Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Am J Physiol Renal Physiol ; 308(3): F244-51, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25391901

RESUMO

With an increase in urine flow there is a significant increase in shear stress against the renal epithelium including the inner medullary collecting duct, resulting in an increase in nitric oxide (NO) production. The mechanisms of the shear stress-mediated increases in NO are undetermined. Previous studies found that shear stress increases epithelial sodium channel (ENaC) open probability and endothelin (ET)-1 production in an ENaC-dependent mechanism in the collecting duct (CD). Given that ET-1 stimulates NO production in the CD, we hypothesized that shear stress-induced NO production is downstream of shear stress-induced ENaC activation and ET-1 production in a negative feedback loop. We determined that nitric oxide synthase 1 (NOS1) and NOS3 contribute to shear stress-mediated NO production in the CD, that is attenuated by low doses of the ENaC inhibitors amiloride and benzamil. Moreover, ETB receptor blockade significantly blunted the shear stress-mediated NO production. We further elucidated whether mice lacking NOS1 in the collecting duct (CDNOS1KO) have an impaired renal ET-1 system in the CD. Although urinary ET-1 production and inner medullary ET receptor expression were similar between flox control and CDNOS1KO mice, acute ET-1 treatment significantly reduced ENaC open probability in CDs from flox mice but not CDNOS1KO mice compared with basal. Basal ENaC activity in CDs was similar between the genotypes. We conclude that during acute shear stress across the CD, ENaC acts in a negative feedback loop to stimulate NO production in an ETB/NOS1-dependent manner resulting in a decrease in ENaC open probability and promoting natriuresis.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Túbulos Renais Coletores/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Sódio/metabolismo , Amilorida/análogos & derivados , Amilorida/farmacologia , Animais , Endotelina-1/metabolismo , Túbulos Renais Coletores/efeitos dos fármacos , Camundongos , Óxido Nítrico/metabolismo , Receptor de Endotelina B
2.
Am J Physiol Renal Physiol ; 308(3): F237-43, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25391898

RESUMO

Arginine vasopressin (AVP) activates the epithelial Na(+) channel (ENaC). The physiological significance of this activation is unknown. The present study tested if activation of ENaC contributes to AVP-sensitive urinary concentration. Consumption of a 3% NaCl solution induced hypernatremia and plasma hypertonicity in mice. Plasma AVP concentration and urine osmolality increased in hypernatremic mice in an attempt to compensate for increases in plasma tonicity. ENaC activity was elevated in mice that consumed 3% NaCl solution compared with mice that consumed a diet enriched in Na(+) with ad libitum tap water; the latter diet does not cause hypernatremia. To determine whether the increase in ENaC activity in mice that consumed 3% NaCl solution served to compensate for hypernatremia, mice were treated with the ENaC inhibitor benzamil. Coadministration of benzamil with 3% NaCl solution decreased urinary osmolality and increased urine flow so that urinary Na(+) excretion increased with no effect on urinary Na(+) concentration. This decrease in urinary concentration further increased plasma Na(+) concentration, osmolality, and AVP concentration in these already hypernatremic mice. Benzamil similarly compromised urinary concentration in water-deprived mice and in mice treated with desmopressin. These results demonstrate that stimulation of ENaC by AVP plays a critical role in water homeostasis by facilitating urinary concentration, which can compensate for hypernatremia or exacerbate hyponatremia. The present findings are consistent with ENaC in addition to serving as a final effector of the renin-angiotensin-aldosterone system and blood pressure homeostasis, also playing a key role in water homeostasis by regulating urine concentration and dilution of plasma.


Assuntos
Arginina Vasopressina/sangue , Arginina Vasopressina/urina , Canais Epiteliais de Sódio/metabolismo , Hiponatremia/urina , Rim/metabolismo , Sódio/metabolismo , Animais , Modelos Animais de Doenças , Homeostase/fisiologia , Hipernatremia/metabolismo , Camundongos Endogâmicos C57BL , Sistema Renina-Angiotensina
3.
Proc Natl Acad Sci U S A ; 109(25): 10095-100, 2012 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-22665796

RESUMO

The epithelial Na(+) channel (ENaC) in the aldosterone-sensitive distal nephron (ASDN) is under negative-feedback regulation by the renin-angiotensin-aldosterone system in protection of sodium balance and blood pressure. We test here whether aldosterone is necessary and sufficient for ENaC expression and activity in the ASDN. Surprisingly, ENaC expression and activity are robust in adrenalectomized (Adx) mice. Exogenous mineralocorticoid increases ENaC activity equally well in control and Adx mice. Plasma [AVP] is significantly elevated in Adx vs. control mice. Vasopressin (AVP) stimulates ENaC. Inhibition of the V(2) AVP receptor represses ENaC activity in Adx mice. The absence of aldosterone combined with elevated AVP release compromises normal feedback regulation of ENaC in Adx mice in response to changes in sodium intake. These results demonstrate that aldosterone is sufficient but not necessary for ENaC activity in the ASDN. Aldosterone-independent stimulation by AVP shifts the role of ENaC in the ASDN from protecting Na(+) balance to promoting water reabsorption. This stimulation of ENaC likely contributes to the hyponatremia of adrenal insufficiency.


Assuntos
Adrenalectomia , Aldosterona/fisiologia , Arginina Vasopressina/fisiologia , Canais Epiteliais de Sódio/fisiologia , Animais , Arginina Vasopressina/sangue , Camundongos
4.
Am J Physiol Renal Physiol ; 306(6): F597-607, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24431204

RESUMO

cAMP is a key mediator of connecting tubule and collecting duct (CD) Na(+) and water reabsorption. Studies performed in vitro have suggested that CD adenylyl cyclase (AC)3 partly mediates the actions of vasopressin; however, the physiological role of CD AC3 has not been determined. To assess this, mice were developed with CD-specific disruption of AC3 [CD AC3 knockout (KO)]. Inner medullary CDs from these mice exhibited 100% target gene recombination and had reduced ANG II- but not vasopressin-induced cAMP accumulation. However, there were no differences in urine volume, urinary urea excretion, or urine osmolality between KO and control mice during normal water intake or varying degrees of water restriction in the presence or absence of chronic vasopressin administration. There were no differences between CD AC3 KO and control mice in arterial pressure or urinary Na(+) or K(+) excretion during a normal or high-salt diet, whereas plasma renin and vasopressin concentrations were similar between the two genotypes. Patch-clamp analysis of split-open cortical CDs revealed no difference in epithelial Na(+) channel activity in the presence or absence of vasopressin. Compensatory changes in AC6 were not responsible for the lack of a renal phenotype in CD AC3 KO mice since combined CD AC3/AC6 KO mice had similar arterial pressure and renal Na(+) and water handling compared with CD AC6 KO mice. In summary, these data do not support a significant role for CD AC3 in the regulation of renal Na(+) and water excretion in general or vasopressin regulation of CD function in particular.


Assuntos
Adenilil Ciclases/deficiência , Túbulos Renais Coletores/fisiologia , Sódio/urina , Adenilil Ciclases/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Diurese , Feminino , Masculino , Camundongos , Camundongos Knockout , Cloreto de Sódio na Dieta/farmacologia
5.
J Am Soc Nephrol ; 24(2): 218-27, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23264685

RESUMO

Vasopressin modulates sodium reabsorption in the collecting duct through adenylyl cyclase-stimulated cyclic AMP, which exists as multiple isoforms; the specific isoform involved in vasopressin-stimulated sodium transport is unknown. To assess this, we studied mice deficient in adenylyl cyclase type VI specifically in the principal cells of the collecting duct. Knockout mice had increased urine volume and reduced urine sodium concentration, but regardless of the level of sodium intake, they did not exhibit significant alterations in urinary sodium excretion, arterial pressure, or pulse rate. Plasma renin concentration was elevated in knockout mice, however, suggesting a compensatory response. Valsartan significantly reduced arterial pressure in knockout mice but not in controls. Knockout mice had decreased renal cortical mRNA content of all three epithelial sodium channel (ENaC) isoforms, and total cell sodium channel isoforms α and γ were reduced in these animals. Patch-clamp analysis of split-open cortical collecting ducts revealed no difference in baseline activity of sodium channels, but knockout mice had abolished vasopressin-stimulated ENaC open probability and apical membrane channel number. In summary, these data suggest that adenylyl cyclase VI mediates vasopressin-stimulated ENaC activity in the kidney.


Assuntos
Adenilil Ciclases/metabolismo , Arginina Vasopressina/farmacologia , Canais Epiteliais de Sódio/metabolismo , Túbulos Renais Coletores/enzimologia , Adenilil Ciclases/genética , Aldosterona/urina , Animais , Antidiuréticos/metabolismo , Antidiuréticos/farmacologia , Anti-Hipertensivos/farmacologia , Aquaporina 2/genética , Arginina Vasopressina/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Canais Epiteliais de Sódio/genética , Feminino , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Túbulos Renais Coletores/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Renina/sangue , Sódio/urina , Cloreto de Sódio na Dieta/farmacologia , Tetrazóis/farmacologia , Valina/análogos & derivados , Valina/farmacologia , Valsartana
6.
J Biol Chem ; 287(46): 38552-8, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-23002235

RESUMO

The epithelial Na(+) channel (ENaC) in the aldosterone-sensitive distal nephron (ASDN) is under tonic inhibition by a local purinergic signaling system responding to changes in dietary sodium intake. Normal BK(Ca) channel function is required for flow-sensitive ATP secretion in the ASDN. We tested here whether ATP secreted through connexin channels in a coupled manner with K(+) efflux through BK(Ca) channels is required for inhibitory purinergic regulation of ENaC in response to increases in sodium intake. Inhibition of connexin channels relieves purinergic inhibition of ENaC. Deletion of the BK-ß4 regulatory subunit, which is required for normal BK(Ca) channel function and flow-sensitive ATP secretion in the ASDN, suppresses increases in urinary ATP in response to increases in sodium intake. As a consequence, ENaC activity, particularly in the presence of high sodium intake, is inappropriately elevated in BK-ß4 null mice. ENaC in BK-ß4 null mice, however, responds normally to exogenous ATP, indicating that increases in activity do not result from end-organ resistance but rather from lowered urinary ATP. Consistent with this, disruption of purinergic regulation increases ENaC activity in wild type but not BK-ß4 null mice. Consequently, sodium excretion is impaired in BK-ß4 null mice. These results demonstrate that the ATP secreted in the ASDN in a BK(Ca) channel-dependent manner is physiologically available for purinergic inhibition of ENaC in response to changes in sodium homeostasis. Impaired sodium excretion resulting form loss of normal purinergic regulation of ENaC in BK-ß4 null mice likely contributes to their elevated blood pressure.


Assuntos
Trifosfato de Adenosina/química , Canais Epiteliais de Sódio/metabolismo , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Néfrons/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Pressão Sanguínea , Conexinas/química , Homeostase , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Técnicas de Patch-Clamp , Sódio/química , Sódio/metabolismo , Sódio na Dieta/metabolismo
7.
Am J Physiol Cell Physiol ; 302(1): C188-94, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21918182

RESUMO

Collecting duct (CD)-derived endothelin-1 (ET-1) acting via endothelin B (ETB) receptors promotes Na(+) excretion. Compromise of ET-1 signaling or ETB receptors in the CD cause sodium retention and increase blood pressure. Activity of the epithelial Na(+) channel (ENaC) is limiting for Na(+) reabsorption in the CD. To test for ETB receptor regulation of ENaC, we combined patch-clamp electrophysiology with CD-specific knockout (KO) of endothelin receptors. We also tested how ET-1 signaling via specific endothelin receptors influences ENaC activity under differing dietary Na(+) regimens. ET-1 significantly decreased ENaC open probability in CD isolated from wild-type (WT) and CD ETA KO mice but not CD ETB KO and CD ETA/B KO mice. ENaC activity in WT and CD ETA but not CD ETB and CD ETA/B KO mice was inversely related to dietary Na(+) intake. ENaC activity in CD ETB and CD ETA/B KO mice tended to be elevated under all dietary Na(+) regimens compared with WT and CD ETA KO mice, reaching significance with high (2%) Na(+) feeding. These results show that the bulk of ET-1 inhibition of ENaC activity is mediated by the ETB receptor. In addition, they could explain the Na(+) retention and elevated blood pressure observed in CD ET-1 KO, CD ETB KO, and CD ETA/B KO mice consistent with ENaC regulation by ET-1 via ETB receptors contributing to the antihypertensive and natriuretic effects of the local endothelin system in the mammalian CD.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Túbulos Renais Coletores/metabolismo , Receptor de Endotelina B/deficiência , Receptor de Endotelina B/genética , Regulação para Cima/genética , Amilorida/farmacologia , Animais , Endotelina-1/metabolismo , Endotelina-1/fisiologia , Bloqueadores do Canal de Sódio Epitelial , Feminino , Hipertensão/genética , Hipertensão/metabolismo , Masculino , Camundongos , Camundongos Knockout , Natriurese/genética , Sódio/metabolismo
8.
J Biol Chem ; 286(2): 1054-60, 2011 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-21075848

RESUMO

We tested whether ATP release through Connexin 30 (Cx30) is part of a local purinergic regulatory system intrinsic to the aldosterone-sensitive distal nephron (ASDN) important for proper control of sodium excretion; if changes in sodium intake influence ATP release via Cx30; and if this allows a normal ENaC response to changes in systemic sodium levels. In addition, we define the consequences of disrupting ATP regulation of ENaC in Cx30(-/-) mice. Urinary ATP levels in wild-type mice increase with sodium intake, being lower and less dependent on sodium intake in Cx30(-/-) mice. Loss of inhibitory ATP regulation causes ENaC activity to be greater in Cx30(-/-) versus wild-type mice, particularly with high sodium intake. This results from compromised ATP release rather than end-organ resistance: ENaC in Cx30(-/-) mice responds to exogenous ATP. Thus, loss of paracrine ATP feedback regulation of ENaC in Cx30(-/-) mice disrupts normal responses to changes in sodium intake. Consequently, ENaC is hyperactive in Cx30(-/-) mice lowering sodium excretion particularly during increases in sodium intake. Clamping mineralocorticoids high in Cx30(-/-) mice fed a high sodium diet causes a marked decline in renal sodium excretion. This is not the case in wild-type mice, which are capable of undergoing aldosterone-escape. This loss of the ability of ENaC to respond to changes in sodium levels contributes to salt-sensitive hypertension in Cx30(-/-) mice.


Assuntos
Conexinas/genética , Conexinas/metabolismo , Canais Epiteliais de Sódio/metabolismo , Hipertensão Renal/metabolismo , Comunicação Parácrina/fisiologia , Trifosfato de Adenosina/urina , Aldosterona/metabolismo , Animais , Conexina 30 , Retroalimentação Fisiológica/fisiologia , Hipertensão Renal/genética , Túbulos Renais Coletores/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Receptores Purinérgicos P2Y2/metabolismo , Transdução de Sinais/fisiologia , Cloreto de Sódio na Dieta/farmacologia , Cloreto de Sódio na Dieta/urina
9.
J Biol Chem ; 286(3): 1683-95, 2011 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-21041305

RESUMO

The epithelial Na(+) channel (ENaC) is modulated by membrane lipid composition. However, the effect of an in vivo change of membrane composition is unknown. We examined the effect of a 70-day enhanced cholesterol diet (ECD) on ENaC and renal Na(+) handling. Rats were fed a standard chow or one supplemented with 1% cholesterol and 0.5% cholic acid (ECD). ECD animals exhibited marked anti-diuresis and anti-natriuresis (40 and 47%), which peaked at 1-3 weeks. Secondary compensation returned urine output and urinary Na(+) excretion to control levels by week 10. During these initial changes, there were no accompanying effects on systolic blood pressure, serum creatinine, or urinary creatinine excretion, indicating that the these effects of ECD preceded those which modify renal filtration and blood pressure. The effects of ECD on ENaC were evaluated by measuring the relative protein content of α, ß, and γ subunits. α and γ blots were further examined for subunit cleavage (a process that activates ENaC). No significant changes were observed in α and ß levels throughout the study. However, levels of cleaved γ were elevated, suggesting that ENaC was activated. The changes of γ persisted at week 10 and were accompanied by additional subunit fragments, indicating potential changes of γ-cleaving proteases. Enhanced protease activity, and specifically that which could act on the second identified cleavage site in γ, was verified in a newly developed urinary protease assay. These results predict enhanced ENaC activity, an effect that was confirmed in patch clamp experiments of principal cells of split open collecting ducts, where ENaC open probability was increased by 40% in the ECD group. These data demonstrate a complex series of events and a new regulatory paradigm that is initiated by ECD prior to the onset of elevated blood pressure. These events lead to changes of renal Na(+) handling, which occur in part by effects on extracellular γ-ENaC cleavage.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Colesterol na Dieta/efeitos adversos , Dieta , Canais Epiteliais de Sódio/metabolismo , Taxa de Filtração Glomerular/efeitos dos fármacos , Túbulos Renais Coletores/metabolismo , Sódio/urina , Animais , Linhagem Celular , Colesterol na Dieta/farmacologia , Ácido Cólico/efeitos adversos , Ácido Cólico/farmacologia , Creatinina/sangue , Creatinina/urina , Cães , Canais Epiteliais de Sódio/genética , Humanos , Masculino , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Ratos
10.
FASEB J ; 24(6): 2056-65, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20097874

RESUMO

Apical release of ATP and UTP can activate P2Y(2) receptors in the aldosterone-sensitive distal nephron (ASDN) and inhibit the open probability (P(o)) of the epithelial sodium channel (ENaC). Little is known, however, about the regulation and physiological relevance of this system. Patch-clamp studies in freshly isolated ASDN provide evidence that increased dietary Na(+) intake in wild-type mice lowers ENaC P(o), consistent with a contribution to Na(+) homeostasis, and is associated with increased urinary concentrations of UTP and the ATP hydrolytic product, ADP. Genetic deletion of P2Y(2) receptors in mice (P2Y(2)(-/-); littermates to wild-type mice) or inhibition of apical P2Y-receptor activation in wild-type mice prevents dietary Na(+)-induced lowering of ENaC P(o). Although they lack suppression of ENaC P(o) by dietary NaCl, P2Y(2)(-/-) mice do not exhibit NaCl-sensitive blood pressure, perhaps as a consequence of compensatory down-regulation of aldosterone levels. Consistent with this hypothesis, clamping mineralocorticoid activity at high levels unmasks greater ENaC activity and NaCl sensitivity of blood pressure in P2Y(2)(-/-) mice. The studies indicate a key role of the apical ATP/UTP-P2Y(2)-receptor system in the inhibition of ENaC P(o) in the ASDN in response to an increase in Na(+) intake, thereby contributing to NaCl homeostasis and blood pressure regulation.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Rim/metabolismo , Receptores Purinérgicos P2/fisiologia , Sódio na Dieta/administração & dosagem , Sódio na Dieta/farmacologia , Trifosfato de Adenosina/urina , Aldosterona/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Células Cultivadas , Eletrofisiologia , Homeostase/efeitos dos fármacos , Rim/citologia , Rim/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mineralocorticoides/farmacologia , Néfrons/citologia , Néfrons/efeitos dos fármacos , Néfrons/metabolismo , Técnicas de Patch-Clamp , Receptores Purinérgicos P2Y2 , Transdução de Sinais , Uridina Trifosfato/urina
11.
J Am Soc Nephrol ; 21(11): 1903-11, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20813869

RESUMO

The mechanisms underlying "aldosterone escape," which refers to the excretion of sodium (Na(+)) during high Na(+) intake despite inappropriately increased levels of mineralocorticoids, are incompletely understood. Because local purinergic tone in the aldosterone-sensitive distal nephron downregulates epithelial Na(+) channel (ENaC) activity, we tested whether this mechanism mediates aldosterone escape. Here, urinary ATP concentration increased with dietary Na(+) intake in mice. Physiologic concentrations of ATP decreased ENaC activity in a dosage-dependent manner. P2Y(2)(-/-) mice, which lack the purinergic receptor, had significantly less increased Na(+) excretion than wild-type mice in response to high-Na(+) intake. Exogenous deoxycorticosterone acetate and deletion of the P2Y(2) receptor each modestly increased the resistance of ENaC to changes in Na(+) intake; together, they markedly increased resistance. Under the latter condition, ENaC could not respond to changes in Na(+) intake. In contrast, as a result of aldosterone escape, wild-type mice had increased Na(+) excretion in response to high-Na(+) intake regardless of the presence of high deoxycorticosterone acetate. These data suggest that control of ENaC by purinergic signaling is necessary for aldosterone escape.


Assuntos
Aldosterona/metabolismo , Regulação para Baixo/fisiologia , Canais Epiteliais de Sódio/metabolismo , Néfrons/metabolismo , Receptores Purinérgicos P2Y2/deficiência , Trifosfato de Adenosina/urina , Animais , Desoxicorticosterona/análogos & derivados , Desoxicorticosterona/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Néfrons/efeitos dos fármacos , Receptores Purinérgicos P2Y2/genética , Receptores Purinérgicos P2Y2/metabolismo , Sódio/urina , Sódio na Dieta/farmacologia
12.
J Am Soc Nephrol ; 20(4): 721-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19158355

RESUMO

Thiazolidinediones are agonists of peroxisome proliferator-activated receptor gamma (PPARgamma) that can induce fluid retention and weight gain through unclear mechanisms. To test a proposed role for the epithelial sodium channel ENaC in thiazolidinedione-induced fluid retention, we used mice with conditionally inactivated alphaENaC in the collecting duct (Scnn1a(loxloxCre) mice). In control mice, rosiglitazone did not alter plasma aldosterone levels or protein expression of ENaC subunits in the kidney, but did increase body weight, plasma volume, and the fluid content of abdominal fat pads, and decreased hematocrit. Scnn1a(loxloxCre) mice provided functional evidence for blunted Na+ uptake in the collecting duct, but still exhibited rosiglitazone-induced fluid retention. Moreover, treatment with rosiglitazone or pioglitazone did not significantly alter the open probability or number of ENaC channels per patch in isolated, split-open cortical collecting ducts of wild-type mice. Finally, patch-clamp studies in primary mouse inner medullary collecting duct cells did not detect ENaC activity but did detect a nonselective cation channel upregulated by pioglitazone. These data argue against a primary and critical role of ENaC in thiazolidinedione-induced fluid retention.


Assuntos
Canais Epiteliais de Sódio/fisiologia , Medula Renal/fisiologia , Túbulos Renais Coletores/fisiologia , Tiazolidinedionas/farmacologia , Animais , Canais Epiteliais de Sódio/efeitos dos fármacos , Medula Renal/efeitos dos fármacos , Túbulos Renais Coletores/efeitos dos fármacos , Camundongos , Técnicas de Patch-Clamp , Pioglitazona , Rosiglitazona , Aumento de Peso/efeitos dos fármacos
13.
Am J Physiol Renal Physiol ; 297(5): F1411-8, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19692483

RESUMO

We used patch-clamp electrophysiology on isolated, split-open murine collecting ducts (CD) to test the hypothesis that regulation of epithelial sodium channel (ENaC) activity is a physiologically important effect of vasopressin. Surprisingly, this has not been tested directly before. We ask whether vasopressin affects ENaC activity distinguishing between acute and chronic effects, as well as, parsing the cellular signaling pathway and molecular mechanism of regulation. In addition, we quantified possible synergistic regulation of ENaC by vasopressin and aldosterone associating this with a requirement for distal nephron Na+ reabsorption during water conservation vs. maintenance of Na+ balance. We find that vasopressin significantly increases ENaC activity within 2-3 min by increasing open probability (P(o)). This activation was dependent on adenylyl cyclase (AC) and PKA. Water restriction (18-24 h) and pretreatment of isolated CD with vasopressin (approximately 30 min) resulted in a similar increase in P(o). In addition, this also increased the number (N) of active ENaC in the apical membrane. Similar to P(o), increases in N were sensitive to inhibitors of AC. Stressing animals with water and salt restriction separately and jointly revealed an important effect of vasopressin: conservation of water and Na+ each independently increased ENaC activity and jointly had a synergistic effect on channel activity. These results demonstrate a quantitatively important action of vasopressin on ENaC suggesting that distal nephron Na+ reabsorption mediated by this channel contributes to maintenance of water reabsorption. In addition, our results support that the combined actions of vasopressin and aldosterone are required to achieve maximally activated ENaC.


Assuntos
Agonistas do Canal de Sódio Epitelial , Canais Epiteliais de Sódio/metabolismo , Túbulos Renais Coletores/metabolismo , Fármacos Renais/farmacologia , Vasopressinas/farmacologia , Água/metabolismo , Absorção , Inibidores de Adenilil Ciclases , Adenilil Ciclases/metabolismo , Aldosterona/farmacologia , Animais , Arginina Vasopressina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Eletrofisiologia , Canais Epiteliais de Sódio/efeitos dos fármacos , Feminino , Túbulos Renais Coletores/efeitos dos fármacos , Masculino , Camundongos , Técnicas de Patch-Clamp , Sódio/deficiência , Sódio/metabolismo
14.
J Gen Physiol ; 130(4): 399-413, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17893193

RESUMO

Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P(2)) and phosphatidylinositol 3,4,5-trisphosphate (PI(3,4,5)P(3)) are physiologically important second messengers. These molecules bind effector proteins to modulate activity. Several types of ion channels, including the epithelial Na(+) channel (ENaC), are phosphoinositide effectors capable of directly interacting with these signaling molecules. Little, however, is known of the regions within ENaC and other ion channels important to phosphoinositide binding and modulation. Moreover, the molecular mechanism of this regulation, in many instances, remains obscure. Here, we investigate modulation of ENaC by PI(3,4,5)P(3) and PI(4,5)P(2) to begin identifying the molecular determinants of this regulation. We identify intracellular regions near the inner membrane interface just following the second transmembrane domains in beta- and gamma- but not alpha-ENaC as necessary for PI(3,4,5)P(2) but not PI(4,5)P(2) modulation. Charge neutralization of conserved basic amino acids within these regions demonstrated that these polar residues are critical to phosphoinositide regulation. Single channel analysis, moreover, reveals that the regions just following the second transmembrane domains in beta- and gamma-ENaC are critical to PI(3,4,5)P(3) augmentation of ENaC open probability, thus, defining mechanism. Unexpectedly, intracellular domains within the extreme N terminus of beta- and gamma-ENaC were identified as being critical to down-regulation of ENaC activity and P(o) in response to depletion of membrane PI(4,5)P(2). These regions of the channel played no identifiable role in a PI(3,4,5)P(3) response. Again, conserved positive-charged residues within these domains were particularly important, being necessary for exogenous PI(4,5)P(2) to increase open probability. We conclude that beta and gamma subunits bestow phosphoinositide sensitivity to ENaC with distinct regions of the channel being critical to regulation by PI(3,4,5)P(3) and PI(4,5)P(2). This argues that these phosphoinositides occupy distinct ligand-binding sites within ENaC to modulate open probability.


Assuntos
Canais Epiteliais de Sódio , Ativação do Canal Iônico , Fosfatidilinositol 4,5-Difosfato/farmacologia , Fosfatos de Fosfatidilinositol/farmacologia , Subunidades Proteicas/metabolismo , Motivos de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação , Células CHO , Linhagem Celular Transformada , Cricetinae , Cricetulus , Agonistas do Canal de Sódio Epitelial , Canais Epiteliais de Sódio/química , Canais Epiteliais de Sódio/metabolismo , Camundongos , Microscopia de Fluorescência , Técnicas de Patch-Clamp , Marcadores de Fotoafinidade , Subunidades Proteicas/química
15.
Curr Opin Nephrol Hypertens ; 17(5): 533-40, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18695396

RESUMO

PURPOSE OF REVIEW: Epithelial sodium channel (ENaC) activity is limiting for sodium reabsorption in the distal nephron. Humans regulate blood pressure by fine-tuning sodium balance through control of ENaC. ENaC dysfunction causes some hypertensive and renal salt wasting diseases. Thus, it is critical to understand the cellular mechanisms controlling ENaC activity. RECENT FINDINGS: ENaC is sensitive to phosphatidylinositol 4,5-bisphosphate (PIP2), the target of phospholipase C-mediated metabolism, and phosphatidylinositiol 3,4,5-trisphosphate (PIP3), the product of phosphatidylinositide 3-OH kinase (PI3-K). PIP2 is permissive for ENaC gating possibly interacting directly with the channel. Activation of distal nephron P2Y receptors tempers ENaC activity by promoting PIP2 metabolism. This is important because gene deletion of P2Y2 receptors causes hypertension associated with hyperactive ENaC. Aldosterone, the final hormone in a negative-feedback cascade activated by decreases in blood pressure, increases ENaC activity. PIP3 sits at a critical bifurcation in the aldosterone-signaling cascade, increasing ENaC open probability and number. PIP3-effectors mediate increases in ENaC number by suppressing channel retrieval. PIP3 binds ENaC, at a site distinct from that important to PIP2 regulation, to modulate directly open probability. SUMMARY: Phosphoinositides play key roles in physiologic control of ENaC and perhaps dysregulation plays a role in disease associated with abnormal renal sodium handling.


Assuntos
Canais Epiteliais de Sódio/fisiologia , Fosfatidilinositóis/fisiologia , Animais , Humanos , Transdução de Sinais
16.
Am J Physiol Renal Physiol ; 294(1): F38-46, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17913833

RESUMO

Activity of the epithelial sodium channel (ENaC) is limiting for Na(+) reabsorption at the distal nephron. Phosphoinositides, such as phosphatidylinositol 4,5-biphosphate [PI(4,5)P(2)] modulate the activity of this channel. Activation of purinergic receptors triggers multiple events, including activation of PKC and PLC, with the latter depleting plasma membrane PI(4,5)P(2). Here, we investigate regulation of ENaC in renal principal cells by purinergic receptors via PLC and PI(4,5)P(2). Purinergic signaling rapidly decreases ENaC open probability and apical membrane PI(4,5)P(2) levels with similar time courses. Moreover, inhibiting purinergic signaling with suramin rescues ENaC activity. The PLC inhibitor U73122, but not U73343, its inactive analog, recapitulates the action of suramin. In contrast, modulating PKC signaling failed to affect purinergic regulation of ENaC. Unexpectedly, inhibiting either purinergic receptors or PLC in resting cells dramatically increased ENaC activity above basal levels, indicating tonic activation of purinergic signaling in these polarized renal epithelial cells. Increased ENaC activity was associated with elevation of apical membrane PI(4,5)P(2) levels. Subsequent treatment with ATP in the presence of inhibited purinergic signaling failed to decrease ENaC activity and apical membrane PI(4,5)P(2) levels. Dwell-time analysis reveals that depletion of PI(4,5)P(2) forces ENaC toward a closed state. In contrast, increasing PI(4,5)P(2) levels above basal values locks the channel in an open state interrupted by brief closings. Thus our results suggest that purinergic control of apical membrane PI(4,5)P(2) levels is a major regulator of ENaC activity in renal epithelial cells.


Assuntos
Membrana Celular/metabolismo , Canais Epiteliais de Sódio/metabolismo , Túbulos Renais Coletores/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Receptores Purinérgicos/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/efeitos dos fármacos , Túbulos Renais Distais/citologia , Túbulos Renais Distais/efeitos dos fármacos , Túbulos Renais Distais/metabolismo , Camundongos , Técnicas de Patch-Clamp , Proteína Quinase C/metabolismo , Transdução de Sinais , Fosfolipases Tipo C/metabolismo
17.
J Biol Chem ; 283(52): 36599-607, 2008 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-18981175

RESUMO

Growing evidence implicates a key role for extracellular nucleotides in cellular regulation, including of ion channels and renal function, but the mechanisms for such actions are inadequately defined. We investigated purinergic regulation of the epithelial Na+ channel (ENaC) in mammalian collecting duct. We find that ATP decreases ENaC activity in both mouse and rat collecting duct principal cells. ATP and other nucleotides, including UTP, decrease ENaC activity via apical P2Y2 receptors. ENaC in collecting ducts isolated from mice lacking this receptor have blunted responses to ATP. P2Y2 couples to ENaC via PLC; direct activation of PLC mimics ATP action. Tonic regulation of ENaC in the collecting duct occurs via locally released ATP; scavenging endogenous ATP and inhibiting P2 receptors, in the absence of other stimuli, rapidly increases ENaC activity. Moreover, ENaC has greater resting activity in collecting ducts from P2Y2-/- mice. Loss of collecting duct P2Y2 receptors in the knock-out mouse is the primary defect leading to increased ENaC activity based on the ability of direct PLC stimulation to decrease ENaC activity in collecting ducts from P2Y2-/- mice in a manner similar to ATP in collecting ducts from wild-type mice. These findings demonstrate that locally released ATP acts in an autocrine/paracrine manner to tonically regulate ENaC in mammalian collecting duct. Loss of this intrinsic regulation leads to ENaC hyperactivity and contributes to hypertension that occurs in P2Y2 receptor-/- mice. P2Y2 receptor activation by nucleotides thus provides physiologically important regulation of ENaC and electrolyte handling in mammalian kidney.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Regulação da Expressão Gênica , Receptores Purinérgicos P2/metabolismo , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Animais , Eletrofisiologia , Epitélio/metabolismo , Canais Iônicos/metabolismo , Túbulos Renais Coletores/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Nucleotídeos/química , Ratos , Receptores Purinérgicos P2Y2
18.
Am J Physiol Renal Physiol ; 295(4): F1063-70, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18667482

RESUMO

We used patch-clamp electrophysiology to investigate regulation of the epithelial Na+ channel (ENaC) by endothelin-1 (ET-1) in isolated, split-open rat collecting ducts. ET-1 significantly decreases ENaC open probability by about threefold within 5 min. ET-1 decreases ENaC activity through basolateral membrane ETB but not ETA receptors. In rat collecting duct, we find no role for phospholipase C or protein kinase C in the rapid response of ENaC to ET-1. ET-1, although, does activate src family tyrosine kinases and their downstream MAPK1/2 effector cascade in renal principal cells. Both src kinases and MAPK1/2 signaling are necessary for ET-1-dependent decreases in ENaC open probability in the split-open collecting duct. We conclude that ET-1 in a physiologically relevant manner rapidly suppresses ENaC activity in native, mammalian principal cells. These findings may provide a potential mechanism for the natriuresis observed in vivo in response to ET-1, as well as a potential cause for the salt-sensitive hypertension found in animals with impaired endothelin signaling.


Assuntos
Endotelina-1/metabolismo , Canais Epiteliais de Sódio/fisiologia , Ativação do Canal Iônico/fisiologia , Túbulos Renais Coletores/metabolismo , Animais , Linhagem Celular Transformada , Endotelina-1/farmacologia , Feminino , Hipertensão Renal/metabolismo , Hipertensão Renal/fisiopatologia , Ativação do Canal Iônico/efeitos dos fármacos , Túbulos Renais Coletores/citologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Quinases da Família src/metabolismo
19.
J Am Soc Nephrol ; 18(6): 1652-61, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17442787

RESUMO

Activity of the epithelial Na(+) channel (ENaC) is limiting for Na(+) reabsorption in the aldosterone-sensitive distal nephron. Hormones, including aldosterone and insulin, increase ENaC activity, in part by stimulating phosphatidylinositide 3-OH kinase (PI3-K) signaling. Recent studies in heterologous expression systems reveal a close spatiotemporal coupling between PI3-K signaling and ENaC activity with the phospholipid product of this kinase, PI(3,4,5)P(3), in some cases, directly binding the channel and increasing open probability (P(o)). This study tested whether this tight coupling plays a physiologic role in modulating ENaC activity in native tissue and polarized epithelial cells. IGF-I was found to increase Na(+) reabsorption across mpkCCD(c14) principal cell monolayers in a PI3-K-sensitive manner. Inhibition of PI3-K signaling, moreover, rapidly decreased Na(+) reabsorption and ENaC activity in mpkCCD(c14) cells that were treated with corticosteroids and IGF-I. These decreases paralleled changes in apical membrane PI(3,4,5)P(3) levels, demonstrating tight spatiotemporal coupling between ENaC activity and PI3-K/PI(3,4,5)P(3) signaling within this membrane. For further probing of the mechanism underpinning this coupling, cortical collecting ducts (CCD) were isolated from rat and split open to expose the apical membrane for patch-clamp analysis. Inhibition of PI3-K signaling with wortmannin and LY294002 but not its inactive analogue rapidly and markedly decreased the P(o) of ENaC. Moreover, IGF-I acutely increased P(o) of ENaC in CCD principal cells in a PI3-K-sensitive manner. Together, these observations stress the importance of tight spatiotemporal coupling between PI3-K signaling and ENaC within the apical membrane of principal cells to the physiologic control of this ion channel.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Túbulos Renais Coletores/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/fisiologia , Junções Íntimas/fisiologia , Androstadienos/farmacologia , Animais , Linhagem Celular Transformada , Polaridade Celular/fisiologia , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Canais Epiteliais de Sódio/genética , Feminino , Fator de Crescimento Insulin-Like I/metabolismo , Túbulos Renais Coletores/citologia , Masculino , Camundongos , Morfolinas/farmacologia , Técnicas de Patch-Clamp , Fosfatidilinositóis/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Sódio/metabolismo , Wortmanina
20.
J Biol Chem ; 282(19): 14576-85, 2007 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-17376773

RESUMO

The epithelial Na(+) channel (ENaC) plays a central role in control of epithelial surface hydration and vascular volume. Similar to other ion channels, ENaC activity is set, in part, by its membrane levels. The small G protein RhoA increases ENaC activity by increasing the membrane levels of this channel. We hypothesize that RhoA increases ENaC activity by promoting channel trafficking to the plasma membrane. Few experimental methods are available to directly visualize trafficking of ion channels to the plasma membrane. Here we combine electrophysiology with two complementary imaging methods, total internal reflection fluorescence microscopy and fluorescence recovery after photobleaching, to study the mechanistic basis of RhoA actions on ENaC. Patch clamp results demonstrate that RhoA increases ENaC activity in an additive manner with dominant-negative dynamin. This is consistent with a mechanism of increased ENaC trafficking to the membrane. Direct visualization of ENaC movement near the plasma membrane with total internal reflection fluorescence-fluorescence recovery after photobleaching revealed that RhoA accelerates ENaC trafficking toward the membrane. RhoA-facilitated movement of the channel was sensitive to disrupting the endomembrane system. Moreover, facilitating retrieval decreased ENaC activity but not trafficking toward the membrane. ENaC at the plasma membrane clustered and was laterally immobile suggesting that the cytoskeleton tethers or corrals membrane resident channels or membrane-directed vesicles containing ENaC. Disrupting microtubules but not microfilaments led to reorganization of ENaC clusters and slowed trafficking toward the membrane. The cytoskeleton is an established target for RhoA signaling. We conclude that RhoA, likely through effects on the cytoskeleton, promotes ENaC trafficking to the plasma membrane to increase channel membrane levels and activity.


Assuntos
Membrana Celular/metabolismo , Canais Epiteliais de Sódio/metabolismo , Recuperação de Fluorescência Após Fotodegradação , Microscopia de Fluorescência , Proteína rhoA de Ligação ao GTP/farmacologia , Animais , Células CHO , Células COS , Chlorocebus aethiops , Cricetinae , Cricetulus , Citoesqueleto/metabolismo , Dinaminas/metabolismo , Eletrofisiologia , Proteínas Luminescentes/metabolismo , Camundongos , Microtúbulos/metabolismo , Transporte Proteico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA