipófise/metabolismo , Canais de Potássio Cálcio-Ativados/metabolismo , Transdução de Sinais/fisiologia , Agonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Compostos de Alumínio/farmacologia , Animais , Ácidos Araquidônicos/farmacologia , Cálcio/metabolismo , Linhagem Celular Tumoral , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Fluoretos/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Isoproterenol/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Modelos Biológicos , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Técnicas de Patch-Clamp/métodos , Fosfolipases A2 , Picrotoxina/farmacologia , Hipófise/efeitos dos fármacos , Hipófise/fisiologia , Propranolol/farmacologia , Ratos , Fatores de Tempo
4.
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
5.
Am J Physiol Cell Physiol ; 281(3): C773-85, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11502554

RESUMO

The A6 cell line was used to study the role of S-adenosyl-L-homocysteine hydrolase (SAHHase) in the aldosterone-induced activation of the epithelial Na(+) channel (ENaC). Because aldosterone increases methylation of several different molecules, and because this methylation is associated with increased Na(+) reabsorption, we tested the hypothesis that aldosterone increases the expression and activity of SAHHase protein. The rationale for this work is that general methylation may be promoted by activation of SAHHase, the only enzyme known to metabolize SAH, a potent end-product inhibitor of methylation. Although aldosterone increased SAHHase activity, steroid did not affect SAHHase expression. Antisense SAHHase oligonucleotide decreased SAHHase expression and activity. Moreover, this oligonucleotide, as well as a pharmacological inhibitor of SAHHase, decreased aldosterone-induced activity of ENaC via a decrease in ENaC open probability. The kinetics of ENaC in cells treated with antisense plus aldosterone were similar to those reported previously for the channel in the absence of steroid. This is the first report showing that active SAHHase, in part, increases ENaC open probability by reducing the transition rate from open states in response to aldosterone. Thus aldosterone-induced SAHHase activity plays a critical role in shifting ENaC from a gating mode with short open and closed times to one with longer open and closed times.


Assuntos
Aldosterona/farmacologia , Hidrolases/genética , Hidrolases/metabolismo , Rim/fisiologia , Canais de Sódio/fisiologia , Urotélio/fisiologia , Adenosil-Homocisteinase , Animais , Membrana Celular/fisiologia , Canais Epiteliais de Sódio , Isomerismo , Rim/citologia , Cinética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Metilação , Oligodesoxirribonucleotídeos/farmacologia , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Técnicas de Patch-Clamp , RNA Mensageiro/genética , Proteínas Recombinantes/metabolismo , Sódio/metabolismo , Canais de Sódio/efeitos dos fármacos , Transcrição Gênica , Transfecção , Tubercidina/farmacologia , Urotélio/citologia , Urotélio/efeitos dos fármacos , Xenopus laevis
6.
Am J Physiol Renal Physiol ; 281(2): F213-21, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11457713

RESUMO

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.


Assuntos
Células Epiteliais/metabolismo , Túbulos Renais/metabolismo , Canais de Sódio/metabolismo , Urotélio/metabolismo , Aldosterona/farmacologia , Animais , Linhagem Celular , Polaridade Celular , Centrifugação com Gradiente de Concentração , Células Epiteliais/efeitos dos fármacos , Canais Epiteliais de Sódio , Immunoblotting , Túbulos Renais/citologia , Túbulos Renais/efeitos dos fármacos , Testes de Precipitina , Subunidades Proteicas , Fatores de Tempo , Urotélio/citologia , Urotélio/efeitos dos fármacos , Xenopus laevis
7.
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
8.
J Biol Chem ; 276(10): 7136-42, 2001 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-11113145

RESUMO

To test the hypothesis that ATP activation of BK channels in GH(3) cells involves cytosolic phospholipase A(2) (cPLA(2)) as a potential protein target for phosphorylation, we first inhibited the activity of cPLA(2) by both pharmacologic and molecular biologic approaches. Both approaches resulted in a decrease rather than an increase in BK channel activity by ATP, suggesting that in the absence of cPLA(2), phosphorylation of other regulatory elements, possibly the BK channel protein itself, results in inactivation rather than activation of the channel. The absence of changes in activity in the presence of the non-substrate ATP analog 5'-adenylyl-beta,gamma-imidodiphosphate verified that ATP hydrolysis was required for channel activation by ATP. Experiments with an activator and inhibitor of protein kinase C (PKC) support the hypothesis that PKC can be involved in the activation of BK channels by ATP; and in the absence of PKC, other kinases appear to phosphorylate additional elements in the regulatory pathway that reduce channel activity. Our data point to cPLA(2)-alpha (but not cPLA(2)-gamma) as one target protein for phosphorylation that is intimately associated with the BK channel protein.


Assuntos
Trifosfato de Adenosina/metabolismo , Ácidos Aristolóquicos , Citosol/enzimologia , Fosfolipases A/metabolismo , Fosfolipases A/fisiologia , Canais de Potássio Cálcio-Ativados , Canais de Potássio/metabolismo , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Adenilil Imidodifosfato/farmacologia , Animais , Western Blotting , Linhagem Celular , Membrana Celular/enzimologia , Inibidores Enzimáticos/farmacologia , Hidrólise , Canais de Potássio Ativados por Cálcio de Condutância Alta , Oligonucleotídeos Antissenso/farmacologia , Técnicas de Patch-Clamp , Fenantrenos/farmacologia , Fosfolipases A/química , Fosfolipases A2 , Fosforilação , Isoformas de Proteínas , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Ratos
9.
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
10.
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
11.
Am J Physiol Cell Physiol ; 279(4): C1211-9, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11003601

RESUMO

Ca(2+)-activated K(+) (BK) channels in GH(3) cells are activated by arachidonic acid (AA). Because cytosolic phospholipase A(2) can produce other unsaturated free fatty acids (FFA), we examined the effects of FFA on BK channels in excised patches. Control recordings were made at several holding potentials. The desired FFA was added to the bath solution, and the voltage paradigm was repeated. AA increased the activity of BK channels by 3.6 +/- 1.6-fold. The cis FFA, palmitoleic, oleic, linoleic, linolenic, eicosapentaenoic, and the triple bond analog of AA, eicosatetraynoic acid, all increased BK channel activity, whereas stearic (saturated) or the trans isomers elaidic, linolelaidic, and linolenelaidic had no effect. The cis unsaturated FFA shifted the open probability vs. voltage relationships to the left without a change in slope, suggesting no change in the sensitivity of the voltage sensor. Measurements of membrane fluidity showed no correlation between the change of membrane fluidity and the change in BK channel activation. In addition, AA effects on BK channels were unaffected in the presence of N-acetylcysteine. Arachidonyl-CoA, a membrane impermeable analog of AA, activates channels when applied to the cytosolic surface of excised patches, suggesting an effect of FFAs from the cytosolic surface of BK channels. Our data imply a direct interaction between cis FFA and the BK channel protein.


Assuntos
Ácidos Graxos/metabolismo , Sistemas Neurossecretores/metabolismo , Hipófise/metabolismo , Canais de Potássio Cálcio-Ativados , Canais de Potássio/metabolismo , Acetilcisteína/farmacologia , Acil Coenzima A/metabolismo , Acil Coenzima A/farmacologia , Animais , Ácido Araquidônico/metabolismo , Ácido Araquidônico/farmacologia , Linhagem Celular , Membrana Celular/metabolismo , Ácidos Graxos/farmacologia , Ácidos Graxos Insaturados/metabolismo , Ácidos Graxos Insaturados/farmacologia , Sequestradores de Radicais Livres/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta , Fluidez de Membrana/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Sistemas Neurossecretores/citologia , Sistemas Neurossecretores/efeitos dos fármacos , Técnicas de Patch-Clamp , Hipófise/citologia , Hipófise/efeitos dos fármacos , Canais de Potássio/efeitos dos fármacos , Ratos , Espécies Reativas de Oxigênio/metabolismo , Relação Estrutura-Atividade
12.
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
13.
J Biol Chem ; 275(22): 16550-9, 2000 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-10747971

RESUMO

We used single channel methods on A6 renal cells to study the regulation by methylation reactions of epithelial sodium channels. 3-Deazaadenosine (3-DZA), a methyltransferase blocker, produced a 5-fold decrease in sodium transport and a 6-fold decrease in apical sodium channel activity by decreasing channel open probability (P(o)). 3-Deazaadenosine also blocked the increase in channel open probability associated with addition of aldosterone. Sodium channel activity in excised "inside-out" patches usually decreased within 1-2 min; in the presence of S-adenosyl-l-methionine (AdoMet), activity persisted for 5-8 min. Sodium channel mean time open (t(open)) before and after patch excision was higher in the presence of AdoMet than in untreated excised patches but less than t(open) in cell-attached patches. Sodium channel activity in excised patches exposed to both AdoMet and GTP usually remained stable for more than 10 min, and P(o) and the number of active channels per patch were close to values in cell-attached patches from untreated cells. These findings suggest that a methylation reaction contributes to the activity of epithelial sodium channels in A6 cells and is directed to some regulatory element closely connected with the channel, whose activity also depends on the presence of intracellular GTP.


Assuntos
Canais de Sódio/metabolismo , Linhagem Celular , Células Epiteliais/metabolismo , Canais Epiteliais de Sódio , Guanosina Trifosfato/farmacologia , Metilação , Metiltransferases/farmacologia , Antagonistas de Receptores de Mineralocorticoides/farmacologia , S-Adenosilmetionina/farmacologia , Tubercidina/farmacologia
14.
J Biol Chem ; 274(50): 35449-54, 1999 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-10585415

RESUMO

Xenopus laevis A6 cells were used as model epithelia to test the hypothesis that K-Ras2A is an aldosterone-induced protein necessary for steroid-regulated Na(+) transport. The possibility that increased K-Ras2A alone is sufficient to mimic aldosterone action on Na(+) transport also was tested. Aldosterone treatment increased K-Ras2A protein expression 2.8-fold within 4 h. Active Ras is membrane associated. After aldosterone treatment, 75% of K-Ras was localized to the plasma membrane compared with 25% in the absence of steroid. Aldosterone also increased the amount of active (phosphorylated) mitogen-activated protein kinase kinase likely through K-Ras2A signaling. Steroid-induced K-Ras2A protein levels and Na(+) transport were decreased with antisense K-ras2A oligonucleotides, showing that K-Ras2A is necessary for the natriferic actions of aldosterone. Aldosterone-induced Na(+) channel activity, was decreased from 0.40 to 0.09 by pretreatment with antisense ras oligonucleotide, implicating the luminal Na(+) channel as one final effector of Ras signaling. Overexpression of K-Ras2A increased Na(+) transport approximately 2.2-fold in the absence of aldosterone. These results suggest that aldosterone signals to the luminal Na(+) channel via multiple pathways and that K-Ras2A levels are limiting for a portion of the aldosterone-sensitive Na(+) transport.


Assuntos
Aldosterona/farmacologia , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Sódio/metabolismo , Animais , Transporte Biológico , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Epiteliais/fisiologia , Canais Epiteliais de Sódio , Regulação da Expressão Gênica/efeitos dos fármacos , Cinética , Modelos Biológicos , Transdução de Sinais , Canais de Sódio/metabolismo , Xenopus laevis
15.
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
16.
J Physiol ; 515 ( Pt 3): 669-83, 1999 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-10066896

RESUMO

1. In cell-attached patches formed on the apical membrane of fetal alveolar epithelium, terbutaline (a specific beta2-adrenergic agonist) increased the open probability (Po) of an amiloride-sensitive Na+-permeable non-selective cation (NSC) channel (control, 0.03 +/- 0.04; terbutaline, 0.62 +/- 0.18; n = 8, P < 0. 00001) by increasing the mean open time 100-fold without any significant change in the mean closed time and without any change in the single channel conductance (control, 27.8 +/- 2.3 pS; terbutaline, 28.2 +/- 2.1 pS; n = 8). 2. The Po of the unstimulated channel increased when the apical membrane was depolarized due to a decrease in the closing rate and an increase in the opening rate, while the Po of the terbutaline-stimulated channel did not depend on the membrane potential. 3. Increased cytosolic [Ca2+] also increased the Po of the channel in a manner consistent with one Ca2+-binding site on the cytosolic surface of the channel. Terbutaline increased the sensitivity of the channel to cytosolic Ca2+ by shifting the concentration of cytosolic Ca2+ ([Ca2+]c) required for half-maximal activation to a lower [Ca2+]c value, leading to an increase in Po. 4. An increase in the cytosolic Cl- concentration ([Cl-]c) decreased the Po of the channel consistent with two Cl--binding sites by increasing the closing rate without any significant change in the opening rate. Terbutaline increased Po by reducing the effect of cytosolic Cl- to promote channel closing. 5. Taken together, these observations indicate that terbutaline activates a Ca2+-activated, Cl--inhibitable, amiloride-sensitive, Na+-permeable NSC channel in fetal rat alveolar epithelium in two ways: first, through an increase in Ca2+ sensitivity, and second, through a reduction in the effect of cytosolic Cl- to promote channel closing.


Assuntos
Amilorida/farmacologia , Cálcio/metabolismo , Cloretos/metabolismo , Células Epiteliais/fisiologia , Alvéolos Pulmonares/fisiologia , Canais de Sódio/fisiologia , Terbutalina/farmacologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Células Cultivadas , Citosol/metabolismo , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Feto , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Modelos Biológicos , Técnicas de Patch-Clamp , Alvéolos Pulmonares/citologia , Ratos , Ratos Wistar , Canais de Sódio/efeitos dos fármacos
17.
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
18.
Am J Physiol ; 275(5): C1216-23, 1998 11.
Artigo em Inglês | MEDLINE | ID: mdl-9814969

RESUMO

Angiotensin II (ANG II) exerts its effects on vascular smooth muscle cells through G protein-coupled AT1 receptors. ANG II stimulation activates the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway by inducing tyrosine phosphorylation, activation, and association of JAK2 with the receptor. Association appears to be required for JAK2 phosphorylation. In the present study, electroporation experiments with neutralizing anti-Src homology phosphatase-1 (SHP-1) and anti-SHP-2 antibodies and time course determinations of SHP-1 and SHP-2 activation and complexation with JAK2 suggest that the tyrosine phosphatases, SHP-1 and SHP-2, have opposite roles in ANG II-induced JAK2 phosphorylation. SHP-1 appears responsible for JAK2 dephosphorylation and termination of the ANG II-induced JAK/STAT cascade. SHP-2 appears to have an essential role in JAK2 phosphorylation and initiation of the ANG II-induced JAK/STAT cascade leading to cell proliferation. The motif in the AT1 receptor that is required for association with JAK2 is also required for association with SHP-2. Furthermore, SHP-2 is required for JAK2-receptor association. SHP-2 may thus play a role as an adaptor protein for JAK2 association with the receptor, thereby facilitating JAK2 phosphorylation and activation.


Assuntos
Angiotensina II/farmacologia , Coração/fisiologia , Miocárdio/enzimologia , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas , Transdução de Sinais/fisiologia , Substituição de Aminoácidos , Animais , Anticorpos/farmacologia , Células Cultivadas , Eletroporação , Ventrículos do Coração , Peptídeos e Proteínas de Sinalização Intracelular , Janus Quinase 2 , Masculino , Miocárdio/citologia , Fosforilação , Fosfotirosina/metabolismo , Mutação Puntual , Proteína Fosfatase 1 , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/genética , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina , Receptor Tipo 2 de Angiotensina , Receptores de Angiotensina/efeitos dos fármacos , Receptores de Angiotensina/fisiologia , Proteínas Recombinantes/metabolismo , Proteínas Tirosina Fosfatases Contendo o Domínio SH2 , Transdução de Sinais/efeitos dos fármacos , Domínios de Homologia de src
19.
J Biol Chem ; 273(46): 30795-800, 1998 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-9804857

RESUMO

Angiotensin II (Ang II) AT1 receptors on vascular smooth muscle cells (VSMCs) are coupled to the Janus-activated kinase (JAK)/signal transducers and activators of transcription (STAT) pathway. We have shown previously that Ang II stimulation of VSMCs results in the tyrosine phosphorylation of JAK2 and STAT1 and the translocation of STAT1 to the nucleus. In the present study, we demonstrate using specific enzyme inhibitors and antisense oligonucleotides that both JAK2 and p59 Fyn tyrosine kinases are required for the Ang II-induced tyrosine phosphorylation and nuclear translocation of STAT1 in VSMCs. Neither tyrosine kinase, however, appears to function upstream from the other in a phosphorylation cascade. Rather, p59 Fyn functions as an Ang II-activated docking protein for both JAK2 and STAT1, a docking interaction that may facilitate JAK2-mediated STAT1 tyrosine phosphorylation. In this study, we have also identified the nuclear dual-specificity phosphatase mitogen-activated protein kinase phosphatase 1 as the enzyme responsible for STAT1 tyrosine dephosphorylation in VSMCs.


Assuntos
Angiotensina II/farmacologia , Proteínas de Ciclo Celular , Proteínas Imediatamente Precoces/metabolismo , Fosfoproteínas Fosfatases , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Transativadores , Tirosina/metabolismo , Animais , Proteínas de Ligação a DNA/metabolismo , Fosfatase 1 de Especificidade Dupla , Proteínas Imediatamente Precoces/genética , Janus Quinase 2 , Masculino , Músculo Liso Vascular/enzimologia , Oligonucleotídeos Antissenso/metabolismo , Fosforilação , Proteína Fosfatase 1 , Proteínas Tirosina Fosfatases/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-fyn , Ratos , Ratos Sprague-Dawley , Fator de Transcrição STAT1 , Transativadores/metabolismo
20.
Kidney Int ; 53(5): 1259-68, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9573541

RESUMO

Erythropoietin (EPO) increases Ca2+ influx in vascular smooth muscle cells and acts both as a direct vasoconstrictor and vascular growth factor (that is, angiogenesis). However, the mechanism by which EPO promotes extracellular Ca2+ entry in contractile cells has not been elucidated. In hematopoietic cells, EPO induces tyrosine kinase (TK)-dependent activation of phospholipase C (PLC)-gamma 1 and Ca2+ influx via a voltage-independent Ca2+ conductance. In contractile mesangial cells, we have recently characterized a voltage-independent, 1 pS Ca2+ channel that is dependent on both TK and PLC-gamma 1 activity. Therefore, we examined cultured rat glomerular mesangial cells after timed exposure to recombinant human EPO (20 U/ml). Erythropoietin increased the tyrosine phosphorylation of PLC-gamma 1, promoted membrane complex formation between PLC-gamma 1 and the EPO receptor itself, and raised the levels of intracellular inositol 1,4,5-trisphosphate and intracellular Ca2+. Consistent with our previous studies, 1 pS Ca2+ channel activity was extremely low under basal, unstimulated conditions in cell-attached patches, but was dramatically increased when EPO was present in the patch pipette. Tyrosine kinase inhibition with 100 micron genistein or 1 micron PP1 (Src; selective tyrosine kinase inhibitor) prevented all of these EPO-induced responses. We conclude that: (1) EPO-induced stimulation of 1 pS Ca2+ channels is mediated via a cytosolic Src TK in glomerular mesangial cells. (2) Stimulation of this Ca2(+)-activated, Ca2(+)-permeable channel is dependent on the tyrosine phosphorylation/activation of PLC-gamma 1. (3) This cascade provides a possible mechanism for the vasoconstriction and hypertension observed with clinical EPO use for the treatment of chronic anemias.


Assuntos
Canais de Cálcio/metabolismo , Isoenzimas/metabolismo , Receptores da Eritropoetina/metabolismo , Fosfolipases Tipo C/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Eritropoetina/farmacologia , Eritropoetina/toxicidade , Mesângio Glomerular/efeitos dos fármacos , Mesângio Glomerular/metabolismo , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Líquido Intracelular/metabolismo , Modelos Biológicos , Fosfolipase C gama , Fosforilação , Ratos , Proteínas Recombinantes , Tirosina/metabolismo , Quinases da Família src/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA