Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Antioxidants (Basel) ; 10(7)2021 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-34203453

RESUMO

(1) Background: One third of patients who receive cisplatin develop an acute kidney injury. We previously demonstrated the Na/H Exchange Regulatory Factor 1 (NHERF1) loss resulted in increased kidney enzyme activity of the pentose phosphate pathway and was associated with more severe cisplatin nephrotoxicity. We hypothesized that changes in proximal tubule biochemical pathways associated with NHERF1 loss alters renal metabolism of cisplatin or response to cisplatin, resulting in exacerbated nephrotoxicity. (2) Methods: 2-4 month-old male wild-type and NHERF1 knock out littermate mice were treated with either vehicle or cisplatin (20 mg/kg dose IP), with samples taken at either 4, 24, or 72 h. Kidney injury was determined by urinary neutrophil gelatinase-associated lipocalin and histology. Glutathione metabolites were measured by HPLC and genes involved in glutathione synthesis were measured by qPCR. Kidney handling of cisplatin was assessed by a kidney cortex measurement of γ-glutamyl transferase activity, Western blot for γ-glutamyl transferase and cysteine S-conjugate beta lyase, and ICP-MS for platinum content. (3) Results: At 24 h knock out kidneys show evidence of greater tubular injury after cisplatin and exhibit a decreased reduced/oxidized glutathione ratio under baseline conditions in comparison to wild-type. KO kidneys fail to show an increase in γ-glutamyl transferase activity and experience a more rapid decline in tissue platinum when compared to wild-type. (4) Conclusions: Knock out kidneys show evidence of greater oxidative stress than wild-type accompanied by a greater degree of early injury in response to cisplatin. NHERF1 loss has no effect on the initial accumulation of cisplatin in the kidney cortex but is associated with an altered redox status which may alter the activity of enzymes involved in cisplatin metabolism.

2.
Antioxidants (Basel) ; 9(9)2020 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-32937931

RESUMO

(1) Background: We previously showed Na/H exchange regulatory factor 1 (NHERF1) loss resulted in increased susceptibility to cisplatin nephrotoxicity. NHERF1-deficient cultured proximal tubule cells and proximal tubules from NHERF1 knockout (KO) mice exhibit altered mitochondrial protein expression and poor survival. We hypothesized that NHERF1 loss results in changes in metabolic pathways and/or mitochondrial dysfunction, leading to increased sensitivity to cisplatin nephrotoxicity. (2) Methods: Two to 4-month-old male wildtype (WT) and KO mice were treated with vehicle or cisplatin (20 mg/kg dose IP). After 72 h, kidney cortex homogenates were utilized for metabolic enzyme activities. Non-treated kidneys were used to isolate mitochondria for mitochondrial respiration via the Seahorse XF24 analyzer. Non-treated kidneys were also used for LC-MS analysis to evaluate kidney ATP abundance, and electron microscopy (EM) was utilized to evaluate mitochondrial morphology and number. (3) Results: KO mouse kidneys exhibit significant increases in malic enzyme and glucose-6 phosphate dehydrogenase activity under baseline conditions but in no other gluconeogenic or glycolytic enzymes. NHERF1 loss does not decrease kidney ATP content. Mitochondrial morphology, number, and area appeared normal. Isolated mitochondria function was similar between WT and KO. Conclusions: KO kidneys experience a shift in metabolism to the pentose phosphate pathway, which may sensitize them to the oxidative stress imposed by cisplatin.

3.
Compr Physiol ; 6(2): 561-601, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-27065162

RESUMO

PTH and Vitamin D are two major regulators of mineral metabolism. They play critical roles in the maintenance of calcium and phosphate homeostasis as well as the development and maintenance of bone health. PTH and Vitamin D form a tightly controlled feedback cycle, PTH being a major stimulator of vitamin D synthesis in the kidney while vitamin D exerts negative feedback on PTH secretion. The major function of PTH and major physiologic regulator is circulating ionized calcium. The effects of PTH on gut, kidney, and bone serve to maintain serum calcium within a tight range. PTH has a reciprocal effect on phosphate metabolism. In contrast, vitamin D has a stimulatory effect on both calcium and phosphate homeostasis, playing a key role in providing adequate mineral for normal bone formation. Both hormones act in concert with the more recently discovered FGF23 and klotho, hormones involved predominantly in phosphate metabolism, which also participate in this closely knit feedback circuit. Of great interest are recent studies demonstrating effects of both PTH and vitamin D on the cardiovascular system. Hyperparathyroidism and vitamin D deficiency have been implicated in a variety of cardiovascular disorders including hypertension, atherosclerosis, vascular calcification, and kidney failure. Both hormones have direct effects on the endothelium, heart, and other vascular structures. How these effects of PTH and vitamin D interface with the regulation of bone formation are the subject of intense investigation.


Assuntos
Doenças das Paratireoides/metabolismo , Hormônio Paratireóideo/metabolismo , Deficiência de Vitamina D/metabolismo , Vitamina D/metabolismo , Animais , Cálcio/metabolismo , Fator de Crescimento de Fibroblastos 23 , Humanos , Hormônio Paratireóideo/sangue , Hormônio Paratireóideo/química , Receptores de Calcitriol/metabolismo , Receptores de Hormônios Paratireóideos/metabolismo , Vitamina D/sangue
4.
Am J Physiol Cell Physiol ; 307(2): C169-79, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24829496

RESUMO

Inflammation-induced vascular endothelial dysfunction can allow plasma proteins to cross the vascular wall, causing edema. Proteins may traverse the vascular wall through two main pathways, the paracellular and transcellular transport pathways. Paracellular transport involves changes in endothelial cell junction proteins, while transcellular transport involves caveolar transcytosis. Since both processes are associated with filamentous actin formation, the two pathways are interconnected. Therefore, it is difficult to differentiate the prevailing role of one or the other pathway during various pathologies causing an increase in vascular permeability. Using a newly developed dual-tracer probing method, we differentiated transcellular from paracellular transport during hyperfibrinogenemia (HFg), an increase in fibrinogen (Fg) content. Roles of cholesterol and sphingolipids in formation of functional caveolae were assessed using a cholesterol chelator, methyl-ß-cyclodextrin, and the de novo sphingolipid synthesis inhibitor myriocin. Fg-induced formation of functional caveolae was defined by association and colocalization of Na+-K+-ATPase and plasmalemmal vesicle-associated protein-1 with use of Förster resonance energy transfer and total internal reflection fluorescence microscopy, respectively. HFg increased permeability of the endothelial cell layer mainly through the transcellular pathway. While MßCD blocked Fg-increased transcellular and paracellular transport, myriocin affected only transcellular transport. Less pial venular leakage of albumin was observed in myriocin-treated HFg mice. HFg induced greater formation of functional caveolae, as indicated by colocalization of Na+-K+-ATPase with plasmalemmal vesicle-associated protein-1 by Förster resonance energy transfer and total internal reflection fluorescence microscopy. Our results suggest that elevated blood levels of Fg alter cerebrovascular permeability mainly by affecting caveolae-mediated transcytosis through modulation of de novo sphingolipid synthesis.


Assuntos
Encéfalo/irrigação sanguínea , Permeabilidade Capilar/fisiologia , Cavéolas/metabolismo , Fibrinogênio/metabolismo , Esfingolipídeos/farmacologia , Animais , Colesterol/metabolismo , Cromatografia Líquida , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Fibrinogênio/genética , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Esfingolipídeos/metabolismo , Espectrometria de Massas em Tandem , Transcitose , Veias/efeitos dos fármacos , Veias/fisiologia
5.
J Endocrinol ; 222(1): R11-24, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24781255

RESUMO

Cardiotonic steroids have been used for the past 200 years in the treatment of congestive heart failure. As specific inhibitors of membrane-bound Na(+)/K(+) ATPase, they enhance cardiac contractility through increasing myocardial cell calcium concentration in response to the resulting increase in intracellular Na concentration. The half-minimal concentrations of cardiotonic steroids required to inhibit Na(+)/K(+) ATPase range from nanomolar to micromolar concentrations. In contrast, the circulating levels of cardiotonic steroids under physiological conditions are in the low picomolar concentration range in healthy subjects, increasing to high picomolar levels under pathophysiological conditions including chronic kidney disease and heart failure. Little is known about the physiological function of low picomolar concentrations of cardiotonic steroids. Recent studies have indicated that physiological concentrations of cardiotonic steroids acutely stimulate the activity of Na(+)/K(+) ATPase and activate an intracellular signaling pathway that regulates a variety of intracellular functions including cell growth and hypertrophy. The effects of circulating cardiotonic steroids on renal salt handling and total body sodium homeostasis are unknown. This review will focus on the role of low picomolar concentrations of cardiotonic steroids in renal Na(+)/K(+) ATPase activity, cell signaling, and blood pressure regulation.


Assuntos
Glicosídeos Cardíacos/metabolismo , Túbulos Renais/metabolismo , Sódio/metabolismo , Transporte Biológico/fisiologia , Pressão Sanguínea/fisiologia , Humanos , Transdução de Sinais/fisiologia , ATPase Trocadora de Sódio-Potássio/metabolismo
6.
Am J Physiol Renal Physiol ; 294(4): F840-9, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18216150

RESUMO

Several lines of evidence show that sodium/hydrogen exchanger regulatory factor 1 (NHERF-1) regulates the expression and activity of the type IIa sodium-dependent phosphate transporter (Npt2a) in renal proximal tubules. We have previously demonstrated that expression of a COOH-terminal ezrin binding domain-deficient NHERF-1 in opossum kidney (OK) cells decreased expression of Npt2a in apical membranes but did not affect responses to parathyroid hormone. We hypothesized that NHERF-1 regulates apical membrane expression of Npt2a in renal proximal tubule cells. To address this hypothesis, we compared regulation of Npt2a expression and function in NHERF-deficient OK cells (OK-H) and wild-type cells (OK-WT). In OK-H cells, phosphate uptake and expression of Npt2a protein in apical membranes were significantly lower than in OK-WT cells. Transient transfection of green fluorescent protein-tagged Npt2a cDNA into OK-H cells resulted in aberrant localization of an Npt2a fragment to the cytosol but not to the apical membrane. OK-H cells also exhibited a marked decrease in Npt2a mRNA expression. As demonstrated by luciferase assay, Npt2a promoter activity was significantly decreased in OK-H cells compared with that shown in OK-WT cells. Transfection of OK-H cells with human NHERF-1 restored Npt2a expression at both the protein and mRNA levels and regulation by parathyroid hormone. Expression of NHERF-1 constructs with mutations in the PDZ domains or the ezrin binding domain in OK-H cells suggested that the PDZ2 domain is critical for apical translocation of Npt2a and for expression at the mRNA level. Our data demonstrate for the first time that NHERF-1 regulates Npt2a transcription and membrane insertion.


Assuntos
Fosfoproteínas/fisiologia , Trocadores de Sódio-Hidrogênio/fisiologia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/genética , Animais , Linhagem Celular , Primers do DNA , DNA Complementar/genética , Genes Reporter , Rim , Gambás , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/deficiência , Transfecção
7.
Br J Nutr ; 98(5): 984-90, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17764602

RESUMO

Ramadan fasting is a unique model of fasting in which Muslims the world over abstain from food and water from dawn to sunset for 1 month. We hypothesized that this model of prolonged intermittent fasting would result in specific adaptive alterations in rat kidney to keep a positive balance of metabolites and inorganic phosphate (Pi). The effect of Ramadan-type fasting was studied on enzymes of carbohydrate metabolism and brush border membrane (BBM) and BBM uptake of 32Pi in different renal tissue zones in the rat model. Rats were fasted (12 h) and then re-fed (12 h) daily for 30 d similar to human Ramadan fasting. Ramadan-type fasting resulted in increased serum Pi and phospholipids, whereas Pi clearance decreased. Serum creatinine and its clearance were not affected. Fasting caused a significant decrease in the activities of lactate and malate dehydrogenases, glucose-6-phosphatase and fructose-1,6-bisphosphatase, both in the renal cortex and medulla. However, the activity of glucose-6-phosphate dehydrogenase profoundly increased but that of malic enzyme decreased. The activities of alkaline phosphatase and gamma-glutamyl transpeptidase in BBM decreased, whereas transport of 32Pi significantly increased. The decrease in enzyme activities and increase in 32Pi transport were due to alterations of both maximal velocities and relative affinities. The results indicate that Ramadan-type fasting caused specific metabolic alterations with enhanced Pi conservation in different kidney tissues in a rat model used for Ramadan fasting in man.


Assuntos
Metabolismo dos Carboidratos/fisiologia , Jejum/fisiologia , Islamismo , Rim/metabolismo , Religião e Medicina , Fenômenos Fisiológicos da Nutrição Animal/fisiologia , Animais , Transporte Biológico Ativo/fisiologia , Ritmo Circadiano/fisiologia , Rim/ultraestrutura , Córtex Renal/enzimologia , Medula Renal/enzimologia , Microvilosidades/enzimologia , Modelos Animais , Fosfatos/metabolismo , Ratos , Ratos Wistar
8.
J Am Soc Nephrol ; 16(9): 2598-607, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16000700

RESUMO

It was demonstrated that expression of murine sodium hydrogen exchanger regulatory factor (NHERF-1) lacking the ezrin-binding domain blocks parathyroid hormone (PTH) regulation of Na+,K+-ATPase in opossum kidney (OK) cells. The hypothesis that the NHERF-1 PDZ domains contribute to PTH regulation of Na+,K+-ATPase was tested by comparison of PTH regulation of Na+,K+-ATPase in wild-type OK (OK-WT) cells, NHERF-deficient OKH cells, OK-WT transfected with siRNA for NHERF (NHERF siRNA OK-WT), and OKH cells that were stably transfected with full-length NHERF-1 or constructs with mutated PDZ domains. OKH cells and NHERF siRNA OK-WT showed decreased expression of NHERF-1 but equivalent expression of ezrin and Na+,K+-ATPase alpha1 subunit when compared with OK-WT cells. PTH decreased Na+,K+-ATPase activity and stimulated phosphorylation of the Na+,K+-ATPase alpha1 in OK-WT cells but not in NHERF-deficient cells. Rubidium (86Rb) uptake was equivalent in OK-WT, OKH, and OKH cells that were transfected with all but the double PDZ domain mutants. PTH decreased 86Rb uptake significantly in OK-WT but not in OKH cells. PTH also significantly inhibited 86Rb uptake in OKH cells that were transfected with full-length NHERF-1 or NHERF-1 with mutated PDZ 2 but not in OKH cells that were transfected with mutated PDZ 1. Transfection with NHERF expressing both mutated PDZ domains resulted in diminished basal 86Rb uptake that was not inhibited further by PTH. PTH stimulated protein kinase Calpha activity and alpha1 subunit phosphorylation in OK-WT but not in NHERF-deficient cells. Transfection of OKH cells with NHERF constructs that contained an intact PDZ1 domain restored PTH-stimulated protein kinase Calpha activity and alpha1 subunit phosphorylation. These results demonstrate that NHERF-1 is necessary for PTH-mediated inhibition of Na+,K+-ATPase activity and that the inhibition is mediated through the PDZ1, not PDZ2, domain.


Assuntos
Hormônio Paratireóideo/farmacologia , Fosfoproteínas/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Rim/citologia , Rim/efeitos dos fármacos , Rim/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Gambás , Fosfoproteínas/química , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Proteína Quinase C-alfa/metabolismo , Estrutura Terciária de Proteína , Subunidades Proteicas , RNA Interferente Pequeno/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trocadores de Sódio-Hidrogênio/química , Trocadores de Sódio-Hidrogênio/genética , ATPase Trocadora de Sódio-Potássio/química , Transfecção
9.
J Biol Chem ; 279(17): 17418-27, 2004 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-14976217

RESUMO

Parathyroid hormone (PTH) inhibits Na(+),K(+)-ATPase activity through protein kinase C- (PKC) and extracellular signal-regulated kinase- (ERK) dependent pathways and increases serine phosphorylation of the alpha(1)-subunit. To determine whether specific serine phosphorylation sites within the Na(+),K(+)-ATPase alpha(1)-subunit are involved in the Na(+),K(+)-ATPase responses to PTH, we examined the effect of PTH in opossum kidney cells stably transfected with wild type rat Na(+),K(+)-ATPase alpha(1)-subunit (WT), serine 11 to alanine mutant alpha(1)-subunit (S11A), or serine 18 to alanine mutant alpha(1)-subunit (S18A). PTH increased phosphorylation and endocytosis of the Na(+),K(+)-ATPase alpha(1)-subunit into clathrin-coated vesicles in cells transfected with WT and S18A rat Na(+),K(+)-ATPase alpha(1)-subunits. PTH did not increase the level of phosphorylation or stimulate translocation of Na(+),K(+)-ATPase alpha(1)-subunits into clathrin-coated vesicles in cells transfected with the S11A mutant. PTH inhibited ouabain-sensitive (86)Rb uptake and Na(+),K(+)-ATPase activity (ouabain-sensitive ATP hydrolysis) in WT- and S18A-transfected opossum kidney cells but not in S11A-transfected cells. Pretreatment of the cells with the PKC inhibitors and ERK inhibitor blocked PTH inhibition of (86)Rb uptake, Na(+),K(+)-ATPase activity, alpha(1)-subunit phosphorylation, and endocytosis in WT and S18A cells. Consistent with the notion that ERK phosphorylates Na(+),K(+)-ATPase alpha(1)-subunit, ERK was shown to be capable of causing phosphorylation of Na(+),K(+)-ATPase alpha(1)-subunit immunoprecipitated from WT and S18A but not from S11A-transfected cells. These results suggest that PTH regulates Na(+),K(+)-ATPase by PKC and ERK-dependent alpha(1)-subunit phosphorylation and that the phosphorylation requires the expression of a serine at the 11 position of the Na(+),K(+)-ATPase alpha(1)-subunit.


Assuntos
Clatrina/metabolismo , Endocitose , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Hormônio Paratireóideo/metabolismo , Serina/química , ATPase Trocadora de Sódio-Potássio/química , Alanina/química , Animais , Butadienos/farmacologia , Bovinos , Membrana Celular/metabolismo , DNA Complementar/metabolismo , Endossomos/metabolismo , Rim/metabolismo , Mutação , Nitrilas/farmacologia , Gambás , Fosforilação , Testes de Precipitina , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/química , Estrutura Terciária de Proteína , Transporte Proteico , Ratos , ATPase Trocadora de Sódio-Potássio/metabolismo , Estaurosporina/farmacologia , Fatores de Tempo , Transfecção
10.
J Am Soc Nephrol ; 14(7): 1711-9, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12819230

RESUMO

Parathyroid hormone (PTH), acting at least in part through a cAMP signaling pathway, regulates three important transporters in the renal proximal convoluted tubule, namely Na-H exchanger 3, Na-K ATPase, and type IIa sodium phosphate cotransporter (NaPi IIa). The regulation of Na-H exchanger 3 by protein kinase A requires a protein co-factor from the sodium-hydrogen exchanger regulatory factor (NHERF) family of proteins (NHERF-1 and NHERF-2). However, the role of NHERF in PTH regulation of Na-K ATPase and NaPi IIa has not been explored. For studying the role of NHERF-1 on PTH regulation of these transporters, wild-type mNHERF-1 (1-355) or mNHERF-1 (1-325) lacking the ezrin-binding domain were expressed in proximal tubule-derived opossum kidney cells. PTH inhibited Na-K ATPase activity in cells expressing wild-type NHERF-1 associated with increased serine phosphorylation of the alpha subunit of the transporter. By contrast, in cells expressing mNHERF (1-325), the phosphorylation of the alpha subunit of Na-K ATPase was blunted and the activity of the transporter was stimulated in response to PTH. Basal sodium-dependent phosphate transport was lower in cells expressing mNHERF-1 (1-325) as compared with cells expressing mNHERF-1 (1-355). Nonetheless, there were no differences in PTH-associated inhibition of the activity or the decrease in membrane expression of the NaPi IIa in any of the cell lines. These experiments document for the first time an association between NHERF-1 and PTH regulation of Na-K ATPase in epithelial cells. These experiments also suggest that the mechanism for retrieval of NaPi IIa transporters from the apical membrane in response to cAMP does not require NHERF.


Assuntos
Células Epiteliais/metabolismo , Regulação Enzimológica da Expressão Gênica , Fosfoproteínas/fisiologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Transporte Biológico , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Proteínas do Citoesqueleto , Glutationa/metabolismo , Glutationa Transferase/metabolismo , Immunoblotting , Rim/citologia , Camundongos , Mutação , Gambás , Hormônio Paratireóideo/metabolismo , Fosfatos/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Fosforilação , Plasmídeos/metabolismo , Testes de Precipitina , Estrutura Terciária de Proteína , Transdução de Sinais , Trocadores de Sódio-Hidrogênio , Transfecção
11.
J Biol Chem ; 278(12): 10134-41, 2003 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-12496250

RESUMO

Parathyroid hormone inhibits sodium-phosphate cotransport in proximal renal tubule cells through activation of several kinases. We tested the hypothesis that the activity of these kinases was coordinated by an A kinase anchoring protein (AKAP) by demonstrating that the type II sodium-phosphate cotransporter (NaPi-4) physically associated with an AKAP and that this association was necessary for regulation of phosphate transport by parathyroid hormone. Immunoprecipitation with anti-NaPi-4 antiserum and glutathione S-transferase pull-down with GST-NaPi-4 showed that NaPi-4 associated with AKAP79, protein kinase A catalytic and regulatory subunits, and the parathyroid hormone receptor in opossum kidney cells. When the regulatory subunit of protein kinase A was uncoupled from the AKAP by a competing peptide, parathyroid hormone lost the ability to inhibit phosphate transport. This result was confirmed by co-transfecting HEK293 cells with the sodium-phosphate cotransporter and wild type AKAP, a mutant AKAP79, or the empty vector. 8-Bromo-cAMP was able to inhibit phosphate transport in cells expressing the wild type AKAP79 but not empty vector or mutant AKAP79. We conclude that parathyroid hormone inhibits proximal renal tubule sodium-phosphate cotransport through a signaling complex dependent upon an AKAP.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte/fisiologia , Túbulos Renais Proximais/metabolismo , Hormônio Paratireóideo/fisiologia , Simportadores/fisiologia , Proteínas de Ancoragem à Quinase A , Sequência de Aminoácidos , Animais , Células Cultivadas , Proteína Quinase Tipo II Dependente de AMP Cíclico , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Humanos , Dados de Sequência Molecular , Gambás , Proteína Quinase C/fisiologia , Receptores de Hormônios Paratireóideos/fisiologia , Proteínas Cotransportadoras de Sódio-Fosfato , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II , Simportadores/química
12.
Am J Physiol Renal Physiol ; 282(3): F512-22, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11832434

RESUMO

Parathyroid hormone (PTH) and dopamine (DA) inhibit Na-K ATPase activity and sodium-phosphate cotransport in proximal tubular cells. We previously showed that PTH and DA inhibit phosphate transport in opossum kidney (OK) cells through different signaling pathways. Therefore, we hypothesized that PTH and DA also inhibit Na-K ATPase through divergent pathways. We measured PTH and DA inhibition of Na-K ATPase activity in the presence of inhibitors of signaling pathways. PTH and DA inhibited Na-K ATPase in a biphasic manner, the early inhibition through protein kinase C (PKC)- and phospholipase A(2) (PLA(2))-dependent pathways and the late inhibition through protein kinase A- and PLA(2)-dependent pathways. Inhibition of extracellular signal-regulated kinase (ERK) activation blocked early and late inhibition of Na-K ATPase by PTH but not by DA. Pertussis toxin blocked early and late inhibition by DA but not by PTH. Treatment with DA, but not PTH, resulted in an early downregulation of basolateral membrane expression of the alpha-subunit, whereas total cellular expression remained constant for both agonists. We conclude that PTH and DA regulate Na-K ATPase by different mechanisms through activation of divergent pathways.


Assuntos
Dopamina/farmacologia , Rim/enzimologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Sulfonamidas , Animais , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Antagonistas de Dopamina/farmacologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Isoquinolinas/farmacologia , Rim/citologia , Sistema de Sinalização das MAP Quinases/fisiologia , Naftalenos/farmacologia , Gambás , Toxina Pertussis , Inibidores de Fosfodiesterase/farmacologia , Fosfolipases A/metabolismo , Proteína Quinase C/metabolismo , Pironas/farmacologia , Fatores de Virulência de Bordetella/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...