Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Bases de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Physiol Lung Cell Mol Physiol ; 306(11): L975-85, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24682451

RESUMEN

Mesenchymal stromal cells (MSCs) or their media (MSC-M) were reported to reverse acute lung injury (ALI)-induced decrease of alveolar fluid clearance. To determine the mechanisms by which MSC-M exert their beneficial effects, an in vitro model of alveolar epithelial injury was created by exposing primary rat alveolar epithelial cells (AECs) to hypoxia (3% O2) plus cytomix, a combination of IL-1ß, TNF-α, and IFN-γ. MSC-M were collected from human MSCs exposed for 12 h to either normoxia (MSC-M) or to hypoxia plus cytomix (HCYT-MSC-M). This latter condition was used to model the effect of alveolar inflammation and hypoxia on paracrine secretion of MSCs in the injured lung. Comparison of paracrine soluble factors in MSC media showed that the IL-1 receptor antagonist and prostaglandin E2 were markedly increased while keratinocyte growth factor (KGF) was twofold lower in HCYT-MSC-M compared with MSC-M. In AECs, hypoxia plus cytomix increased protein permeability, reduced amiloride-sensitive short-circuit current (AS-Isc), and also decreased the number of α-epithelial sodium channel (α-ENaC) subunits in the apical membrane. To test the effects of MSC media, MSC-M and HCYT-MSC-M were added for an additional 12 h to AECs exposed to hypoxia plus cytomix. MSC-M and HCYT-MSC-M completely restored epithelial permeability to normal. MSC-M, but not HCYT-MSC-M, significantly prevented the hypoxia plus cytomix-induced decrease of ENaC activity and restored apical α-ENaC channels. Interestingly, KGF-deprived MSC-M were unable to restore amiloride-sensitive sodium transport, indicating a possible role for KGF in the beneficial effect of MSC-M. These results indicate that MSC-M may be a preferable therapeutic option for ALI.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Células Madre Mesenquimatosas/metabolismo , Sodio/metabolismo , Animales , Apoptosis , Transporte Biológico , Hipoxia de la Célula , Permeabilidad de la Membrana Celular , Células Cultivadas , Medios de Cultivo Condicionados , Dinoprostona/metabolismo , Canales Epiteliales de Sodio/metabolismo , Factor 7 de Crecimiento de Fibroblastos/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Masculino , Comunicación Paracrina , Alveolos Pulmonares/patología , Ratas , Ratas Sprague-Dawley
2.
Am J Pathol ; 181(2): 605-15, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22705055

RESUMEN

CAP1/Prss8 is a membrane-bound serine protease involved in the regulation of several different effectors, such as the epithelial sodium channel ENaC, the protease-activated receptor PAR2, the tight junction proteins, and the profilaggrin polypeptide. Recently, the V170D and the G54-P57 deletion mutations within the CAP1/Prss8 gene, identified in mouse frizzy (fr) and rat hairless (fr(CR)) animals, respectively, have been proposed to be responsible for their skin phenotypes. In the present study, we analyzed those mutations, revealing a change in the protein structure, a modification of the glycosylation state, and an overall reduction in the activation of ENaC of the two mutant proteins. In vivo analyses demonstrated that both fr and fr(CR) mutant animals present analogous reduction of embryonic viability, similar histologic aberrations at the level of the skin, and a significant decrease in the activity of ENaC in the distal colon compared with their control littermates. Hairless rats additionally had dehydration defects in skin and intestine and significant reduction in the body weight. In conclusion, we provided molecular and functional evidence that CAP1/Prss8 mutations are accountable for the defects in fr and fr(CR) animals, and we furthermore demonstrate a decreased function of the CAP1/Prss8 mutant proteins. Therefore, fr and fr(CR) animals are suitable models to investigate the consequences of CAP1/Prss8 action on its target proteins in the whole organism.


Asunto(s)
Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Canales Epiteliales de Sodio/metabolismo , Mutación/genética , Serina Endopeptidasas/genética , Piel/embriología , Piel/patología , Secuencia de Aminoácidos , Animales , Peso Corporal , Deshidratación/metabolismo , Deshidratación/patología , Regulación de la Expresión Génica , Células HEK293 , Cabello/patología , Humanos , Patrón de Herencia/genética , Activación del Canal Iónico , Ratones , Modelos Animales , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Especificidad de Órganos , Fenotipo , Estructura Terciaria de Proteína , Ratas , Serina Endopeptidasas/química , Serina Endopeptidasas/metabolismo , Piel/metabolismo , Piel/fisiopatología , Homología Estructural de Proteína , Xenopus
3.
EMBO Mol Med ; 2(1): 26-37, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20043279

RESUMEN

Sodium transport via epithelial sodium channels (ENaC) expressed in alveolar epithelial cells (AEC) provides the driving force for removal of fluid from the alveolar space. The membrane-bound channel-activating protease 1 (CAP1/Prss8) activates ENaC in vitro in various expression systems. To study the role of CAP1/Prss8 in alveolar sodium transport and lung fluid balance in vivo, we generated mice lacking CAP1/Prss8 in the alveolar epithelium using conditional Cre-loxP-mediated recombination. Deficiency of CAP1/Prss8 in AEC induced in vitro a 40% decrease in ENaC-mediated sodium currents. Sodium-driven alveolar fluid clearance (AFC) was reduced in CAP1/Prss8-deficient mice, due to a 48% decrease in amiloride-sensitive clearance, and was less sensitive to beta(2)-agonist treatment. Intra-alveolar treatment with neutrophil elastase, a soluble serine protease activating ENaC at the cell surface, fully restored basal AFC and the stimulation by beta(2)-agonists. Finally, acute volume-overload increased alveolar lining fluid volume in CAP1/Prss8-deficient mice. This study reveals that CAP1 plays a crucial role in the regulation of ENaC-mediated alveolar sodium and water transport and in mouse lung fluid balance.


Asunto(s)
Canales Epiteliales de Sodio/metabolismo , Agua Pulmonar Extravascular/metabolismo , Pulmón/metabolismo , Alveolos Pulmonares/metabolismo , Serina Endopeptidasas/metabolismo , Equilibrio Hidroelectrolítico , Agonistas Adrenérgicos beta/farmacología , Animales , Células Cultivadas , Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/genética , Eliminación de Gen , Expresión Génica , Ratones , Alveolos Pulmonares/citología , Edema Pulmonar/metabolismo , Mucosa Respiratoria/metabolismo , Serina Endopeptidasas/genética , Equilibrio Hidroelectrolítico/efectos de los fármacos
4.
Am J Physiol Lung Cell Mol Physiol ; 294(3): L409-16, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18024719

RESUMEN

Transepithelial alveolar sodium (Na+) transport mediated by the amiloride-sensitive epithelial sodium channel (ENaC) constitutes the driving force for removal of fluid from the alveolar space. To define the role of the beta-ENaC subunit in vivo in the mature lung, we studied a previously established mouse strain harboring a disruption of the beta-ENaC gene locus resulting in low levels of beta-ENaC mRNA expression. Real-time RT-PCR experiments confirmed that beta-ENaC mRNA levels were decreased by >90% in alveolar epithelial cells from homozygous mutant (m/m) mice. beta-ENaC protein was undetected in lung homogenates from m/m mice by Western blotting, but alpha- and gamma-ENaC proteins were increased by 83% and 45%, respectively, compared with wild-type (WT) mice. At baseline, Na+-driven alveolar fluid clearance (AFC) was significantly reduced by 32% in m/m mice. Amiloride at the concentration 1 mM inhibited AFC by 75% and 34% in WT and m/m mice, respectively, whereas a higher concentration (5 mM) induced a 75% inhibition of AFC in both groups. The beta2-agonist terbutaline significantly increased AFC in WT but not in m/m mice. These results show that despite the compensatory increase in alpha- and gamma-ENaC protein expression observed in mutant mouse lung, low expression of beta-ENaC results in a moderate impairment of baseline AFC and in decreased AFC sensitivity to amiloride, suggesting a possible change in the stoichiometry of ENaC channels. Finally, adequate beta-ENaC expression appears to be required for AFC stimulation by beta2-agonists.


Asunto(s)
Canales Epiteliales de Sodio/biosíntesis , Agua Pulmonar Extravascular/metabolismo , Alveolos Pulmonares/metabolismo , Animales , Expresión Génica , Ratones , Ratones Transgénicos , Mutación , Subunidades de Proteína/biosíntesis , Alveolos Pulmonares/efectos de los fármacos , ARN Mensajero/metabolismo , Terbutalina/farmacología
5.
J Physiol ; 582(Pt 2): 777-88, 2007 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-17430990

RESUMEN

Transepithelial sodium transport via alveolar epithelial Na(+) channels and Na(+),K(+)-ATPase constitutes the driving force for removal of alveolar oedema fluid. Decreased activity of the amiloride-sensitive epithelial Na(+) channel (ENaC) in the apical membrane of alveolar epithelial cells impairs sodium-driven alveolar fluid clearance (AFC) and predisposes to pulmonary oedema. We hypothesized that hyperactivity of ENaC in the distal lung could improve AFC and facilitate the resolution of pulmonary oedema. AFC and lung fluid balance were studied at baseline and under conditions of hydrostatic pulmonary oedema in the beta-Liddle (L) mouse strain harbouring a gain-of-function mutation (R(566)(stop)) within the Scnn1b gene. As compared with wild-type (+/+), baseline AFC was increased by 2- and 3-fold in heterozygous (+/L) and homozygous mutated (L/L) mice, respectively, mainly due to increased amiloride-sensitive AFC. The beta(2)-agonist terbutaline stimulated AFC in +/+ and +/L mice, but not in L/L mice. Acute volume overload induced by saline infusion (40% of body weight over 2 h) significantly increased extravascular (i.e. interstitial and alveolar) lung water as assessed by the bloodless wet-to-dry lung weight ratio in +/+ and L/L mice, as compared with baseline. However, the increase was significantly larger in +/+ than in L/L groups (P=0.01). Volume overload also increased the volume of the alveolar epithelial lining fluid in +/+ mice, indicating the presence of alveolar oedema, but not in L/L mice. Cardiac function as evaluated by echocardiography was comparable in both groups. These data show that constitutive ENaC activation improved sodium-driven AFC in the mouse lung, and attenuated the severity of hydrostatic pulmonary oedema.


Asunto(s)
Líquidos Corporales/metabolismo , Canales Epiteliales de Sodio/genética , Mutación , Alveolos Pulmonares/metabolismo , Edema Pulmonar/fisiopatología , Agonistas Adrenérgicos beta/farmacología , Amilorida/farmacología , Animales , Bronquios/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/antagonistas & inhibidores , Diuréticos/farmacología , Canales Epiteliales de Sodio/metabolismo , Glicina/análogos & derivados , Glicina/farmacología , Corazón/fisiopatología , Presión Hidrostática , Pulmón/patología , Ratones , Ratones Mutantes , Ratones Transgénicos , Isoformas de Proteínas/metabolismo , Alveolos Pulmonares/efectos de los fármacos , Edema Pulmonar/genética , Edema Pulmonar/patología , Índice de Severidad de la Enfermedad , Terbutalina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA