Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 14(9): 2050-2058, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26923587

RESUMEN

Mutations in the filamin A (FlnA) gene are frequently associated with severe arterial abnormalities, although the physiological role for this cytoskeletal element remains poorly understood in vascular cells. We used a conditional mouse model to selectively delete FlnA in smooth muscle (sm) cells at the adult stage, thus avoiding the developmental effects of the knockout. Basal blood pressure was significantly reduced in conscious smFlnA knockout mice. Remarkably, pressure-dependent tone of the resistance caudal artery was lost, whereas reactivity to vasoconstrictors was preserved. Impairment of the myogenic behavior was correlated with a lack of calcium influx in arterial myocytes upon an increase in intraluminal pressure. Notably, the stretch activation of CaV1.2 was blunted in the absence of smFlnA. In conclusion, FlnA is a critical upstream element of the signaling cascade underlying the myogenic tone. These findings allow a better understanding of the molecular basis of arterial autoregulation and associated disease states.


Asunto(s)
Arterias/fisiología , Filaminas/fisiología , Animales , Presión Sanguínea , Señalización del Calcio , Células Cultivadas , Femenino , Riñón/irrigación sanguínea , Masculino , Mecanotransducción Celular , Ratones Noqueados , Desarrollo de Músculos , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/fisiología
2.
Pflugers Arch ; 468(7): 1151-1160, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27023351

RESUMEN

Human mutations in the X-linked FLNA gene are associated with a remarkably diverse phenotype, including severe arterial morphological anomalies. However, the role for filamin A (FlnA) in vascular cells remains partially understood. We used a smooth muscle (sm)-specific conditional mouse model to delete FlnA at the adult stage, thus avoiding the developmental effects of the knock-out. Inactivation of smFlnA in adult mice significantly lowered blood pressure, together with a decrease in pulse pressure. However, both the aorta and carotid arteries showed a major outward hypertrophic remodeling, resistant to losartan, and normally occurring in hypertensive conditions. Notably, arterial compliance was significantly enhanced in the absence of smFlnA. Moreover, reactivity of thoracic aorta rings to a variety of vasoconstrictors was elevated, while basal contractility in response to KCl depolarization was reduced. Enhanced reactivity to the thromboxane A2 receptor agonist U46619 was fully reversed by the ROCK inhibitor Y27632. We discuss the possibility that a reduction in arterial stiffness upon smFlnA inactivation might cause a compensatory increase in conduit artery diameter for normalization of parietal tension, independently of the ROCK pathway. In conclusion, deletion of smFlnA in adult mice recapitulates the vascular phenotype of human bilateral periventricular nodular heterotopia, culminating in aortic dilatation.


Asunto(s)
Arterias Carótidas/metabolismo , Arterias Carótidas/fisiología , Filaminas/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aorta/fisiología , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Arterias Carótidas/efectos de los fármacos , Humanos , Masculino , Ratones , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiología , Fenotipo , Rigidez Vascular/efectos de los fármacos , Rigidez Vascular/fisiología , Vasoconstrictores/farmacología
3.
Cell Rep ; 13(6): 1161-1171, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26526998

RESUMEN

The mechanically activated non-selective cation channel Piezo1 is a determinant of vascular architecture during early development. Piezo1-deficient embryos die at midgestation with disorganized blood vessels. However, the role of stretch-activated ion channels (SACs) in arterial smooth muscle cells in the adult remains unknown. Here, we show that Piezo1 is highly expressed in myocytes of small-diameter arteries and that smooth-muscle-specific Piezo1 deletion fully impairs SAC activity. While Piezo1 is dispensable for the arterial myogenic tone, it is involved in the structural remodeling of small arteries. Increased Piezo1 opening has a trophic effect on resistance arteries, influencing both diameter and wall thickness in hypertension. Piezo1 mediates a rise in cytosolic calcium and stimulates activity of transglutaminases, cross-linking enzymes required for the remodeling of small arteries. In conclusion, we have established the connection between an early mechanosensitive process, involving Piezo1 in smooth muscle cells, and a clinically relevant arterial remodeling.


Asunto(s)
Arterias/metabolismo , Hipertensión/metabolismo , Canales Iónicos/metabolismo , Miocitos del Músculo Liso/metabolismo , Remodelación Vascular , Animales , Arterias/patología , Calcio/metabolismo , Hipertensión/patología , Canales Iónicos/genética , Ratones , Ratones Endogámicos C57BL , Transglutaminasas/metabolismo
4.
EMBO Rep ; 14(12): 1143-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24157948

RESUMEN

Mechanical forces associated with fluid flow and/or circumferential stretch are sensed by renal epithelial cells and contribute to both adaptive or disease states. Non-selective stretch-activated ion channels (SACs), characterized by a lack of inactivation and a remarkably slow deactivation, are active at the basolateral side of renal proximal convoluted tubules. Knockdown of Piezo1 strongly reduces SAC activity in proximal convoluted tubule epithelial cells. Similarly, overexpression of Polycystin-2 (PC2) or, to a greater extent its pathogenic mutant PC2-740X, impairs native SACs. Moreover, PC2 inhibits exogenous Piezo1 SAC activity. PC2 coimmunoprecipitates with Piezo1 and deletion of its N-terminal domain prevents both this interaction and inhibition of SAC activity. These findings indicate that renal SACs depend on Piezo1, but are critically conditioned by PC2.


Asunto(s)
Células Epiteliales/metabolismo , Canales Iónicos/metabolismo , Mecanotransducción Celular , Canales Catiónicos TRPP/metabolismo , Potenciales de Acción , Animales , Sitios de Unión , Células COS , Células Cultivadas , Chlorocebus aethiops , Células Epiteliales/fisiología , Túbulos Renales/citología , Mutación , Unión Proteica , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPP/química , Canales Catiónicos TRPP/genética
5.
Cell Rep ; 1(3): 241-50, 2012 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-22832196

RESUMEN

How renal epithelial cells respond to increased pressure and the link with kidney disease states remain poorly understood. Pkd1 knockout or expression of a PC2 pathogenic mutant, mimicking the autosomal dominant polycystic kidney disease, dramatically enhances mechanical stress-induced tubular apoptotic cell death. We show the presence of a stretch-activated K(+) channel dependent on the TREK-2 K(2P) subunit in proximal convoluted tubule epithelial cells. Our findings further demonstrate that polycystins protect renal epithelial cells against apoptosis in response to mechanical stress, and this function is mediated through the opening of stretch-activated K(2P) channels. Thus, to our knowledge, we establish for the first time, both in vitro and in vivo, a functional relationship between mechanotransduction and mechanoprotection. We propose that this mechanism is at play in other important pathologies associated with apoptosis and in which pressure or flow stimulation is altered, including heart failure or atherosclerosis.


Asunto(s)
Apoptosis , Citoprotección , Activación del Canal Iónico , Mecanotransducción Celular , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Estrés Mecánico , Canales Catiónicos TRPP/metabolismo , Acidosis/metabolismo , Acidosis/patología , Acidosis/fisiopatología , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Células COS , Chlorocebus aethiops , Citoprotección/efectos de los fármacos , Ácidos Docosahexaenoicos/farmacología , Técnicas de Inactivación de Genes , Activación del Canal Iónico/efectos de los fármacos , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Mecanotransducción Celular/efectos de los fármacos , Ratones , Ratones Noqueados , Proteínas Mutantes/metabolismo , Subunidades de Proteína/metabolismo
6.
Lab Invest ; 91(1): 24-32, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20856231

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is a multisystem disorder characterized by renal, hepatic and pancreatic cyst formation and cardiovascular complications. The condition is caused by mutations in the PKD1 or PKD2 gene. In mice with reduced expression of Pkd1, dissecting aneurysms with prominent media thickening have been seen. To study the effect of selective disruption of Pkd1 in vascular smooth muscle cells (SMCs), we have generated mice in which a floxed part of the Pkd1 gene was deleted by Cre under the control of the SM22 promotor (SM22-Pkd1(del/del) mice). Cre activity was confirmed by X-gal staining using lacZ expressing Cre reporter mice (R26R), and quantitative PCR indicated that in the aorta Pkd1 gene expression was strongly reduced, whereas Pkd2 levels remained unaltered. Histopathological analysis revealed cyst formation in pancreas, liver and kidneys as the result of extravascular Cre activity in pancreatic ducts, bile ducts and in the glomerular Bowman's capsule. Remarkably, we did not find any spontaneous gross structural blood vessel abnormalities in mice with somatic Pkd1 gene disruption in SMCs or simultaneous disruption of Pkd1 in SMCs and endothelial cells (ECs). Extensive isometric myographic analysis of the aorta did not reveal differences in response to KCl, acetylcholine, phenylephrin or serotonin, except for a significant increase in contractility induced by phenylephrin on arteries from 40 weeks old Pkd1(del/+) germ-line mice. However, SM22-Pkd1(del/del) mice showed significantly reduced decrease in heart rate on angiotensin II-induced hypertension. The present findings further demonstrate in vivo, that adaptation to hypertension is altered in SM22-Pkd1(del/del) mice.


Asunto(s)
Hipertensión/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Canales Catiónicos TRPP/metabolismo , Animales , Aorta/metabolismo , Aorta/fisiopatología , Presión Sanguínea , Células Endoteliales/metabolismo , Femenino , Frecuencia Cardíaca , Hipertensión/genética , Hipertensión/fisiopatología , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso Vascular/citología , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales Catiónicos TRPP/genética
7.
Cell ; 139(3): 587-96, 2009 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-19879844

RESUMEN

Autosomal-dominant polycystic kidney disease, the most frequent monogenic cause of kidney failure, is induced by mutations in the PKD1 or PKD2 genes, encoding polycystins TRPP1 and TRPP2, respectively. Polycystins are proposed to form a flow-sensitive ion channel complex in the primary cilium of both epithelial and endothelial cells. However, how polycystins contribute to cellular mechanosensitivity remains obscure. Here, we show that TRPP2 inhibits stretch-activated ion channels (SACs). This specific effect is reversed by coexpression with TRPP1, indicating that the TRPP1/TRPP2 ratio regulates pressure sensing. Moreover, deletion of TRPP1 in smooth muscle cells reduces SAC activity and the arterial myogenic tone. Inversely, depletion of TRPP2 in TRPP1-deficient arteries rescues both SAC opening and the myogenic response. Finally, we show that TRPP2 interacts with filamin A and demonstrate that this actin crosslinking protein is critical for SAC regulation. This work uncovers a role for polycystins in regulating pressure sensing.


Asunto(s)
Presión , Canales Catiónicos TRPP/metabolismo , Actinas/metabolismo , Animales , Proteínas Contráctiles/metabolismo , Filaminas , Mecanotransducción Celular , Ratones , Proteínas de Microfilamentos/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Presorreceptores/metabolismo
8.
EMBO J ; 27(22): 3047-55, 2008 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-18923424

RESUMEN

Acid-sensing ion channels (ASICs) are cationic channels activated by extracellular acidosis that are expressed in both central and peripheral nervous systems. Although peripheral ASICs seem to be natural sensors of acidic pain (e.g., in inflammation, ischaemia, lesions or tumours), a direct demonstration is still lacking. We show that approximately 60% of rat cutaneous sensory neurons express ASIC3-like currents. Native as well as recombinant ASIC3 respond synergistically to three different inflammatory signals that are slight acidifications (approximately pH 7.0), hypertonicity and arachidonic acid (AA). Moderate pH, alone or in combination with hypertonicity and AA, increases nociceptors excitability and produces pain suppressed by the toxin APETx2, a specific blocker of ASIC3. Both APETx2 and the in vivo knockdown of ASIC3 with a specific siRNA also have potent analgesic effects against primary inflammation-induced hyperalgesia in rat. Peripheral ASIC3 channels are thus essential sensors of acidic pain and integrators of molecular signals produced during inflammation where they contribute to primary hyperalgesia.


Asunto(s)
Inflamación/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Dolor/metabolismo , Canales de Sodio/metabolismo , Canales Iónicos Sensibles al Ácido , Acidosis/metabolismo , Potenciales de Acción/fisiología , Animales , Ácido Araquidónico/farmacología , Células Cultivadas , Venenos de Cnidarios/metabolismo , Ganglios Espinales/citología , Calor/efectos adversos , Humanos , Soluciones Hipertónicas , Inflamación/metabolismo , Masculino , Proteínas del Tejido Nervioso/genética , Neuronas Aferentes/citología , Neuronas Aferentes/metabolismo , Dimensión del Dolor , Péptidos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Wistar , Piel/efectos de los fármacos , Piel/inervación , Canales de Sodio/genética , Venenos de Araña/metabolismo
9.
J Biol Chem ; 281(3): 1796-807, 2006 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-16234233

RESUMEN

Acid-sensing ion channels (ASICs) are cationic channels activated by extracellular protons. The ASIC3 subunit is largely expressed in the peripheral nervous system, where it contributes to pain perception and to some aspects of mechanosensation. We report here a PDZ-dependent and protein kinase C-modulated association between ASIC3 and the Na+/H+ exchanger regulatory factor-1 (NHERF-1) adaptor protein. We show that NHERF-1 and ASIC3 are co-expressed in dorsal root ganglion neurons. NHERF-1 enhances the ASIC3 peak current in heterologous cells, including F-11 dorsal root ganglion cells, by increasing the amount of channel at the plasma membrane. Perhaps more importantly, we show that the plateau current of ASIC3 can be dramatically increased (10-30-fold) by association with NHERF-1, leading to a significant sustained current at pH 6.6. In the presence of NHERF-1, the ASIC3 subcellular localization is modified, and the channel co-localizes with ezrin, a member of the ezrin-radixin-moesin family of actin-binding proteins, providing the first direct link between ASIC3 and the cortical cytoskeleton. Given the importance of the ASIC3 sustained current in nociceptor excitability, it is likely that NHERF-1 participates in channel functions associated with nociception and mechanosensation.


Asunto(s)
Proteínas Portadoras/fisiología , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Neuronas Aferentes/fisiología , Fosfoproteínas/fisiología , Canales de Sodio/fisiología , Canales Iónicos Sensibles al Ácido , Animales , Células COS , Membrana Celular/fisiología , Chlorocebus aethiops , Ganglios Espinales/fisiología , Nociceptores/fisiología , Ratas , Intercambiadores de Sodio-Hidrógeno
10.
EMBO Rep ; 6(7): 642-8, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15976821

RESUMEN

TREK-1 (KCNK2) is a K(2P) channel that is highly expressed in fetal neurons. This K(+) channel is opened by a variety of stimuli, including membrane stretch and cellular lipids. Here, we show that the expression of TREK-1 markedly alters the cytoskeletal network and induces the formation of actin- and ezrin-rich membrane protrusions. The genetic inactivation of TREK-1 significantly alters the growth cone morphology of cultured embryonic striatal neurons. Cytoskeleton remodelling is crucially dependent on the protein kinase A phosphorylation site S333 and the interactive proton sensor E306, but is independent of channel permeation. Conversely, the actin cytoskeleton tonically represses TREK-1 mechano-sensitivity. Thus, the dialogue between TREK-1 and the actin cytoskeleton might influence both synaptogenesis and neuronal electrogenesis.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Neuronas/metabolismo , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Animales , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Silenciador del Gen , Activación del Canal Iónico , Mecanotransducción Celular , Ratones , Ratones Noqueados , Mutación , Neuronas/química , Neuronas/citología , Fosfoproteínas/metabolismo , Fosforilación , Canales de Potasio de Dominio Poro en Tándem/análisis , Canales de Potasio de Dominio Poro en Tándem/genética , Seudópodos/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...