Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Am J Physiol Renal Physiol ; 317(6): F1536-F1548, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31588796

RESUMO

The sodium-chloride cotransporter (NCC) in the distal convoluted tubule contributes importantly to sodium balance and blood pressure (BP) regulation. NCC phosphorylation determines transport activity and has a diurnal rhythm influenced by glucocorticoids. Disturbing this rhythm induces "nondipping" BP, an abnormality that increases cardiovascular risk. The receptor through which glucocorticoids regulate NCC is not known. In this study, we found that acute administration of corticosterone to male C57BL6 mice doubled NCC phosphorylation without affecting total NCC abundance in both adrenalectomized and adrenal-intact mice. Corticosterone also increased the whole kidney expression of canonical clock genes: period circadian protein homolog 1 (Per1), Per2, cryptochrome 1, and aryl hydrocarbon receptor nuclear translocator-like protein 1. In adrenal-intact mice, chronic blockade of glucocorticoid receptor (GR) with RU486 did not change total NCC but prevented corticosterone-induced NCC phosphorylation and activation of clock genes. Blockade of mineralocorticoid receptor (MR) with spironolactone reduced the total pool of NCC but did not affect stimulation by corticosterone. The diurnal rhythm of NCC phosphorylation, measured at 6-h intervals, was blunted by chronic GR blockade, and a similar dampening of diurnal variation was seen in GR heterozygous null mice. These effects on NCC phosphorylation did not reflect altered rhythmicity of plasma corticosterone or serum and glucocorticoid-induced kinase 1 activity. Both mineralocorticoids and glucocorticoids emerge as regulators of NCC, acting via distinct receptor pathways. MR activation provides maintenance of the NCC protein pool; GR activation dynamically regulates NCC phosphorylation and establishes the diurnal rhythm of NCC activity. This study has implications for circadian BP homeostasis, particularly in individuals with abnormal glucocorticoid signaling as is found in chronic stress and corticosteroid therapy.


Assuntos
Ritmo Circadiano/fisiologia , Rim/metabolismo , Receptores de Glucocorticoides/metabolismo , Simportadores de Cloreto de Sódio/metabolismo , Adrenalectomia , Animais , Proteínas CLOCK/biossíntese , Proteínas CLOCK/genética , Ritmo Circadiano/efeitos dos fármacos , Corticosterona/sangue , Corticosterona/farmacologia , Rim/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mifepristona/farmacologia , Antagonistas de Receptores de Mineralocorticoides/farmacologia , Fosforilação/efeitos dos fármacos , Receptores de Glucocorticoides/antagonistas & inibidores , Receptores de Glucocorticoides/efeitos dos fármacos , Espironolactona/farmacologia
3.
Pflugers Arch ; 470(2): 295-304, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28861610

RESUMO

The epithelial sodium channel (ENaC) marks the tightly regulated, rate-limiting step of sodium re-absorption in the aldosterone-sensitive distal nephron (ASDN). Stimulation of ENaC activity by aldosterone involves the serum and glucocorticoid-induced kinase 1 (SGK1) and is mediated via complex mechanisms including inhibition of channel retrieval. Retrieved channels may be recycled or degraded, e.g. by the proteasomal pathway. The aim of the present study was to investigate whether inhibitors of the proteasome affect ENaC activity and surface expression, and to explore a possible involvement of SGK1. Short circuit current (I SC) measurements were performed on confluent mCCDcl1 murine cortical collecting duct cells to investigate the effect of two distinct proteasomal inhibitors, MG132 and bortezomib, on amiloride-sensitive ENaC-mediated I SC. Both inhibitors robustly stimulated amiloride-sensitive I SC. The time course and magnitude of the stimulatory effect of the proteasomal inhibitors on I SC were similar to those of aldosterone. Both, MG132 and aldosterone, significantly increased the abundance of ß-ENaC at the cell surface. SGK1 activity was assessed by monitoring the phosphorylation of a downstream target, NDRG1, and was found to be increased by MG132. Importantly, inhibiting SGK1 activity prevented not only the stimulatory effect of aldosterone but also that of proteasomal inhibition. In conclusion, these data suggest that ENaC stimulation following proteasomal inhibition is due to an accumulation of active SGK1 resulting in increased expression of ENaC at the cell surface. Thus, inhibition of the proteasome mimics SGK1-dependent stimulation of ENaC by aldosterone.


Assuntos
Aldosterona/farmacologia , Canais Epiteliais de Sódio/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Inibidores de Proteassoma/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Amilorida/farmacologia , Animais , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Bloqueadores do Canal de Sódio Epitelial/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/efeitos dos fármacos , Túbulos Renais Coletores/metabolismo , Camundongos
4.
Am J Physiol Renal Physiol ; 308(5): F450-8, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25520009

RESUMO

There is good evidence for a causal link between excessive sympathetic drive to the kidney and hypertension. We hypothesized that sympathetic regulation of tubular Na(+) absorption may occur in the aldosterone-sensitive distal nephron, where the fine tuning of renal Na(+) excretion takes place. Here, the appropriate regulation of transepithelial Na(+) transport, mediated by the amiloride-sensitive epithelial Na(+) channel (ENaC), is critical for blood pressure control. To explore a possible effect of the sympathetic transmitter norepinephrine on ENaC-mediated Na(+) transport, we performed short-circuit current (Isc) measurements on confluent mCCDcl1 murine cortical collecting duct cells. Norepinephrine caused a complex Isc response with a sustained increase of amiloride-sensitive Isc by ∼44%. This effect was concentration dependent and mediated via basolateral α2-adrenoceptors. In cells pretreated with aldosterone, the stimulatory effect of norepinephrine was reduced. Finally, we demonstrated that noradrenergic nerve fibers are present in close proximity to ENaC-expressing cells in murine kidney slices. We conclude that the sustained stimulatory effect of locally elevated norepinephrine on ENaC-mediated Na(+) absorption may contribute to the hypertensive effect of increased renal sympathetic activity.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Túbulos Renais Coletores/metabolismo , Norepinefrina/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Sódio/metabolismo , Aldosterona , Animais , Linhagem Celular , Camundongos Endogâmicos C57BL
5.
J Gen Physiol ; 152(8)2020 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-32442241

RESUMO

Prostaglandin E2 (PGE2) is the most abundant prostanoid in the kidney, affecting a wide range of renal functions. Conflicting data have been reported regarding the effects of PGE2 on tubular water and ion transport. The amiloride-sensitive epithelial sodium channel (ENaC) is rate limiting for transepithelial sodium transport in the aldosterone-sensitive distal nephron. The aim of the present study was to explore a potential role of PGE2 in regulating ENaC in cortical collecting duct (CCD) cells. Short-circuit current (ISC) measurements were performed using the murine mCCDcl1 cell line known to express characteristic properties of CCD principal cells and to be responsive to physiological concentrations of aldosterone and vasopressin. PGE2 stimulated amiloride-sensitive ISC via basolateral prostaglandin E receptors type 4 (EP4) with an EC50 of ∼7.1 nM. The rapid stimulatory effect of PGE2 on ISC resembled that of vasopressin. A maximum response was reached within minutes, coinciding with an increased abundance of ß-ENaC at the apical plasma membrane and elevated cytosolic cAMP levels. The effects of PGE2 and vasopressin were nonadditive, indicating similar signaling cascades. Exposing mCCDcl1 cells to aldosterone caused a much slower (∼2 h) increase of the amiloride-sensitive ISC. Interestingly, the rapid effect of PGE2 was preserved even after aldosterone stimulation. Furthermore, application of arachidonic acid also increased the amiloride-sensitive ISC involving basolateral EP4 receptors. Exposure to arachidonic acid resulted in elevated PGE2 in the basolateral medium in a cyclooxygenase 1 (COX-1)-dependent manner. These data suggest that in the cortical collecting duct, locally produced and secreted PGE2 can stimulate ENaC-mediated transepithelial sodium transport.


Assuntos
Dinoprostona/farmacologia , Canais Epiteliais de Sódio , Túbulos Renais Coletores , Animais , Linhagem Celular , Agonistas do Canal de Sódio Epitelial , Canais Epiteliais de Sódio/fisiologia , Transporte de Íons , Túbulos Renais Coletores/citologia , Camundongos
6.
Br J Pharmacol ; 176(24): 4708-4719, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31423568

RESUMO

BACKGROUND AND PURPOSE: Aldosterone stimulates epithelial Na+ channel (ENaC)-dependent Na+ retention in the cortical collecting duct (CCD) of the kidney by activating mineralocorticoid receptors that promote expression of serum and glucocorticoid-inducible kinase 1 (SGK1). This response is critical to BP homeostasis. It has previously been suggested that inhibiting lysine deacetylases (KDACs) can post-transcriptionally disrupt this response by promoting acetylation of the mineralocorticoid receptor. The present study critically evaluates this hypothesis. EXPERIMENTAL APPROACH: Electrometric and molecular methods were used to define the effects of a pan-KDAC inhibitor, trichostatin A, on the responses to a physiologically relevant concentration of aldosterone (3 nM) in murine mCCDcl1 cells. KEY RESULTS: Aldosterone augmented ENaC-induced Na+ absorption and increased SGK1 activity and abundance, as expected. In the presence of trichostatin A, these responses were suppressed. Trichostatin A-induced inhibition of KDAC was confirmed by increased acetylation of histone H3, H4, and α-tubulin. Trichostatin A did not block the electrometric response to insulin, a hormone that activates SGK1 independently of increased transcription, indicating that trichostatin A has no direct effect upon the SGK1/ENaC pathway. CONCLUSIONS AND IMPLICATIONS: Inhibition of lysine de-acetylation suppresses aldosterone-dependent control over the SGK1-ENaC pathway but does not perturb post-transcriptional signalling, providing a physiological basis for the anti-hypertensive action of KDAC inhibition seen in vivo.


Assuntos
Aldosterona/farmacologia , Canais Epiteliais de Sódio/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Proteínas Imediatamente Precoces/metabolismo , Túbulos Renais Coletores/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Sódio/metabolismo , Acetilação , Animais , Células Cultivadas , Eletrofisiologia , Histona Desacetilases/metabolismo , Histonas/metabolismo , Túbulos Renais Coletores/metabolismo , Camundongos , Transdução de Sinais , Tubulina (Proteína)/metabolismo
8.
Kidney Int Rep ; 1(3): 197-203, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27722209

RESUMO

Hypertension is known as the "silent killer," driving the global public health burden of cardiovascular and renal disease. Blood pressure homeostasis is intimately associated with sodium balance and the distribution of sodium between fluid compartments and within tissues. On a population level, most societies consume 10 times more salt that the 0.5 g required by physiological need. This high salt intake is strongly linked to hypertension and to the World Health Organization targeting a ∼30% relative reduction in mean population salt intake to arrest the global mortality due to cardiovascular disease. But how does a habitually high-salt diet cause blood pressure to rise? In this focused review, we discuss 2 "evolutionary medicine" concepts, presented at the ISN Forefront Meeting "Immunomodulation of Cardio-renal Function." We first examine how ancestral variants in genes that conferred a selection advantage during early human development are now maladaptive. We then discuss the conservation of "renal" sodium transport processes across multiple organ systems, including the brain. These systems influence sodium appetite and can exert an often-overlooked effect on long-term blood pressure control.

9.
Physiol Rep ; 4(10)2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27225626

RESUMO

Serum and glucocorticoid-inducible kinase 1 (SGK1) is a protein kinase that contributes to the hormonal control of renal Na(+) retention by regulating the abundance of epithelial Na(+) channels (ENaC) at the apical surface of the principal cells of the cortical collecting duct (CCD). Although glucocorticoids and insulin stimulate Na(+) transport by activating SGK1, the responses follow different time courses suggesting that these hormones act by different mechanisms. We therefore explored the signaling pathways that allow dexamethasone and insulin to stimulate Na(+) transport in mouse CCD cells (mpkCCDcl4). Dexamethasone evoked a progressive augmentation of electrogenic Na(+) transport that became apparent after ~45 min latency and was associated with increases in SGK1 activity and abundance and with increased expression of SGK1 mRNA Although the catalytic activity of SGK1 is maintained by phosphatidylinositol-OH-3-kinase (PI3K), dexamethasone had no effect upon PI3K activity. Insulin also stimulated Na(+) transport but this response occurred with no discernible latency. Moreover, although insulin also activated SGK1, it had no effect upon SGK1 protein or mRNA abundance. Insulin did, however, evoke a clear increase in cellular PI3K activity. Our data are consistent with earlier work, which shows that glucocorticoids regulate Na(+) retention by inducing sgk1 gene expression, and also establish that this occurs independently of increased PI3K activity. Insulin, on the other hand, stimulates Na(+) transport via a mechanism independent of sgk1 gene expression that involves PI3K activation. Although both hormones act via SGK1, our data show that they activate this kinase by distinct physiological mechanisms.


Assuntos
Dexametasona/farmacologia , Proteínas Imediatamente Precoces/metabolismo , Insulina/farmacologia , Córtex Renal/enzimologia , Túbulos Renais Coletores/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Córtex Renal/efeitos dos fármacos , Túbulos Renais Coletores/efeitos dos fármacos , Camundongos
10.
Br J Pharmacol ; 161(8): 1778-92, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20735411

RESUMO

BACKGROUND AND PURPOSE: Although the serum and glucocorticoid-inducible protein kinase 1 (SGK1) appears to be involved in controlling epithelial Na(+) absorption, its role in this physiologically important ion transport process is undefined. As SGK1 activity is dependent upon target of rapamycin complex 2 (TORC2)-catalysed phosphorylation of SGK1-Ser(422) , we have explored the effects of inhibiting TORC2 and/or TORC1 upon the hormonal control of Na(+) absorption. EXPERIMENTAL APPROACH: Na(+) absorption was quantified electrometrically in mouse cortical collecting duct cells (mpkCCD) grown to confluence on permeable membranes. Kinase activities were assessed by monitoring endogenous protein phosphorylation, with or without TORC1/2 inhibitors (TORIN1 and PP242) and the TORC1 inhibitor: rapamycin. KEY RESULTS: Inhibition of TORC1/2 (TORIN1, PP242) suppressed basal SGK1 activity, prevented insulin- and dexamethasone-induced SGK1 activation, and caused modest (10-20%) inhibition of basal Na(+) absorption and substantial (∼80%) inhibition of insulin/dexamethasone-induced Na(+) transport. Inhibition of TORC1 did not impair SGK1 activation or insulin-induced Na(+) transport, but did inhibit (∼80%) dexamethasone-induced Na(+) absorption. Arginine vasopressin stimulated Na(+) absorption via a TORC1/2-independent mechanism. CONCLUSION AND IMPLICATIONS: Target of rapamycin complex 2, but not TORC1, is important to SGK1 activation. Signalling via phosphoinositide-3-kinase/TORC2/SGK1 can explain insulin-induced Na(+) absorption. TORC2, but not TORC1, is also involved in glucocorticoid-induced SGK1 activation but its role is permissive. Glucocorticoid-induced Na(+) transport displayed a requirement for TORC1 activity. Therefore, TORC1 and TORC2 contribute to the regulation of Na(+) absorption. Pharmacological manipulation of TORC1/2 signalling may provide novel therapies for Na(+)-sensitive hypertension.


Assuntos
Absorção/fisiologia , Sódio/metabolismo , Serina-Treonina Quinases TOR/fisiologia , Aldosterona/farmacologia , Animais , Arginina Vasopressina/farmacologia , Células Cultivadas , Dexametasona/farmacologia , Células Epiteliais/metabolismo , Proteínas Imediatamente Precoces/antagonistas & inibidores , Proteínas Imediatamente Precoces/metabolismo , Insulina/farmacologia , Túbulos Renais Coletores/efeitos dos fármacos , Túbulos Renais Coletores/enzimologia , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Naftiridinas/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Sirolimo/farmacologia
11.
Br J Pharmacol ; 161(3): 571-88, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20880397

RESUMO

BACKGROUND AND PURPOSE: Insulin-induced Na(+) retention in the distal nephron may contribute to the development of oedema/hypertension in patients with type 2 diabetes. This response to insulin is usually attributed to phosphatidylinositol-3-kinase (PI3K)/serum and glucocorticoid-inducible kinase 1 (SGK1) but a role for protein kinase B (PKB) has been proposed. The present study therefore aimed to clarify the way in which insulin can evoke Na(+) retention. EXPERIMENTAL APPROACH: We examined the effects of nominally selective inhibitors of PI3K (wortmannin, PI103, GDC-0941), SGK1 (GSK650394A) and PKB (Akti-1/2) on Na(+) transport in hormone-deprived and insulin-stimulated cortical collecting duct (mpkCCD) cells, while PI3K, SGK1 and PKB activities were assayed by monitoring the phosphorylation of endogenous proteins. KEY RESULTS: Wortmannin substantially inhibited basal Na(+) transport whereas PI103 and GDC-0941 had only very small effects. However, these PI3K inhibitors all abolished insulin-induced Na(+) absorption and inactivated PI3K, SGK1 and PKB fully. GSK650394A and Akti-1/2 also inhibited insulin-evoked Na(+) absorption and while GSK650394A inhibited SGK1 without affecting PKB, Akti-1/2 inactivated both kinases. CONCLUSION AND IMPLICATIONS: While studies undertaken using PI103 and GDC-0941 show that hormone-deprived cells can absorb Na(+) independently of PI3K, PI3K seems to be essential for insulin induced Na(+) transport. Akti-1/2 does not act as a selective inhibitor of PKB and data obtained using this compound must therefore be treated with caution. GSK650394A, on the other hand, selectively inhibits SGK1 and the finding that GSK650394A suppressed insulin-induced Na(+) absorption suggests that this response is dependent upon signalling via PI3K/SGK1.


Assuntos
Células Epiteliais/efeitos dos fármacos , Proteínas Imediatamente Precoces/antagonistas & inibidores , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Sódio/metabolismo , Absorção , Androstadienos/farmacologia , Animais , Benzoatos/farmacologia , Benzilaminas/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Células Cultivadas , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Furanos/farmacologia , Proteínas Imediatamente Precoces/metabolismo , Indazóis/farmacologia , Insulina/farmacologia , Transporte de Íons/efeitos dos fármacos , Túbulos Renais Coletores/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridinas/farmacologia , Pirimidinas/farmacologia , Quinoxalinas/farmacologia , Sulfonamidas/farmacologia , Wortmanina
12.
Br J Pharmacol ; 159(3): 678-88, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20105179

RESUMO

BACKGROUND AND PURPOSE: Peroxisome proliferator-activated receptor gamma (PPARgamma) agonists, such as rosiglitazone and pioglitazone, sensitize cells to insulin, and are therefore used to treat type 2 diabetes. However, in some patients, these drugs induce oedema, and the present study tests the hypothesis that this side effect reflects serum and glucocorticoid-inducible kinase 1 (SGK1)-dependent enhancement of epithelia Na(+) absorption. EXPERIMENTAL APPROACH: Na(+) absorbing epithelial cells (H441 cells, mpkCCD cells) on permeable membranes were mounted in Ussing chambers, and the effects of rosiglitazone (2 microM) and pioglitazone (10 microM) on transepithelial Na(+) absorption were quantified electrometrically. Changes in SGK1 activity were assessed by monitoring phosphorylation of residues within an endogenous protein. KEY RESULTS: Both cell types absorbed Na(+) via an electrogenic process that was enhanced by insulin. In mpkCCD cells, this stimulation of Na(+) transport was associated with increased activity of SGK1, whereas insulin regulated Na(+) transport in H441 cells through a mechanism that did not involve activation of this kinase. Rosiglitazone and pioglitazone had no discernible effect on transepithelial Na(+) absorption in unstimulated or insulin-stimulated cells and failed to alter cellular SGK1 activity. CONCLUSIONS AND IMPLICATIONS: Our results do not support the view that PPARgamma agonists stimulate epithelial Na(+) absorption or alter the control of cellular SGK1 activity. It is therefore likely that other mechanisms are involved in PPARgamma-mediated fluid retention, and a better understanding of these mechanisms may help with the identification of patients likely to develop oedema or heart failure when treated with these drugs.


Assuntos
Células Epiteliais/metabolismo , Epitélio/metabolismo , PPAR gama/agonistas , PPAR gama/metabolismo , Tiazolidinedionas/farmacologia , Transporte Biológico/efeitos dos fármacos , Contagem de Células , Glucocorticoides/metabolismo , Glucocorticoides/farmacologia , Humanos , Insulina/metabolismo , Insulina/farmacologia , Rim/metabolismo , Pulmão/metabolismo , Fosforilação , Fosfotransferases/metabolismo , Pioglitazona , Rosiglitazona , Tiazolidinedionas/agonistas , Tiazolidinedionas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA