Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Physiol Rev ; 95(1): 297-340, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25540145

RESUMEN

Transcription and translation require a high concentration of potassium across the entire tree of life. The conservation of a high intracellular potassium was an absolute requirement for the evolution of life on Earth. This was achieved by the interplay of P- and V-ATPases that can set up electrochemical gradients across the cell membrane, an energetically costly process requiring the synthesis of ATP by F-ATPases. In animals, the control of an extracellular compartment was achieved by the emergence of multicellular organisms able to produce tight epithelial barriers creating a stable extracellular milieu. Finally, the adaptation to a terrestrian environment was achieved by the evolution of distinct regulatory pathways allowing salt and water conservation. In this review we emphasize the critical and dual role of Na(+)-K(+)-ATPase in the control of the ionic composition of the extracellular fluid and the renin-angiotensin-aldosterone system (RAAS) in salt and water conservation in vertebrates. The action of aldosterone on transepithelial sodium transport by activation of the epithelial sodium channel (ENaC) at the apical membrane and that of Na(+)-K(+)-ATPase at the basolateral membrane may have evolved in lungfish before the emergence of tetrapods. Finally, we discuss the implication of RAAS in the origin of the present pandemia of hypertension and its associated cardiovascular diseases.


Asunto(s)
Aldosterona/metabolismo , Evolución Biológica , Canales Epiteliales de Sodio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Sodio/metabolismo , Animales , Canales Epiteliales de Sodio/química , Canales Epiteliales de Sodio/genética , Genoma Humano , Humanos , Nefronas/fisiología , Transducción de Señal/fisiología , ATPasa Intercambiadora de Sodio-Potasio/química , ATPasa Intercambiadora de Sodio-Potasio/genética
2.
Rev Med Suisse ; 16(701): 1450-1455, 2020 Aug 05.
Artículo en Francés | MEDLINE | ID: mdl-32833367

RESUMEN

The Covid 19 pandemic remains a serious public health problem until effective drugs and/or vaccines are available. Can we explain why so many people remain asymptomatic but nevertheless highly contagious explaining the speed with which the pandemic has spread around the world? Can we explain why the acute respiratory distress syndrome (ARDS) appears late but can so quickly have a fatal outcome? In the lung, mucociliary clearance (CMC) and alveolar clearance (CA) depend on the transport of sodium through the plasma membrane of epithelial cells. This transport is mediated by a highly selective sodium channel (Epithelial Sodium Channel = ENaC) which could be a key element in the pulmonary pathophysiology of SARS-CoV-2 infection.


La pandémie Covid-19 reste un grave problème de santé publique tant que l'on ne disposera pas de médicaments et/ou d'un vaccin efficaces. Peut-on expliquer pourquoi de si nombreuses personnes restent asymptomatiques mais néanmoins hautement contagieuses expliquant la rapidité avec laquelle la pandémie s'est répandue dans le monde ? Pourquoi le syndrome de détresse respiratoire aigu (SDRA) apparaît-il tardivement mais peut rapidement avoir une issue fatale ? Dans le poumon, la clairance muco-ciliaire (CMC) et la clairance alvéolaire (CA) dépendent du transport de sodium à travers la membrane plasmique des cellules épithéliales. Ce transport est médié par un canal ionique hautement sélectif pour le sodium (Epithelial Sodium Channel = ENaC), qui pourrait être un élément clé de la physiopathologie pulmonaire de l'infection à SARS-CoV-2.


Asunto(s)
Betacoronavirus/patogenicidad , Infecciones por Coronavirus/metabolismo , Modelos Biológicos , Neumonía Viral/metabolismo , Sodio/metabolismo , COVID-19 , Infecciones por Coronavirus/virología , Humanos , Transporte Iónico , Pandemias , Neumonía Viral/virología , SARS-CoV-2
3.
J Am Soc Nephrol ; 29(3): 977-990, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29371419

RESUMEN

The amiloride-sensitive epithelial sodium channel (ENaC) and the thiazide-sensitive sodium chloride cotransporter (NCC) are key regulators of sodium and potassium and colocalize in the late distal convoluted tubule of the kidney. Loss of the αENaC subunit leads to a perinatal lethal phenotype characterized by sodium loss and hyperkalemia resembling the human syndrome pseudohypoaldosteronism type 1 (PHA-I). In adulthood, inducible nephron-specific deletion of αENaC in mice mimics the lethal phenotype observed in neonates, and as in humans, this phenotype is prevented by a high sodium (HNa+)/low potassium (LK+) rescue diet. Rescue reflects activation of NCC, which is suppressed at baseline by elevated plasma potassium concentration. In this study, we investigated the role of the γENaC subunit in the PHA-I phenotype. Nephron-specific γENaC knockout mice also presented with salt-wasting syndrome and severe hyperkalemia. Unlike mice lacking αENaC or ßΕΝaC, an HNa+/LK+ diet did not normalize plasma potassium (K+) concentration or increase NCC activation. However, when K+ was eliminated from the diet at the time that γENaC was deleted, plasma K+ concentration and NCC activity remained normal, and progressive weight loss was prevented. Loss of the late distal convoluted tubule, as well as overall reduced ßENaC subunit expression, may be responsible for the more severe hyperkalemia. We conclude that plasma K+ concentration becomes the determining and limiting factor in regulating NCC activity, regardless of Na+ balance in γENaC-deficient mice.


Asunto(s)
Canales Epiteliales de Sodio/genética , Hiperpotasemia/genética , Potasio/sangre , Seudohipoaldosteronismo/sangre , Seudohipoaldosteronismo/genética , Animales , Quelantes/uso terapéutico , Suplementos Dietéticos , Hiperpotasemia/sangre , Hiperpotasemia/tratamiento farmacológico , Ratones , Ratones Noqueados , Nefronas , Poliestirenos/uso terapéutico , Potasio en la Dieta/administración & dosificación , Sodio en la Dieta/administración & dosificación , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo
4.
Physiology (Bethesda) ; 32(2): 112-125, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28202622

RESUMEN

Hypertension affects over 1.2 billion individuals worldwide and has become the most critical and expensive public health problem. Hypertension is a multifactorial disease involving environmental and genetic factors together with risk-conferring behaviors. The cause of the disease is identified in ∼10% of the cases (secondary hypertension), but in 90% of the cases no etiology is found (primary or essential hypertension). For this reason, a better understanding of the mechanisms controlling blood pressure in normal and hypertensive patients is the aim of very active experimental and clinical research. In this article, we review the importance of the renin-angiotensin-aldosterone system (RAAS) for the control of blood pressure, focusing on the evolution of the system and its critical importance for adaptation of vertebrates to a terrestrial and dry environment. The evolution of blood pressure control during the evolution of primates, hominins, and humans is discussed, together with the role of common genetic factors and the possible causes of the current hypertension pandemic in the light of evolutionary medicine.


Asunto(s)
Evolución Biológica , Hipertensión/epidemiología , Pandemias , Sistema Renina-Angiotensina , Animales , Presión Sanguínea , Interacción Gen-Ambiente , Humanos , Hipertensión/genética , Hipertensión/fisiopatología , Riñón/metabolismo , Riñón/fisiopatología , Potasio en la Dieta/metabolismo , Sodio en la Dieta/metabolismo
5.
J Am Soc Nephrol ; 27(8): 2309-18, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26701978

RESUMEN

Systemic pseudohypoaldosteronism type 1 (PHA-1) is a severe salt-losing syndrome caused by loss-of-function mutations of the amiloride-sensitive epithelial sodium channel (ENaC) and characterized by neonatal life-threatening hypovolemia and hyperkalemia. The very high plasma aldosterone levels detected under hypovolemic or hyperkalemic challenge can lead to increased or decreased sodium reabsorption, respectively, through the Na(+)/Cl(-) cotransporter (NCC). However, the role of ENaC deficiency remains incompletely defined, because constitutive inactivation of individual ENaC subunits is neonatally lethal in mice. We generated adult inducible nephron-specific αENaC-knockout mice (Scnn1a(Pax8/LC1)) that exhibit hyperkalemia and body weight loss when kept on a regular-salt diet, thus mimicking PHA-1. Compared with control mice fed a regular-salt diet, knockout mice fed a regular-salt diet exhibited downregulated expression and phosphorylation of NCC protein, despite high plasma aldosterone levels. In knockout mice fed a high-sodium and reduced-potassium diet (rescue diet), although plasma aldosterone levels remained significantly increased, NCC expression returned to control levels, and body weight, plasma and urinary electrolyte concentrations, and excretion normalized. Finally, shift to a regular diet after the rescue diet reinstated the symptoms of severe PHA-1 syndrome and significantly reduced NCC phosphorylation. In conclusion, lack of ENaC-mediated sodium transport along the nephron cannot be compensated for by other sodium channels and/or transporters, only by a high-sodium and reduced-potassium diet. We further conclude that hyperkalemia becomes the determining factor in regulating NCC activity, regardless of sodium loss, in the ENaC-mediated salt-losing PHA-1 phenotype.


Asunto(s)
Canales Epiteliales de Sodio/genética , Hiperpotasemia/genética , Seudohipoaldosteronismo/genética , Animales , Ratones , Ratones Noqueados , Nefronas , Índice de Severidad de la Enfermedad
6.
J Am Soc Nephrol ; 25(7): 1453-64, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24480829

RESUMEN

Aldosterone promotes electrogenic sodium reabsorption through the amiloride-sensitive epithelial sodium channel (ENaC). Here, we investigated the importance of ENaC and its positive regulator channel-activating protease 1 (CAP1/Prss8) in colon. Mice lacking the αENaC subunit in colonic superficial cells (Scnn1a(KO)) were viable, without fetal or perinatal lethality. Control mice fed a regular or low-salt diet had a significantly higher amiloride-sensitive rectal potential difference (∆PDamil) than control mice fed a high-salt diet. In Scnn1a(KO) mice, however, this salt restriction-induced increase in ∆PDamil did not occur, and the circadian rhythm of ∆PDamil was blunted. Plasma and urinary sodium and potassium did not change with regular or high-salt diets or potassium loading in control or Scnn1a(KO) mice. However, Scnn1a(KO) mice fed a low-salt diet lost significant amounts of sodium in their feces and exhibited high plasma aldosterone and increased urinary sodium retention. Mice lacking the CAP1/Prss8 in colonic superficial cells (Prss8(KO)) were viable, without fetal or perinatal lethality. Compared with controls, Prss8(KO) mice fed regular or low-salt diets exhibited significantly reduced ∆PDamil in the afternoon, but the circadian rhythm was maintained. Prss8(KO) mice fed a low-salt diet also exhibited sodium loss through feces and higher plasma aldosterone levels. Thus, we identified CAP1/Prss8 as an in vivo regulator of ENaC in colon. We conclude that, under salt restriction, activation of the renin-angiotensin-aldosterone system in the kidney compensated for the absence of ENaC in colonic surface epithelium, leading to colon-specific pseudohypoaldosteronism type 1 with mineralocorticoid resistance without evidence of impaired potassium balance.


Asunto(s)
Aldosterona/metabolismo , Colon/metabolismo , Canales Epiteliales de Sodio/fisiología , Sodio/metabolismo , Animales , Canales Epiteliales de Sodio/deficiencia , Femenino , Masculino , Ratones , Serina Endopeptidasas/fisiología
7.
Am J Physiol Renal Physiol ; 304(11): F1390-7, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23515718

RESUMEN

Since nitric oxide (NO) participates in the renal regulation of blood pressure, in part, by modulating transport of Na⁺ and Cl⁻ in the kidney, we asked whether NO regulates net Cl⁻ flux (JCl) in the cortical collecting duct (CCD) and determined the transporter(s) that mediate NO-sensitive Cl⁻ absorption. Cl⁻ absorption was measured in CCDs perfused in vitro that were taken from aldosterone-treated mice. Administration of an NO donor (10 µM MAHMA NONOate) reduced JCl and transepithelial voltage (VT) both in the presence or absence of angiotensin II. However, reducing endogenous NO production by inhibiting NO synthase (100 µM N(G)-nitro-L-arginine methyl ester) increased JCl only in the presence of angiotensin II, suggesting that angiotensin II stimulates NO synthase activity. To determine the transport process that mediates NO-sensitive changes in JCl, we examined the effect of NO on JCl following either genetic ablation or chemical inhibition of transporters in the CCD. Since the application of hydrochlorothiazide (100 µM) or bafilomycin (5 nM) to the perfusate or ablation of the gene encoding pendrin did not alter NO-sensitive JCl, NO modulates JCl independent of the Na⁺-dependent Cl⁻/HCO3⁻ exchanger (NDCBE, Slc4a8), the A cell apical plasma membrane H⁺-ATPase and pendrin. In contrast, both total and NO-sensitive JCl and VT were abolished with application of an epithelial Na(+) channel (ENaC) inhibitor (3 µM benzamil) to the perfusate. We conclude that NO reduces Cl⁻ absorption in the CCD through a mechanism that is ENaC-dependent.


Asunto(s)
Cloruros/metabolismo , Canales Epiteliales de Sodio/fisiología , Túbulos Renales Colectores/metabolismo , Óxido Nítrico/fisiología , Absorción/fisiología , Aldosterona/administración & dosificación , Amilorida/análogos & derivados , Amilorida/farmacología , Angiotensina II/farmacología , Animales , Proteínas de Transporte de Anión/deficiencia , Proteínas de Transporte de Anión/fisiología , Antiportadores de Cloruro-Bicarbonato/fisiología , Canales Epiteliales de Sodio/efectos de los fármacos , Canales Epiteliales de Sodio/genética , Femenino , Técnicas In Vitro , Masculino , Ratones , Ratones Noqueados , NG-Nitroarginina Metil Éster/farmacología , Donantes de Óxido Nítrico/administración & dosificación , Óxido Nítrico Sintasa/antagonistas & inhibidores , Sodio/farmacología , Transportadores de Sulfato
8.
Annu Rev Physiol ; 71: 361-79, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18928407

RESUMEN

The study of human monogenic diseases [pseudohypoaldosteronism type 1 (PHA-1) and Liddle's syndrome] as well as mouse models mimicking the salt-losing syndrome (PHA-1) or salt-sensitive hypertension (Liddle's syndrome) have established the epithelial sodium channel ENaC as a limiting factor in vivo in the control of ionic composition of the extracellular fluid, regulation of blood volume and blood pressure, lung alveolar clearance, and airway mucociliary clearance. In this review, we discuss more specifically the activation of ENaC by serine proteases. Recent in vitro and in vivo experiments indicate that membrane-bound serine proteases are of critical importance in the activation of ENaC in different organs, such as the kidney, the lung, or the cochlea. Progress in understanding the basic mechanism of proteolytic activation of ENaC is accelerating, but uncertainty about the most fundamental aspects persists, leaving numerous still-unanswered questions.


Asunto(s)
Canales Epiteliales de Sodio/fisiología , Serina Endopeptidasas/fisiología , Animales , Cóclea/citología , Cóclea/fisiología , Líquido Extracelular/fisiología , Humanos , Riñón/citología , Riñón/fisiología , Pulmón/citología , Pulmón/fisiología
9.
Am J Physiol Renal Physiol ; 303(1): F45-55, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22496413

RESUMEN

In cortical collecting ducts (CCDs) perfused in vitro, inhibiting the epithelial Na(+) channel (ENaC) reduces Cl(-) absorption. Since ENaC does not transport Cl(-), the purpose of this study was to determine how ENaC modulates Cl(-) absorption. Thus, Cl(-) absorption was measured in CCDs perfused in vitro that were taken from mice given aldosterone for 7 days. In wild-type mice, we observed no effect of luminal hydrochlorothiazide on either Cl(-) absorption or transepithelial voltage (V(T)). However, application of an ENaC inhibitor [benzamil (3 µM)] to the luminal fluid or application of a Na(+)-K(+)-ATPase inhibitor to the bath reduced Cl(-) absorption by ∼66-75% and nearly obliterated lumen-negative V(T). In contrast, ENaC inhibition had no effect in CCDs from collecting duct-specific ENaC-null mice (Hoxb7:CRE, Scnn1a(loxlox)). Whereas benzamil-sensitive Cl(-) absorption did not depend on CFTR, application of a Na(+)-K(+)-2Cl(-) cotransport inhibitor (bumetanide) to the bath or ablation of the gene encoding Na(+)-K(+)-2Cl(-) cotransporter 1 (NKCC1) blunted benzamil-sensitive Cl(-) absorption, although the benzamil-sensitive component of V(T) was unaffected. In conclusion, first, in CCDs from aldosterone-treated mice, most Cl(-) absorption is benzamil sensitive, whereas thiazide-sensitive Cl(-) absorption is undetectable. Second, benzamil-sensitive Cl(-) absorption occurs by inhibition of ENaC, possibly due to elimination of lumen-negative V(T). Finally, benzamil-sensitive Cl(-) flux occurs, at least in part, through transcellular transport through a pathway that depends on NKCC1.


Asunto(s)
Cloruros/metabolismo , Bloqueadores del Canal de Sodio Epitelial , Túbulos Renales Colectores/metabolismo , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Absorción/efectos de los fármacos , Aldosterona/farmacología , Amilorida/análogos & derivados , Amilorida/farmacología , Animales , Transporte Biológico/efectos de los fármacos , Canales Epiteliales de Sodio/genética , Canales Epiteliales de Sodio/metabolismo , Hidroclorotiazida/farmacología , Túbulos Renales Colectores/efectos de los fármacos , Ratones , Ratones Noqueados , Sodio/metabolismo , Inhibidores de los Simportadores del Cloruro de Sodio/farmacología , Miembro 2 de la Familia de Transportadores de Soluto 12
10.
J Am Soc Nephrol ; 22(2): 253-61, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21051735

RESUMEN

Lithium-induced nephrogenic diabetes insipidus (NDI) is accompanied by polyuria, downregulation of aquaporin 2 (AQP2), and cellular remodeling of the collecting duct (CD). The amiloride-sensitive epithelial sodium channel (ENaC) is a likely candidate for lithium entry. Here, we subjected transgenic mice lacking αENaC specifically in the CD (knockout [KO] mice) and littermate controls to chronic lithium treatment. In contrast to control mice, KO mice did not markedly increase their water intake. Furthermore, KO mice did not demonstrate the polyuria and reduction in urine osmolality induced by lithium treatment in the control mice. Lithium treatment reduced AQP2 protein levels in the cortex/outer medulla and inner medulla (IM) of control mice but only partially reduced AQP2 levels in the IM of KO mice. Furthermore, lithium induced expression of H(+)-ATPase in the IM of control mice but not KO mice. In conclusion, the absence of functional ENaC in the CD protects mice from lithium-induced NDI. These data support the hypothesis that ENaC-mediated lithium entry into the CD principal cells contributes to the pathogenesis of lithium-induced NDI.


Asunto(s)
Diabetes Insípida Nefrogénica/inducido químicamente , Canales Epiteliales de Sodio/fisiología , Cloruro de Litio/toxicidad , Absorción , Animales , Acuaporina 2/análisis , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/patología , Cloruro de Litio/farmacocinética , Ratones , Ratones Noqueados , ATPasas de Translocación de Protón/análisis
11.
Physiol Genomics ; 43(13): 844-54, 2011 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-21558422

RESUMEN

Despite large changes in salt intake, the mammalian kidney is able to maintain the extracellular sodium concentration and osmolarity within very narrow margins, thereby controlling blood volume and blood pressure. In the aldosterone-sensitive distal nephron (ASDN), aldosterone tightly controls the activities of epithelial sodium channel (ENaC) and Na,K-ATPase, the two limiting factors in establishing transepithelial sodium transport. It has been proposed that the ENaC/degenerin gene family is restricted to Metazoans, whereas the α- and ß-subunits of Na,K-ATPase have homologous genes in prokaryotes. This raises the question of the emergence of osmolarity control. By exploring recent genomic data of diverse organisms, we found that: 1) ENaC/degenerin exists in all of the Metazoans screened, including nonbilaterians and, by extension, was already present in ancestors of Metazoa; 2) ENaC/degenerin is also present in Naegleria gruberi, an eukaryotic microbe, consistent with either a vertical inheritance from the last common ancestor of Eukaryotes or a lateral transfer between Naegleria and Metazoan ancestors; and 3) The Na,K-ATPase ß-subunit is restricted to Holozoa, the taxon that includes animals and their closest single-cell relatives. Since the ß-subunit of Na,K-ATPase plays a key role in targeting the α-subunit to the plasma membrane and has an additional function in the formation of cell junctions, we propose that the emergence of Na,K-ATPase, together with ENaC/degenerin, is linked to the development of multicellularity in the Metazoan kingdom. The establishment of multicellularity and the associated extracellular compartment ("internal milieu") precedes the emergence of other key elements of the aldosterone signaling pathway.


Asunto(s)
Aldosterona/metabolismo , Canales Epiteliales de Sodio/genética , Evolución Molecular , ATPasa Intercambiadora de Sodio-Potasio/genética , Sodio/metabolismo , Canales Iónicos Sensibles al Ácido , Animales , Canales de Sodio Degenerina , Humanos , Transporte Iónico/efectos de los fármacos , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Fosfoproteínas/genética , Filogenia
12.
J Biol Chem ; 285(35): 26945-26955, 2010 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-20566636

RESUMEN

Studies in cystic fibrosis patients and mice overexpressing the epithelial Na(+) channel beta-subunit (betaENaC-Tg) suggest that raised airway Na(+) transport and airway surface liquid (ASL) depletion are central to the pathogenesis of cystic fibrosis lung disease. However, patients or mice with Liddle gain-of-function betaENaC mutations exhibit hypertension but no lung disease. To investigate this apparent paradox, we compared the airway phenotype (nasal versus tracheal) of Liddle with CFTR-null, betaENaC-Tg, and double mutant mice. In mouse nasal epithelium, the region that functionally mimics human airways, high levels of CFTR expression inhibited Liddle epithelial Nat channel (ENaC) hyperfunction. Conversely, in mouse trachea, low levels of CFTR failed to suppress Liddle ENaC hyperfunction. Indeed, Na(+) transport measured in Ussing chambers ("flooded" conditions) was raised in both Liddle and betaENaC-Tg mice. Because enhanced Na(+) transport did not correlate with lung disease in these mutant mice, measurements in tracheal cultures under physiologic "thin film" conditions and in vivo were performed. Regulation of ASL volume and ENaC-mediated Na(+) absorption were intact in Liddle but defective in betaENaC-Tg mice. We conclude that the capacity to regulate Na(+) transport and ASL volume, not absolute Na(+) transport rates in Ussing chambers, is the key physiologic function protecting airways from dehydration-induced lung disease.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Canales Epiteliales de Sodio/biosíntesis , Regulación de la Expresión Génica , Síndrome de Liddle/metabolismo , Mutación , Sodio/metabolismo , Animales , Fibrosis Quística/metabolismo , Fibrosis Quística/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Canales Epiteliales de Sodio/genética , Humanos , Transporte Iónico/genética , Síndrome de Liddle/patología , Ratones , Ratones Noqueados , Mucosa Nasal/metabolismo , Mucosa Nasal/patología , Técnicas de Cultivo de Órganos , Tráquea/metabolismo , Tráquea/patología
13.
Pflugers Arch ; 462(6): 871-83, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21938401

RESUMEN

Inositol Inpp5k (or Pps, SKIP) is a member of the inositol polyphosphate 5-phosphatases family with a poorly characterized function in vivo. In this study, we explored the function of this inositol 5-phosphatase in mice and cells overexpressing the 42-kDa mouse Inpp5k protein. Inpp5k transgenic mice present defects in water metabolism characterized by a reduced plasma osmolality at baseline, a delayed urinary water excretion following a water load, and an increased acute response to vasopressin. These defects are associated with the expression of the Inpp5k transgene in renal collecting ducts and with alterations in the arginine vasopressin/aquaporin-2 signalling pathway in this tubular segment. Analysis in a mouse collecting duct mCCD cell line revealed that Inpp5k overexpression leads to increased expression of the arginine vasopressin receptor type 2 and increased cAMP response to arginine vasopressin, providing a basis for increased aquaporin-2 expression and plasma membrane localization with increased osmotically induced water transport. Altogether, our results indicate that Inpp5k 5-phosphatase is important for the control of the arginine vasopressin/aquaporin-2 signalling pathway and water transport in kidney collecting ducts.


Asunto(s)
Acuaporina 2/metabolismo , Túbulos Renales Colectores/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Vasopresinas/metabolismo , Equilibrio Hidroelectrolítico/fisiología , Animales , Células Cultivadas , Femenino , Humanos , Túbulos Renales Colectores/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Monoéster Fosfórico Hidrolasas/genética , Transducción de Señal/fisiología , Agua/metabolismo
14.
Kidney Int ; 79(8): 843-52, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21178974

RESUMEN

Water balance is achieved through the ability of the kidney to control water reabsorption in the connecting tubule and the collecting duct. In a mouse cortical collecting duct cell line (mCCD(c11)), physiological concentrations of arginine vasopressin increased both electrogenic, amiloride-sensitive, epithelial sodium channel (ENaC)-mediated sodium transport measured by the short-circuit current (Isc) method and water flow (Jv apical to basal) measured by gravimetry with similar activation coefficient K(1/2) (6 and 12 pM, respectively). Jv increased linearly according to the osmotic gradient across the monolayer. A small but highly significant Jv was also measured under isoosmotic conditions. To test the coupling between sodium reabsorption and water flow, mCCD(c11) cells were treated for 24 h under isoosmotic condition with either diluent, amiloride, vasopressin or vasopressin and amiloride. Isc, Jv, and net chemical sodium fluxes were measured across the same monolayers. Around 30% of baseline and 50% of vasopressin-induced water flow is coupled to an amiloride-sensitive, ENaC-mediated, electrogenic sodium transport, whereas the remaining flow is coupled to an amiloride-insensitive, nonelectrogenic sodium transport mediated by an unknown electroneutral transporter. The mCCD(c11) cell line is a first example of a mammalian tight epithelium allowing quantitative study of the coupling between sodium and water transport. Our data are consistent with the 'near isoosmotic' fluid transport model.


Asunto(s)
Arginina Vasopresina/metabolismo , Túbulos Renales Colectores/fisiología , Sodio/metabolismo , Agua/metabolismo , Animales , Acuaporinas/metabolismo , Arginina Vasopresina/farmacología , Línea Celular , Fenómenos Electrofisiológicos , Canales Epiteliales de Sodio/metabolismo , Transporte Iónico/efectos de los fármacos , Corteza Renal/citología , Corteza Renal/efectos de los fármacos , Corteza Renal/fisiología , Túbulos Renales Colectores/citología , Túbulos Renales Colectores/efectos de los fármacos , Ratones , Ósmosis
15.
J Am Soc Nephrol ; 21(11): 1942-51, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20947633

RESUMEN

Mutations in α, ß, or γ subunits of the epithelial sodium channel (ENaC) can downregulate ENaC activity and cause a severe salt-losing syndrome with hyperkalemia and metabolic acidosis, designated pseudohypoaldosteronism type 1 in humans. In contrast, mice with selective inactivation of αENaC in the collecting duct (CD) maintain sodium and potassium balance, suggesting that the late distal convoluted tubule (DCT2) and/or the connecting tubule (CNT) participates in sodium homeostasis. To investigate the relative importance of ENaC-mediated sodium absorption in the CNT, we used Cre-lox technology to generate mice lacking αENaC in the aquaporin 2-expressing CNT and CD. Western blot analysis of microdissected cortical CD (CCD) and CNT revealed absence of αENaC in the CCD and weak αENaC expression in the CNT. These mice exhibited a significantly higher urinary sodium excretion, a lower urine osmolality, and an increased urine volume compared with control mice. Furthermore, serum sodium was lower and potassium levels were higher in the genetically modified mice. With dietary sodium restriction, these mice experienced significant weight loss, increased urinary sodium excretion, and hyperkalemia. Plasma aldosterone levels were significantly elevated under both standard and sodium-restricted diets. In summary, αENaC expression within the CNT/CD is crucial for sodium and potassium homeostasis and causes signs and symptoms of pseudohypoaldosteronism type 1 if missing.


Asunto(s)
Canales Epiteliales de Sodio/metabolismo , Túbulos Renales Colectores/metabolismo , Túbulos Renales/metabolismo , Potasio/metabolismo , Sodio/metabolismo , Aldosterona/sangre , Animales , Acuaporina 2/metabolismo , Canales Epiteliales de Sodio/genética , Femenino , Homeostasis/fisiología , Corteza Renal/citología , Corteza Renal/efectos de los fármacos , Corteza Renal/metabolismo , Túbulos Renales/citología , Túbulos Renales/efectos de los fármacos , Túbulos Renales Colectores/citología , Túbulos Renales Colectores/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Sodio en la Dieta/farmacología
16.
Am J Physiol Renal Physiol ; 299(6): F1473-85, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20861076

RESUMEN

Aldosterone and corticosterone bind to mineralocorticoid (MR) and glucocorticoid receptors (GR), which, upon ligand binding, are thought to translocate to the cell nucleus to act as transcription factors. Mineralocorticoid selectivity is achieved by the 11ß-hydroxysteroid dehydrogenase type 2 (11ß-HSD2) that inactivates 11ß-hydroxy glucocorticoids. High expression levels of 11ß-HSD2 characterize the aldosterone-sensitive distal nephron (ASDN), which comprises the segment-specific cells of late distal convoluted tubule (DCT2), connecting tubule (CNT), and collecting duct (CD). We used MR- and GR-specific antibodies to study localization and regulation of MR and GR in kidneys of rats with altered plasma aldosterone and corticosterone levels. In control rats, MR and GR were found in cell nuclei of thick ascending limb (TAL), DCT, CNT, CD cells, and intercalated cells (IC). GR was also abundant in cell nuclei and the subapical compartment of proximal tubule (PT) cells. Dietary NaCl loading, which lowers plasma aldosterone, caused a selective removal of GR from cell nuclei of 11ß-HSD2-positive ASDN. The nuclear localization of MR was unaffected. Adrenalectomy (ADX) resulted in removal of MR and GR from the cell nuclei of all epithelial cells. Aldosterone replacement rapidly relocated the receptors in the cell nuclei. In ASDN cells, low-dose corticosterone replacement caused nuclear localization of MR, but not of GR. The GR was redistributed to the nucleus only in PT, TAL, early DCT, and IC that express no or very little 11ß-HSD2. In ASDN cells, nuclear GR localization was only achieved when corticosterone was replaced at high doses. Thus ligand-induced nuclear translocation of MR and GR are part of MR and GR regulation in the kidney and show remarkable segment- and cell type-specific characteristics. Differential regulation of MR and GR may alter the level of heterodimerization of the receptors and hence may contribute to the complexity of corticosteroid effects on ASDN function.


Asunto(s)
Túbulos Renales Distales/efectos de los fármacos , Transporte de Proteínas , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Adrenalectomía , Aldosterona/metabolismo , Aldosterona/farmacología , Animales , Especificidad de Anticuerpos , Corticosterona/administración & dosificación , Corticosterona/metabolismo , Corticosterona/farmacología , Túbulos Renales Distales/metabolismo , Masculino , Ratones , Nefronas/metabolismo , ARN Mensajero/metabolismo , Ratas , Receptores de Glucocorticoides/inmunología , Receptores de Mineralocorticoides/inmunología , Sodio en la Dieta/administración & dosificación , Sodio en la Dieta/farmacología
17.
Function (Oxf) ; 1(2): zqaa024, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33201937

RESUMEN

The Coronavirus Disease 2019 (COVID-19) pandemic remains a serious public health problem and will continue to be until effective drugs and/or vaccines are available. The rational development of drugs critically depends on our understanding of disease mechanisms, that is, the physiology and pathophysiology underlying the function of the organ targeted by the virus. Since the beginning of the pandemic, tireless efforts around the globe have led to numerous publications on the virus, its receptor, its entry into the cell, its cytopathic effects, and how it triggers innate and native immunity but the role of apical sodium transport mediated by the epithelial sodium channel (ENaC) during the early phases of the infection in the airways has received little attention. We propose a pathophysiological model that defines the possible role of ENaC in this process.


Asunto(s)
COVID-19 , Humanos , Sodio/metabolismo , Transporte Biológico , Transporte Iónico , Canales Epiteliales de Sodio/metabolismo
18.
Kidney Int ; 76(1): 44-53, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19367330

RESUMEN

Lithium therapy frequently induces nephrogenic diabetes insipidus; amiloride appears to prevent its occurrence in some clinical cases. Amiloride blocks the epithelial sodium channel (ENaC) located in the apical membrane of principal cells; hence one possibility is that ENaC is the main entry site for lithium and the beneficial effect of amiloride may be through inhibiting lithium entry. Using a mouse collecting duct cell line, we found that vasopressin caused an increase in Aquaporin 2 (AQP2) expression which was reduced by clinically relevant lithium concentrations similar to what is seen with in vivo models of this disease. Further amiloride or benzamil administration prevented this lithium-induced downregulation of AQP2. Amiloride reduced transcellular lithium transport, intracellular lithium concentration, and lithium-induced inactivation of glycogen synthase kinase 3beta. Treatment of rats with lithium downregulated AQP2 expression, reduced the principal-to-intercalated cell ratio, and caused polyuria, while simultaneous administration of amiloride attenuated all these changes. These results show that ENaC is the major entry site for lithium in principal cells both in vitro and in vivo. Blocking lithium entry with amiloride attenuates lithium-induced diabetes insipidus, thus providing a rationale for its use in treating this disorder.


Asunto(s)
Amilorida/farmacología , Diabetes Insípida Nefrogénica/metabolismo , Litio/metabolismo , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/metabolismo , Animales , Acuaporina 2/metabolismo , Línea Celular , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Inmunohistoquímica , Concentración 50 Inhibidora , Túbulos Renales Colectores/metabolismo , Masculino , Ratones , Ratas , Ratas Wistar , Canales de Sodio/genética
20.
Nephrol Dial Transplant ; 23(5): 1636-41, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18424465

RESUMEN

BACKGROUND: Type 1 pseudohypoaldosteronism (PHA1) is a salt-wasting syndrome caused by mineralocorticoid resistance. Autosomal recessive and dominant hereditary forms are caused by Epithelial Na Channel and Mineralocorticoid Receptor mutation respectively, while secondary PHA1 is usually associated with urological problems. METHODS: Ten patients were studied in four French pediatric units in order to characterize PHA1 spectrum in infants. Patients were selected by chart review. Genetic, clinical and biochemistry data were collected and analyzed. RESULTS: Autosomal recessive PHA1 (n = 3) was diagnosed at 6 and 7 days of life in three patients presenting with severe hyperkalaemia and weight loss. After 8 months, 3 and 5 years on follow-up, neurological development and longitudinal growth was normal with high sodium supplementation. Autosomal dominant PHA1 (n = 4) was revealed at 15, 19, 22 and 30 days of life because of failure to thrive. At 8 months, 3 and 21 years of age, longitudinal growth was normal in three patients who were given salt supplementation; no significant catch-up growth was obtained in the last patient at 20 months of age. Secondary PHA1 (n = 3) was diagnosed at 11, 26 days and 5 months of life concomitantly with acute pyelonephritis in three children with either renal hypoplasia, urinary duplication or bilateral megaureter. The outcome was favourable and salt supplementation was discontinued after 3, 11 and 13 months. CONCLUSIONS: PHA1 should be suspected in case of severe hyperkalemia and weight loss in infants and need careful management. Pathogenesis of secondary PHA1 is still challenging and further studies are mandatory to highlight the link between infection, developing urinary tract and pseudohypoaldosteronism.


Asunto(s)
Seudohipoaldosteronismo/diagnóstico , Canales Epiteliales de Sodio/química , Canales Epiteliales de Sodio/genética , Femenino , Genes Dominantes , Genes Recesivos , Humanos , Lactante , Recién Nacido , Masculino , Modelos Moleculares , Mutación , Seudohipoaldosteronismo/clasificación , Seudohipoaldosteronismo/etiología , Seudohipoaldosteronismo/genética , Pielonefritis/complicaciones , Receptores de Mineralocorticoides/genética , Estudios Retrospectivos , Sistema Urinario/anomalías
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA