Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 300(4): 105785, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38401845

RESUMEN

The epithelial sodium channel (ENaC) is essential for mediating sodium absorption in several epithelia. Its impaired function leads to severe disorders, including pseudohypoaldosteronism type 1 and respiratory distress. Therefore, pharmacological ENaC activators have potential therapeutic implications. Previously, a small molecule ENaC activator (S3969) was developed. So far, little is known about molecular mechanisms involved in S3969-mediated ENaC stimulation. Here, we identified an S3969-binding site in human ENaC by combining structure-based simulations with molecular biological methods and electrophysiological measurements of ENaC heterologously expressed in Xenopus laevis oocytes. We confirmed a previous observation that the extracellular loop of ß-ENaC is essential for ENaC stimulation by S3969. Molecular dynamics simulations predicted critical residues in the thumb domain of ß-ENaC (Arg388, Phe391, and Tyr406) that coordinate S3969 within a binding site localized at the ß-γ-subunit interface. Importantly, mutating each of these residues reduced (R388H; R388A) or nearly abolished (F391G; Y406A) the S3969-mediated ENaC activation. Molecular dynamics simulations also suggested that S3969-mediated ENaC stimulation involved a movement of the α5 helix of the thumb domain of ß-ENaC away from the palm domain of γ-ENaC. Consistent with this, the introduction of two cysteine residues (ßR437C - γS298C) to form a disulfide bridge connecting these two domains prevented ENaC stimulation by S3969 unless the disulfide bond was reduced by DTT. Finally, we demonstrated that S3969 stimulated ENaC endogenously expressed in cultured human airway epithelial cells (H441). These new findings may lead to novel (patho-)physiological and therapeutic concepts for disorders associated with altered ENaC function.


Asunto(s)
Agonistas del Canal de Sodio Epitelial , Canales Epiteliales de Sodio , Indoles , Animales , Humanos , Sitios de Unión , Agonistas del Canal de Sodio Epitelial/metabolismo , Agonistas del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/química , Canales Epiteliales de Sodio/metabolismo , Simulación de Dinámica Molecular , Oocitos/efectos de los fármacos , Xenopus laevis , Unión Proteica , Indoles/metabolismo , Indoles/farmacología
2.
J Pharmacol Exp Ther ; 379(2): 156-165, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34465632

RESUMEN

Acute respiratory distress syndrome (ARDS), a common and fatal clinical condition, is characterized by the destruction of epithelium and augmented permeability of the alveolar-capillary barrier. Resolvin conjugates in tissue regeneration 1 (RCTR1) is an endogenous lipid mediator derived from docosahexaenoic acid , exerting proresolution effects in the process of inflammation. In our research, we evaluated the role of RCTR1 in alveolar fluid clearance (AFC) in lipopolysaccharide-induced ARDS/acute lung injury (ALI) rat model. Rats were injected with RCTR1 (5 µg/kg) via caudal veins 8 hours after lipopolysaccharide (LPS) (14 mg/kg) treatment, and then AFC was estimated after 1 hour of ventilation. Primary type II alveolar epithelial cells were incubated with LPS (1 ug/ml) with or without RCTR1 (10 nM) for 8 hours. Our results showed that RCTR1 significantly enhanced the survival rate, promoted the AFC, and alleviated LPS-induced ARDS/ALI in vivo. Furthermore, RCTR1 remarkably elevated the protein expression of sodium channels and Na, K-ATPase and the activity of Na, K-ATPase in vivo and in vitro. Additionally, RCTR1 also decreased neural precursor cell expressed developmentally downregulated 4-2 (Nedd4-2) level via upregulating Ser473-phosphorylated-Akt expression. Besides this, inhibitors of receptor for lipoxin A4 (ALX), cAMP, and phosphatidylinositol 3-kinase (PI3K) (BOC-2, KH-7, and LY294002) notably inhibited the effects of RCTR1 on AFC. In summary, RCTR1 enhances the protein levels of sodium channels and Na, K-ATPase and the Na, K-ATPase activity to improve AFC in ALI through ALX/cAMP/PI3K/Nedd4-2 pathway, suggesting that RCTR1 may become a therapeutic drug for ARDS/ALI. SIGNIFICANCE STATEMENT: RCTR1, an endogenous lipid mediator, enhanced the rate of AFC to accelerate the resolution of inflammation in the LPS-induced murine lung injury model. RCTR1 upregulates the expression of epithelial sodium channels (ENaCs) and Na, K-ATPase in vivo and in vitro to accelerate the AFC. The efficacy of RCTR1 on the ENaC and Na, K-ATPase level was in an ALX/cAMP/PI3K/Nedd4-2-dependent manner.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Ácidos Docosahexaenoicos/farmacología , Agonistas del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/metabolismo , Alveolos Pulmonares/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/tratamiento farmacológico , Animales , Ácidos Docosahexaenoicos/análogos & derivados , Ácidos Docosahexaenoicos/uso terapéutico , Lipopolisacáridos/toxicidad , Masculino , Alveolos Pulmonares/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
3.
Biol Pharm Bull ; 43(4): 725-730, 2020 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-32009028

RESUMEN

Epithelial sodium channel (ENaC) is an amiloride-sensitive sodium ion channel that is expressed in epithelial tissues. ENaC overexpression and/or hyperactivation in airway epithelial cells cause sodium over-absorption and dysregulated ciliary movement for mucus clearance; however, the agents that suppress constitutive airway ENaC activation are yet to be clinically available. Here, we focused on macrolides, which are widely used antibiotics that have many potential immunomodulatory effects. We examined whether macrolides could modulate constitutive ENaC activity and downstream events that typify cystic fibrosis (CF) and chronic obstructive pulmonary diseases (COPD) in in vitro and in vivo models of ENaC overexpression. Treatment of ENaC-overexpressing human bronchial epithelial cells (ß/γENaC-16HBE14o- cells) with three macrolides (erythromycin, clarithromycin, azithromycin) confirmed dose-dependent suppression of ENaC function. For in vivo studies, mice harboring airway specific ßENaC overexpression (C57BL/6J-ßENaC-transgenic mice) were treated orally with azithromycin, a well-established antimicrobial agent that has been widely prescribed. Azithromycin treatment modulated pulmonary mechanics, emphysematous phenotype and pulmonary dysfunction. Notably, a lower dose (3 mg kg-1) of azithromycin significantly increased forced expiratory volume in 0.1 s (FEV0.1), an inverse indicator of bronchoconstriction. Although not statistically significant, improvement of pulmonary obstructive parameters such as emphysema and lung dysfunction (FEV0.1%) was observed. Our results demonstrate that macrolides directly attenuate constitutive ENaC function in vitro and may be promising for the treatment of obstructive lung diseases with defective mucociliary clearance, possibly by targeting ENaC hyperactivation.


Asunto(s)
Antibacterianos/farmacología , Azitromicina/farmacología , Agonistas del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/fisiología , Animales , Línea Celular , Canales Epiteliales de Sodio/genética , Volumen Espiratorio Forzado , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiología , Masculino , Ratones Transgénicos , Capacidad Vital
4.
Biosci Biotechnol Biochem ; 83(2): 243-250, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30343635

RESUMEN

The epithelial sodium channel (ENaC) plays a pivotal role in sodium homeostasis, and the development of drugs that modulate ENaC activity is of great potential therapeutic relevance. We screened 6100 chemicals for their ability to activate sodium permeability of ENaC. We used a two-step strategy: a high throughput cell-based assay and an electrophysiological assay. Five compounds were identified showing common structural features including an indole or benzothiophene ring. ENaC consists of three subunits: α, ß, and γ. Changing the heteromeric combination of human and mouse ENaC αßγ subunits, we found that all five compounds activated the human ß subunit but not the mouse subunit. However, four of them exhibited lower activity when the human γ subunit was substituted by the mouse γ subunit. Our findings provide a structural basis for designing human ENaC activity modulators. Abbreviations: ENaC: Epithelial sodium channel; ΔRFU: delta relative fluorescence units; EC50: Half-maximal effective concentration; Emax: maximum effect value.


Asunto(s)
Agonistas del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/efectos de los fármacos , Indoles/química , Tiofenos/química , Animales , Agonistas del Canal de Sodio Epitelial/química , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones
5.
Br J Pharmacol ; 175(8): 1305-1317, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28409833

RESUMEN

BACKGROUND AND PURPOSE: Our recent studies show that the reduced activity of epithelial sodium channels (ENaC) in endothelial cells accounts for the adaptation of vasculature to salt in Sprague-Dawley rats. The present study examines a hypothesis that enhanced ENaC activity mediates the loss of vasorelaxation in Dahl salt-sensitive (SS) rats. EXPERIMENTAL APPROACH: We used the cell-attached patch-clamp technique to record ENaC activity in split-open mesenteric arteries. Western blot and immunofluorescence staining were used to evaluate the levels of aldosterone, ENaC, eNOS and NO. Blood pressure was measured with the tail-cuff method and the artery relaxation was measured with the wire myograph assay. KEY RESULTS: High-salt (HS) diet significantly increased plasma aldosterone and ENaC activity in the endothelial cells of Dahl SS rats. The endothelium-dependent artery relaxation was blunted by HS challenge in these rats. Amiloride, a potent blocker of ENaC, increased both phosphorylated eNOS and NO and therefore prevented the HS-induced loss of vasorelaxation. As, in SS rats, endogenous aldosterone was already elevated by HS challenge, exogenous aldosterone did not further elevate ENaC activity in the rats fed with HS. Eplerenone, a mineralocorticoid receptor antagonist, attenuated the effects of HS on both ENaC activity and artery relaxation. CONCLUSIONS AND IMPLICATIONS: These data suggest that HS diet blunts artery relaxation and causes hypertension via a pathway associated with aldosterone-dependent activation of ENaC in endothelial cells. This pathway provides one of the mechanisms by which HS causes hypertension in Dahl SS rats. LINKED ARTICLES: This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Agonistas del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/fisiología , Cloruro de Sodio Dietético/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Células Cultivadas , Células Endoteliales/fisiología , Masculino , Arterias Mesentéricas/citología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ratas Endogámicas Dahl , Vasodilatación/efectos de los fármacos
6.
Channels (Austin) ; 11(5): 388-398, 2017 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-28636485

RESUMEN

Renal sodium reabsorption depends on the activity of the Na+,K+-ATPase α/ß heterodimer. Four α (α1-4) and 3 ß (ß1-3) subunit isoforms have been described. It is accepted that renal tubule cells express α1/ß1 dimers. Aldosterone stimulates Na+,K+-ATPase activity and may modulate α1/ß1 expression. However, some studies suggest the presence of ß3 in the kidney. We hypothesized that the ß3 isoform of the Na+,K+-ATPase is expressed in tubular cells of the distal nephron, and modulated by mineralocorticoids. We found that ß3 is highly expressed in collecting duct of rodents, and that mineralocorticoids decreased the expression of ß3. Thus, we describe a novel molecular mechanism of sodium pump modulation that may contribute to the effects of mineralocorticoids on sodium reabsorption.


Asunto(s)
Túbulos Renales/metabolismo , Mineralocorticoides/farmacología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Aldosterona/farmacología , Animales , Línea Celular , Membrana Celular/metabolismo , Agonistas del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/metabolismo , Masculino , Ratas Sprague-Dawley
7.
J Biol Chem ; 291(45): 23440-23451, 2016 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-27645999

RESUMEN

Regulation of the epithelial sodium channel (ENaC), which regulates fluid homeostasis and blood pressure, is complex and remains incompletely understood. The TIP peptide, a mimic of the lectin-like domain of TNF, activates ENaC by binding to glycosylated residues in the extracellular loop of ENaC-α, as well as to a hitherto uncharacterized internal site. Molecular docking studies suggested three residues, Val567, Glu568, and Glu571, located at the interface between the second transmembrane and C-terminal domains of ENaC-α, as a critical site for binding of the TIP peptide. We generated Ala replacement mutants in this region of ENaC-α and examined its interaction with TIP peptide (3M, V567A/E568A/E571A; 2M, V567A/E568A; and 1M, E571A). 3M and 2M ENaC-α, but not 1M ENaC-α, displayed significantly reduced binding capacity to TIP peptide and to TNF. When overexpressed in H441 cells, 3M mutant ENaC-α formed functional channels with similar gating and density characteristics as the WT subunit and efficiently associated with the ß and γ subunits in the plasma membrane. We subsequently assayed for increased open probability time and membrane expression, both of which define ENaC activity, following addition of TIP peptide. TIP peptide increased open probability time in H441 cells overexpressing wild type and 1M ENaC-α channels, but not 3M or 2M ENaC-α channels. On the other hand, TIP peptide-mediated reduction in ENaC ubiquitination was similar in cells overexpressing either WT or 3M ENaC-α subunits. In summary, this study has identified a novel site in ENaC-α that is crucial for activation of the open probability of the channel, but not membrane expression, by the lectin-like domain of TNF.


Asunto(s)
Agonistas del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/metabolismo , Péptidos Cíclicos/farmacología , Línea Celular Tumoral , Canales Epiteliales de Sodio/química , Canales Epiteliales de Sodio/genética , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , Mutación Puntual , Dominios Proteicos/efectos de los fármacos , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ubiquitinación/efectos de los fármacos
8.
J Pharm Sci ; 105(1): 333-8, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26852863

RESUMEN

Vitamin D is implicated in the pathogenesis of asthma, acute lung injury, and other respiratory diseases. 1,25-Dihydroxyvitamin D (1,25(OH)2D3), the hormonal form of vitamin D, has been shown to reduce vascular permeability and ameliorate lung edema. Therefore, we speculate that 1,25(OH)2D3 may regulate alveolar Na(+) transport via targeting epithelial Na(+) channels (ENaC), a crucial pathway for alveolar fluid clearance. In vivo total alveolar fluid clearance was 39.4 ± 3.8% in 1,25(OH)2D3-treated mice, significantly greater than vehicle-treated controls (24.7 ± 1.9 %, n = 10, p < 0.05). 1,25(OH)2D3 increased amiloride-sensitive short-circuit currents in H441 monolayers, and whole-cell patch-clamp data confirmed that ENaC currents in single H441 cell were enhanced in 1,25(OH)2D3-treated cells. Western blot showed that the expression of α-ENaC was significantly elevated in 1,25(OH)2D3-treated mouse lungs and 1,25(OH)2D3-treated H441 cells. These observations suggest that vitamin D augments transalveolar fluid clearance, and vitamin D therapy may potentially be used to ameliorate pulmonary edema.


Asunto(s)
Calcitriol/farmacología , Agonistas del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/biosíntesis , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Animales , Permeabilidad Capilar/efectos de los fármacos , Línea Celular , Canales Epiteliales de Sodio/genética , Agua Pulmonar Extravascular/metabolismo , Pulmón/citología , Pulmón/metabolismo , Ratones , Ratones Endogámicos BALB C , Técnicas de Placa-Clamp , Edema Pulmonar/tratamiento farmacológico , ARN Mensajero/biosíntesis , ARN Mensajero/genética
9.
J Biomol Screen ; 20(2): 242-53, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25278498

RESUMEN

The epithelial sodium channel (ENaC) plays a crucial role in salt and water homeostasis and is primarily involved in sodium reabsorption in the kidney and lung. Modulators of ENaC function, particularly within lung epithelia, could offer potential treatments for a number of diseases. As a constitutively active sodium channel, ENaC expression at the cell membrane is highly regulated through rapid turnover. This short half-life of the channel at the membrane and cytotoxicity from overexpression pose a problem for reagent generation and assay development in drug discovery. We have generated an HEK293 stable cell line expressing ENaC ß and γ subunits containing the PY motif trafficking mutations found in Liddle's syndrome to overcome rapid channel turnover at the membrane. A BacMam virus was used to transiently express the ENaC α subunit to reconstitute channel function to reduce the toxicity associated with long-term overexpression. We have configured a 384-well FLIPR membrane potential antagonist assay for high-throughput screening and an IonWorks Quattro electrophysiology antagonist assay that is predictive of potency values derived from primary lung epithelial cell short-circuit measurements. The triage strategy for compound screening and profiling against this target using these assays has resulted in the discovery of novel chemotypes.


Asunto(s)
Evaluación Preclínica de Medicamentos , Agonistas del Canal de Sodio Epitelial/farmacología , Bloqueadores del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Ensayos Analíticos de Alto Rendimiento/normas , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/química , Canales Epiteliales de Sodio/genética , Expresión Génica , Células HEK293 , Humanos , Potenciales de la Membrana/efectos de los fármacos , Técnicas de Placa-Clamp , Mucosa Respiratoria/metabolismo , Bibliotecas de Moléculas Pequeñas
11.
Curr Mol Pharmacol ; 6(1): 13-27, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23547931

RESUMEN

Lungs contain a particular amount of fluid that is crucial for proper lung function. This fluid content is tightly controlled within certain limits. Fluid accumulation in the alveolar airspace impairs gas exchange and represents a lifethreatening condition referred to as pulmonary edema. Ion transport processes by pulmonary epithelia represent a mechanism, responsible for fluid absorption from the airspace. Thus, it is obvious to consider ion transport processes as target for therapeutic interventions in pulmonary edema. The principle mechanism responsible for fluid absorption from the airspace is: Na(+) diffuses through luminal Na(+) channels into epithelial cells and is extruded by Na(+)/K(+)-ATPases at the basolateral side. This process generates an osmotic gradient that represents the driving force for fluid absorption. The rate of Na(+) absorption is limited by the number/activity of Na+ channels in the luminal membrane of alveolar epithelial cells. Although different Na+ channels have been identified, the epithelial Na+ channel (ENaC) is a major player that participates in Na(+)-driven fluid absorption and thus a suitable target for the treatment of pulmonary edema. This article reviews cellular mechanisms by which ENaC activity can be increased in alveolar epithelia (lectins, proteases, ß-adrenoceptors, mineralo-/glucocorticoid-receptors). These mechanisms are involved in regulating ENaC-dependent fluid absorption under physiological conditions. Additionally, pre-clinical as well as some preliminary clinical studies revealed that "ENaC-activators/stimulators" (ß2-adrenoceptor agonists and mineralo-/glucocorticoid-receptor agonists) could be beneficial for therapeutic interventions in patients with pulmonary edema. However, the outcome of subsequently performed multicenter clinical trials with "ENaC-activators/stimulators" for treatment of patients with pulmonary edema was disappointing.


Asunto(s)
Agonistas del Canal de Sodio Epitelial/uso terapéutico , Canales Epiteliales de Sodio/metabolismo , Edema Pulmonar/tratamiento farmacológico , Agonistas Adrenérgicos beta/farmacología , Agonistas Adrenérgicos beta/uso terapéutico , Agonistas del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/química , Agua Pulmonar Extravascular/efectos de los fármacos , Glucocorticoides/farmacología , Glucocorticoides/uso terapéutico , Humanos , Lectinas/metabolismo , Péptido Hidrolasas/metabolismo , Edema Pulmonar/metabolismo , Edema Pulmonar/patología , Receptores Adrenérgicos beta/química , Receptores Adrenérgicos beta/metabolismo
12.
Curr Mol Pharmacol ; 6(1): 35-43, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23547933

RESUMEN

The epithelial sodium channel (ENaC) plays an essential role in transepithelial sodium reabsorption in the renal connecting tubule and collecting duct. Therefore, controlling ENaC activity is an important regulatory event in electrolyte and extracellular volume homeostasis, and thus in the control of blood pressure. Many independent signaling pathways converge on ENaC, although the most important for its physiological role is the enhancement of channel activity by the steroid hormone aldosterone. In this review, we briefly summarize current knowledge about ENaC regulation and the different chemical compounds available to directly or indirectly modify channel function. In addition, current and possible clinical uses of ENaC and aldosterone antagonists are highlighted.


Asunto(s)
Canales Epiteliales de Sodio/metabolismo , Enfermedades Renales/metabolismo , Aldosterona/farmacología , Aldosterona/uso terapéutico , Diuréticos/uso terapéutico , Agonistas del Canal de Sodio Epitelial/farmacología , Agonistas del Canal de Sodio Epitelial/uso terapéutico , Bloqueadores del Canal de Sodio Epitelial/farmacología , Bloqueadores del Canal de Sodio Epitelial/uso terapéutico , Canales Epiteliales de Sodio/química , Humanos , Hipertensión/tratamiento farmacológico , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/patología , Receptores de Mineralocorticoides/química , Receptores de Mineralocorticoides/metabolismo , Transducción de Señal/efectos de los fármacos , Sodio/metabolismo
13.
Am J Respir Cell Mol Biol ; 48(5): 610-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23470626

RESUMEN

Edema fluid resorption is critical for gas exchange, and both alveolar epithelial sodium channel (ENaC) and Na,K-ATPase are accredited with key roles in the resolution of pulmonary edema. Alveolar fluid clearance (AFC) was measured in in situ ventilated lungs by instilling isosmolar 5% BSA solution with Evans Blue-labeled albumin tracer (5 ml/kg) and measuring the change in Evans Blue-labeled albumin concentration over time. Treatment with lipoxin A4 and lipoxin receptor agonist (5(S), 6(R)-7-trihydroxymethyl 17 heptanoate) significantly stimulated AFC in oleic acid (OA)-induced lung injury, with the outcome of decreased pulmonary edema. Lipoxin A4 and 5(S), 6(R)-7-trihydroxymethyl 17 heptanoate not only up-regulated the ENaC α and ENaC γ subunits protein expression, but also increased Na,K-ATPase α1 subunit protein expression and Na,K-ATPase activity in lung tissues. There was no significant difference of intracellular cAMP level between the lipoxin A4 treatment and OA group. However, the intracellular cGMP level was significantly decreased after lipoxin A4 treatment. The beneficial effects of lipoxin A4 were abrogated by butoxycarbonyl-Phe-Leu-Phe-Leu-Ph (lipoxin A4 receptor antagonist) in OA-induced lung injury. In primary rat alveolar type II epithelial cells stimulated with LPS, lipoxin A4 increased ENaC α and ENaC γ subunits protein expression and Na,K-ATPase activity. Lipoxin A4 stimulated AFC through activation of alveolar epithelial ENaC and Na,K-ATPase.


Asunto(s)
Agonistas del Canal de Sodio Epitelial/administración & dosificación , Canales Epiteliales de Sodio/metabolismo , Lipoxinas/administración & dosificación , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/metabolismo , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/inmunología , Animales , Células Cultivadas , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Agonistas del Canal de Sodio Epitelial/farmacología , Bloqueadores del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/genética , Expresión Génica/efectos de los fármacos , Ácidos Heptanoicos/farmacología , Lipopolisacáridos/farmacología , Lipoxinas/farmacología , Masculino , Depuración Mucociliar , Oligopéptidos/farmacología , Peroxidasa/metabolismo , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/fisiopatología , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/metabolismo
14.
Am J Physiol Lung Cell Mol Physiol ; 304(6): L428-37, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23292809

RESUMEN

We and others have shown that epithelial Na(+) channels (ENaC) in alveolar type 2 (AT2) cells are activated by ß2 agonists, steroid hormones, elevated oxygen tension, and by dopamine. Although acetylcholine receptors (AChRs) have been previously described in the lung, there are few reports of whether cholinergic agonists alter sodium transport in the alveolar epithelium. Therefore, we investigated how cholinergic receptors regulate ENaC activity in primary cultures of rat AT2 cells using cell-attached patch-clamp recordings to assess ENaC activity. We found that the muscarinic agonists, carbachol (CCh) and oxotremorine, activated ENaC in a dose-dependent manner but that nicotine did not. CCh-induced activation of ENaC was blocked by atropine. Western blotting and immunohistochemistry suggested that muscarinic M2 and M3 receptors (mAChRs) but not nicotinic receptors were present in AT2 cells. Endogenous RhoA and GTP-RhoA increased in response to CCh and the increase was reduced by pretreatment with atropine. We showed that Y-27632, an inhibitor of Rho-associated protein kinase (ROCK), abolished endogenous ENaC activity and inhibited the activation of ENaC by CCh. We also showed that ROCK signaling was necessary for ENaC stability in 2F3 cells, a model for AT2 cells. Our results showed that muscarinic agonists activated ENaC in rat AT2 cells through M2 and/or M3 mAChRs probably via a RhoA/ROCK signaling pathway.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Canales Epiteliales de Sodio/metabolismo , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/fisiología , Amidas/farmacología , Animales , Atropina/farmacología , Carbacol/farmacología , Células Cultivadas , Activación Enzimática , Agonistas del Canal de Sodio Epitelial/farmacología , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Agonistas Muscarínicos/farmacología , Antagonistas Muscarínicos/farmacología , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Oxotremorina/farmacología , Técnicas de Placa-Clamp , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptor Muscarínico M2/metabolismo , Receptor Muscarínico M2/fisiología , Receptor Muscarínico M3/metabolismo , Receptor Muscarínico M3/fisiología , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
15.
Br J Pharmacol ; 168(6): 1329-40, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23083067

RESUMEN

BACKGROUND AND PURPOSE: Pharmacological enhancement of vectorial Na⁺ transport may be useful to increase alveolar fluid clearance. Herein, we investigated the influence of the benzimidazolones 1-ethyl-1,3-dihydro-2-benzimidazolone (1-EBIO), 5,6-dichloro-1-EBIO (DC-EBIO) and chlorzoxazone on vectorial epithelial Na⁺ transport. EXPERIMENTAL APPROACH: Effects on vectorial Na⁺ transport and amiloride-sensitive apical membrane Na⁺ permeability were determined by measuring short-circuit currents (I(SC)) in rat fetal distal lung epithelial (FDLE) monolayers. Furthermore, amiloride-sensitive membrane conductance and the open probability of epithelial Na⁺ channels (ENaC) were determined by patch clamp experiments using A549 cells. KEY RESULTS: I(SC) was increased by approximately 50% after addition of 1-EBIO, DC-EBIO and chlorzoxazone. With permeabilized basolateral membranes in the presence of a 145:5 apical to basolateral Na⁺ gradient, the benzimidazolones markedly increased amiloride-sensitive I(SC). 5-(N-Ethyl-N-isopropyl)amiloride-induced inhibition of I(SC) was not affected. The benzamil-sensitive I(SC) was increased in benzimidazolone-stimulated monolayers. Pretreating the apical membrane with amiloride, which inhibits ENaC, completely prevented the stimulating effects of benzimidazolones on I(SC). Furthermore, 1-EBIO (1 mM) and DC-EBIO (0.1 mM) significantly increased (threefold) the open probability of ENaC without influencing current amplitude. Whole cell measurements showed that DC-EBIO (0.1 mM) induced an amiloride-sensitive increase in membrane conductance. CONCLUSION AND IMPLICATIONS: Benzimidazolones have a stimulating effect on vectorial Na⁺ transport. The antagonist sensitivity of this effect suggests the benzimidazolones elicit this action by activating the highly selective ENaC currents. Thus, the results demonstrate a possible new strategy for directly enhancing epithelial Na⁺ transport.


Asunto(s)
Bencimidazoles/farmacología , Clorzoxazona/farmacología , Agonistas del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/metabolismo , Alveolos Pulmonares/efectos de los fármacos , Mucosa Respiratoria/efectos de los fármacos , Animales , Bencimidazoles/antagonistas & inhibidores , Línea Celular , Permeabilidad de la Membrana Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Células Cultivadas , Clorzoxazona/antagonistas & inhibidores , Agonistas del Canal de Sodio Epitelial/antagonistas & inhibidores , Bloqueadores del Canal de Sodio Epitelial/farmacología , Feto/citología , Humanos , Potenciales de la Membrana/efectos de los fármacos , Técnicas de Placa-Clamp , Alveolos Pulmonares/citología , Alveolos Pulmonares/metabolismo , Ratas , Mucosa Respiratoria/citología , Mucosa Respiratoria/metabolismo , Análisis de la Célula Individual , Sodio/metabolismo
17.
Am J Physiol Cell Physiol ; 303(11): C1129-38, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22895258

RESUMEN

Alcohol affects total body sodium balance, but the molecular mechanism of its effect remains unclear. We used single-channel methods to examine how ethanol affects epithelial sodium channels (ENaC) in A6 distal nephron cells. The data showed that ethanol significantly increased both ENaC open probability (P(o)) and the number of active ENaC in patches (N). 1-Propanol and 1-butanol also increased ENaC activity, but iso-alcohols did not. The effects of ethanol were mimicked by acetaldehyde, the first metabolic product of ethanol, but not by acetone, the metabolic product of 2-propanol. Besides increasing open probability and apparent density of active channels, confocal microscopy and surface biotinylation showed that ethanol significantly increased α-ENaC protein in the apical membrane. The effects of ethanol on ENaC P(o) and N were abolished by a superoxide scavenger, 4-hydroxy-2,2,6,6-tetramethylpiperidinyloxy (TEMPOL) and blocked by the phosphatidylinositol 3-kinase inhibitor LY294002. Consistent with an effect of ethanol-induced reactive oxygen species (ROS) on ENaC, primary alcohols and acetaldehyde elevated intracellular ROS, but secondary alcohols did not. Taken together with our previous finding that ROS stimulate ENaC, the current results suggest that ethanol stimulates ENaC by elevating intracellular ROS probably via its metabolic product acetaldehyde.


Asunto(s)
Agonistas del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/efectos de los fármacos , Etanol/farmacología , Especies Reactivas de Oxígeno/metabolismo , Acetaldehído/farmacología , Animales , Línea Celular , Cromonas/farmacología , Óxidos N-Cíclicos/farmacología , Inhibidores Enzimáticos/farmacología , Canales Epiteliales de Sodio/metabolismo , Depuradores de Radicales Libres/farmacología , Morfolinas/farmacología , Nefronas/efectos de los fármacos , Nefronas/metabolismo , Técnicas de Placa-Clamp , Marcadores de Spin , Xenopus laevis
18.
Pflugers Arch ; 464(4): 353-65, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22864553

RESUMEN

Proteolytic processing of the amiloride-sensitive epithelial sodium channel (ENaC) by serine proteases is known to be important for channel activation. Inappropriate ENaC activation by proteases may contribute to the pathophysiology of cystic fibrosis and could be involved in sodium retention and the pathogenesis of arterial hypertension in the context of renal disease. We hypothesized that in addition to serine proteases, cathepsin proteases may activate ENaC. Cathepsin proteases belong to the group of cysteine proteases and play a pathophysiological role in inflammatory diseases. Under pathophysiological conditions, cathepsin-S (Cat-S) may reach ENaC in the apical membrane of epithelial cells. The aim of this study was to investigate the effect of purified Cat-S on human ENaC heterologously expressed in Xenopus laevis oocytes and on ENaC-mediated sodium transport in cultured M-1 mouse renal collecting duct cells. We demonstrated that Cat-S activates amiloride-sensitive whole-cell currents in ENaC-expressing oocytes. The stimulatory effect of Cat-S was preserved at pH 5. ENaC stimulation by Cat-S was associated with the appearance of a γENaC cleavage fragment at the plasma membrane indicating proteolytic channel activation. Mutating two valine residues (V182 and V193) in the critical region of γENaC prevented proteolytic activation of ENaC by Cat-S. Pre-incubation of the oocytes with the Cat-S inhibitor morpholinurea-leucine-homophenylalanine-vinylsulfone-phenyl (LHVS) prevented the stimulatory effect of Cat-S on ENaC. In contrast, LHVS had no effect on ENaC activation by the prototypical serine proteases trypsin and chymotrypsin. Cat-S also stimulated ENaC in differentiated renal epithelial cells. These findings demonstrate that the cysteine protease Cat-S can activate ENaC which may be relevant under pathophysiological conditions.


Asunto(s)
Catepsinas/metabolismo , Agonistas del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/metabolismo , Amilorida/farmacología , Secuencia de Aminoácidos , Animales , Catepsinas/antagonistas & inhibidores , Membrana Celular/metabolismo , Quimotripsina/metabolismo , Dipéptidos/farmacología , Bloqueadores del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/química , Canales Epiteliales de Sodio/genética , Humanos , Transporte Iónico , Ratones , Datos de Secuencia Molecular , Mutación , Proteolisis , Sodio/metabolismo , Sulfonas/farmacología , Tripsina/metabolismo , Valina/genética , Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...