Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Am J Physiol Cell Physiol ; 303(11): C1156-72, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23034388

RESUMEN

Previous studies in pulmonary artery smooth muscle cells (PASMCs) showed that acute hypoxia activates capacitative Ca(2+) entry (CCE) but the molecular candidate(s) mediating CCE caused by acute hypoxia remain unclear. The present study aimed to determine if transient receptor potential canonical 1 (TRPC1) and Orai1 interact with stromal interacting molecule 1 (STIM1) and mediate CCE caused by acute hypoxia in mouse PASMCs. In primary cultured PASMCs loaded with fura-2, acute hypoxia caused a transient followed by a sustained rise in intracellular Ca(2+) concentration ([Ca(2+)](i)). The transient but not sustained rise in [Ca(2+)](i) was partially inhibited by nifedipine. Acute hypoxia also increased the rate of Mn(2+) quench of fura-2 fluorescence that was inhibited by SKF 96365, Ni(2+), La(3+), and Gd(3+), exhibiting pharmacological properties characteristic of CCE. The nifedipine-insensitive rise in [Ca(2+)](i) and the increase in Mn(2+) quench rate were both inhibited in cells treated with TRPC1 antibody or TRPC1 small interfering (si)RNA, in STIM1 siRNA-transfected cells and in Orai1 siRNA-transfected cells. Moreover, overexpression of STIM1 resulted in a marked increase in [Ca(2+)](i) and Mn(2+) quench rate caused by acute hypoxia, and they were reduced in cells treated with TRPC1 antibody and in cells transfected with Orai1 siRNA. Furthermore, TRPC1 and Orai1 coimmunoprecipitated with STIM1 and the precipitation levels of TRPC1 and Orai1 were increased in cells exposed to acute hypoxia. Immunostaining showed colocalizations of TRPC1-STIM1 and Orai1-STIM1, and the colocalizations of these proteins were more apparent in acute hypoxia. These data provide direct evidence that TRPC1 and Orai1 channels mediate CCE through activation of STIM1 in acute hypoxic mouse PASMCs.


Asunto(s)
Canales de Calcio/fisiología , Calcio/fisiología , Glicoproteínas de Membrana/fisiología , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/fisiología , Arteria Pulmonar/fisiopatología , Canales Catiónicos TRPC/fisiología , Animales , Anticuerpos Neutralizantes/farmacología , Calcio/análisis , Bloqueadores de los Canales de Calcio/farmacología , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Células Cultivadas , Gadolinio/farmacología , Silenciador del Gen , Imidazoles/farmacología , Lantano/farmacología , Masculino , Manganeso/química , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Níquel/farmacología , Nifedipino/farmacología , Proteína ORAI1 , Arteria Pulmonar/efectos de los fármacos , Molécula de Interacción Estromal 1 , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/inmunología
2.
Am J Physiol Cell Physiol ; 299(5): C1079-90, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20739625

RESUMEN

Previous studies in mouse pulmonary arterial smooth muscle cells (PASMCs) showed that cannonical transient receptor potential channel TRPC1 and stromal interaction molecule 1 (STIM1) mediate the sustained component of capacitative Ca(2+) entry (CCE), but the molecular candidate(s) that mediate the transient component of CCE remain unknown. The aim of the present study was to examine whether Orai1 mediates the transient component of CCE through activation of STIM1 in mouse PASMCs. In primary cultured mouse PASMCs loaded with fura-2, cyclopiazonic acid (CPA) caused a transient followed by a sustained rise in intracellular Ca(2+) concentration ([Ca(2+)](i)). The transient but not the sustained rise in [Ca(2+)](i) was partially inhibited by nifedipine. The nifedipine-insensitive transient rise in [Ca(2+)](i) and the increase in Mn(2+) quench of fura-2 fluorescence caused by CPA were both reduced in cells treated with Orai1 siRNA. These responses to CPA were further reduced in cells treated with Orai1 and STIM1 small interfering (si)RNA. Moreover, overexpression of STIM1 enhanced the rise in [Ca(2+)](i) and the increase in Mn(2+) quench of fura-2 fluorescence caused by CPA, and these responses were reduced in cells treated with Orai1 siRNA. RT-PCR revealed Orai1 and STIM1 mRNAs, and Western blot analysis identified Orai1 and STIM1 proteins in mouse PASMCs. Furthermore, Orai1 was found to coimmunoprecipitate with STIM1, and the precipitation level of Orai1 was increased in cells subjected to store-depletion. Immunostaining revealed colocalization of Orai1 and STIM1 proteins, and the colocalization of these proteins was more apparent after store-depletion. These data provide direct evidence that the transient component of CCE is mediated by Orai1 channel as a result of STIM1 activation in mouse PASMCs.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Glicoproteínas de Membrana/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Arteria Pulmonar/citología , Animales , Canales de Calcio/genética , Células Cultivadas , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/citología , Proteína ORAI1 , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Molécula de Interacción Estromal 1 , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo
3.
Adv Exp Med Biol ; 661: 123-35, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20204727

RESUMEN

Capacitative calcium entry (CCE) through store-operated channels (SOCs) has been shown to contribute to the rise in intracellular calcium concentration ([Ca(2+)](i)) and mediate pulmonary artery smooth muscle contraction. CCE is activated as a result of depletion of intracellular Ca(2+) stores but there is a great deal of controversy surrounding the underlying signal that active CCE and the molecular makeup of SOCs. The discovery of canonical subgroup of transient receptor potential channels (TRPC) and recent identification of stromal-interacting molecule 1 (STIM1) protein have opened a door to the study of the identity of SOCs and the signal that activates these channels. Among all the TRPC channels, TRPC1 is widely studied in many cell types and shown to be part of SOCs components, whereas STIM1 protein is found to act as a Ca(2+) sensor in the intracellular Ca(2+) stores and activates SOCs. However, there is very little evidence for the roles of TRPC1 and STIM1 in the contribution of CCE in pulmonary artery. This chapter outlines the roles of TRPC1 and STIM1 in pulmonary artery smooth muscle cells and discusses our recent findings that TRPC1 and STIM1 are functionally interact with each other to mediate CCE in these cells. We also propose a model for the molecular makeup of SOCs formed by TRPC1 and STIM1 in pulmonary artery.


Asunto(s)
Calcio/metabolismo , Proteínas de la Membrana/metabolismo , Isoformas de Proteínas/metabolismo , Arteria Pulmonar/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Humanos , Proteínas de la Membrana/genética , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Isoformas de Proteínas/genética , Arteria Pulmonar/citología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal/fisiología , Canales Catiónicos TRPC/genética
4.
Adv Exp Med Biol ; 661: 237-47, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20204734

RESUMEN

Volume-sensitive outwardly rectifying anion channels (VSOACs) are expressed in pulmonary artery smooth muscle cells (PASMCs) and have been implicated in cell proliferation, growth, apoptosis and protection against oxidative stress. In this chapter, we review the properties of native VSOACs in PASMCs, and consider the evidence that ClC-3, a member of the ClC superfamily of voltage dependent Cl- channels, may be responsible for native VSOACs in PASMCs. Finally, we examine whether or not native VSOACs and heterologously expressed ClC-3 channels function as bona fide chloride channels or as chloride/proton antiporters.


Asunto(s)
Canales de Cloruro/metabolismo , Pulmón/irrigación sanguínea , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , Animales , Aniones/metabolismo , Línea Celular , Cloruros/metabolismo , Humanos , Pulmón/metabolismo , Miocitos del Músculo Liso/citología , Protones , Circulación Pulmonar/fisiología
5.
J Mol Cell Cardiol ; 48(1): 211-9, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19615374

RESUMEN

Native volume-sensitive outwardly rectifying anion channels (VSOACs) play a significant role in cell volume homeostasis in mammalian cells. However, the molecular correlate of VSOACs has been elusive to identify. The short isoform of ClC-3 (sClC-3) is a member of the mammalian ClC gene family and has been proposed to be a molecular candidate for VSOACs in cardiac myocytes and vascular smooth muscle cells. To directly test this hypothesis, and assess the physiological role of ClC-3 in cardiac function, we generated a novel line of cardiac-specific inducible ClC-3 knock-out mice. These transgenic mice were maintained on a doxycycline diet to preserve ClC-3 expression; removal of doxycycline activates Cre recombinase to inactivate the Clcn3 gene. Echocardiography revealed dramatically reduced ejection fraction and fractional shortening, and severe signs of myocardial hypertrophy and heart failure in the knock-out mice at both 1.5 and 3 weeks off doxycycline. In mice off doxycycline, time-dependent inactivation of ClC-3 gene expression was confirmed in atrial and ventricular cells by qRT-PCR and Western blot analysis. Electrophysiological examination of native VSOACs in isolated atrial and ventricular myocytes 3 weeks off doxycycline revealed a complete elimination of the currents, whereas at 1.5 weeks, VSOAC current densities were significantly reduced, compared to age-matched control mice maintained on doxycycline. These results indicate that ClC-3 is a key component of native VSOACs in mammalian heart and plays a significant cardioprotective role against cardiac hypertrophy and failure.


Asunto(s)
Cardiomegalia/genética , Canales de Cloruro/metabolismo , Corazón/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Animales , Western Blotting , Encéfalo/metabolismo , Células Cultivadas , Canales de Cloruro/genética , Eliminación de Gen , Inmunohistoquímica , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa
6.
J Physiol ; 587(Pt 11): 2429-42, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19332490

RESUMEN

Previous studies in pulmonary arterial smooth muscle cells (PASMCs) showed that the TRPC1 channel mediates capacitative Ca(2+) entry (CCE), but the molecular signal(s) that activate TRPC1 in PASMCs remains unknown. The aim of the present study was to determine if TRPC1 mediates CCE through activation of STIM1 protein in mouse PASMCs. In primary cultured mouse PASMCs loaded with fura-2, cyclopiazonic acid (CPA) caused a transient followed by a sustained rise in intracellular Ca(2+) concentration ([Ca(2+)](i)). The transient but not the sustained rise in [Ca(2+)](i) was partially inhibited by nifedipine. In addition, CPA increased the rate of Mn(2+) quench of fura-2 fluorescence that was inhibited by SKF 96365, Ni(2+), La(3+) and Gd(3+), exhibiting pharmacological properties characteristic of CCE. The nifedipine-insensitive sustained rise in [Ca(2+)](i) and the increase in Mn(2+) quench of fura-2 fluorescence caused by CPA were both inhibited in cells pretreated with antibody raised against an extracellular epitope of TRPC1. Moreover, STIM1 siRNA reduced the rise in [Ca(2+)](i) and Mn(2+) quench of fura-2 fluorescence caused by CPA, whereas overexpression of STIM1 resulted in a marked increase in these responses. RT-PCR revealed TRPC1 and STIM1 mRNAs, and Western blot analysis identified TRPC1 and STIM1 proteins in mouse PASMCs. Furthermore, TRPC1 was found to co-immunoprecipitate with STIM1, and the precipitation level of TRPC1 was increased in cells subjected to store depletion. Taken together, store depletion causes activation of voltage-operated Ca(2+) entry and CCE. These data provide direct evidence that CCE is mediated by TRPC1 channel through activation of STIM1 in mouse PASMCs.


Asunto(s)
Señalización del Calcio , Glicoproteínas de Membrana/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Canales Catiónicos TRPC/metabolismo , Aminoquinolinas/farmacología , Animales , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio , ATPasas Transportadoras de Calcio/antagonistas & inhibidores , ATPasas Transportadoras de Calcio/metabolismo , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Gadolinio/metabolismo , Indoles/farmacología , Lantano/metabolismo , Magnesio/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Potenciales de la Membrana , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/enzimología , Níquel/metabolismo , Nifedipino/farmacología , Unión Proteica , Arteria Pulmonar/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , Molécula de Interacción Estromal 1 , Canales Catiónicos TRPC/genética , Factores de Tiempo , Transfección
7.
Clin Exp Pharmacol Physiol ; 36(4): 386-93, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18986326

RESUMEN

1. ClC-3 has been proposed as a molecular candidate responsible for volume-sensitive outwardly rectifying anion channels (VSOAC) in cardiac and smooth muscle cells. To further test this hypothesis, we produced a novel line of transgenic mice with cardiac-specific overexpression of the human short ClC-3 isoform (hsClC-3). 2. Northern and western blot analyses demonstrated that mRNA and protein levels of the short isoform (sClC-3) in the heart were significantly increased in hsClC-3-overexpressing (OE) mice compared with wild-type (WT) mice. Heart weight : bodyweight ratios for OE mice were significantly smaller compared with age-matched WT mice. 3. Electrocardiogram recordings indicated no difference at rest, whereas echocardiographic recordings revealed consistent reductions in left ventricular diastolic diameter, left ventricular posterior wall thickness at end of diastole and interventricular septum thickness in diastole in OE mice. 4. The VSOAC current densities in atrial cardiomyocytes were significantly increased by ClC-3 overexpression compared with WT cells. No differences in VSOAC current properties in OE and WT atrial myocytes were observed in terms of outward rectification, anion permeability (I(-) > Cl(-) > Asp(-)) and inhibition by 4,4'-diisothiocyanatostilbene-2,2'-disulphonic acid and glibenclamide. The VSOAC in atrial myocytes from both groups were totally abolished by phorbol-12,13-dibutyrate (a protein kinase C activator) and by intracellular dialysis of an N-terminal anti-ClC-3 antibody. 5. Cardiac cell volume measurements revealed a significant acceleration of the rate of regulatory volume decrease (RVD) in OE myocytes compared with WT. 6. In conclusion, enhanced VSOAC currents and acceleration of the time-course of RVD in atrial myocytes of OE mice is strong evidence supporting an essential role of sClC-3 in native VSOAC function in mouse atrial myocytes.


Asunto(s)
Canales de Cloruro/genética , Miocardio/metabolismo , Animales , Función Atrial/genética , Canales de Cloruro/metabolismo , Electrofisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Especificidad de Órganos/genética , Técnicas de Placa-Clamp , Fenotipo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Regulación hacia Arriba/genética
8.
Vascul Pharmacol ; 50(3-4): 89-97, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19084078

RESUMEN

Caffeine is a well described and characterized ryanodine receptor (RyR) activator. Previous evidence from independent research studies also indicate caffeine inhibits InsP3 receptor functionality, which is important to activation of capacitative Ca2+ entry (CCE) in some cell types. In addition, RyR activation elicits excitatory-coupled Ca2+ entry (ECCE) in skeletal muscle myotubes. Recent studies by our group show that canine pulmonary arterial smooth muscle cells (PASMCs) have functional InsP3 receptors as well as RyRs, and that CCE is dependent on InsP3 receptor activity. The potential for caffeine to activate ECCE as well as inhibit InsP3 receptor function and CCE was examined using fura-2 fluorescent imaging in canine PASMCs. The data show caffeine causes transient as well as sustained cytosolic Ca2+ increases, though this is not due to CCE or ECCE activity as evidenced by a lack of an increase in Mn2+ quench of fura-2. The experiments also show caffeine reversibly inhibits 5-HT elicited-InsP3 mediated Ca2+ responses with an IC50 of 6.87x10(-4) M and 10 mM caffeine fully inhibits CCE. These studies provide the first evidence that caffeine is an inhibitor of InsP3 generated Ca2+ signals and CCE in PASMCs.


Asunto(s)
Cafeína/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Calcio/metabolismo , Inositol 1,4,5-Trifosfato/antagonistas & inhibidores , Miocitos del Músculo Liso/efectos de los fármacos , Arteria Pulmonar/metabolismo , Animales , Señalización del Calcio/efectos de los fármacos , Línea Celular , Perros , Femenino , Humanos , Técnicas In Vitro , Masculino , Miocitos del Músculo Liso/metabolismo , Serotonina/farmacología
9.
Am J Physiol Cell Physiol ; 294(1): C313-23, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17977940

RESUMEN

Previous studies have shown that, in acutely dispersed canine pulmonary artery smooth muscle cells (PASMCs), depletion of both functionally independent inositol 1,4,5-trisphosphate (IP(3))- and ryanodine-sensitive Ca(2+) stores activates capacitative Ca(2+) entry (CCE). The present study aimed to determine if cell culture modifies intracellular Ca(2+) stores and alters Ca(2+) entry pathways caused by store depletion and hypoxia in canine PASMCs. Intracellular Ca(2+) concentration ([Ca(2+)](i)) was measured in fura 2-loaded cells. Mn(2+) quench of fura 2 signal was performed to study divalent cation entry, and the effects of hypoxia were examined under oxygen tension of 15-18 mmHg. In acutely isolated PASMCs, depletion of IP(3)-sensitive Ca(2+) stores with cyclopiazonic acid (CPA) did not affect initial caffeine-induced intracellular Ca(2+) transients but abolished 5-HT-induced Ca(2+) transients. In contrast, CPA significantly reduced caffeine- and 5-HT-induced Ca(2+) transients in cultured PASMCs. In cultured PASMCs, store depletion or hypoxia caused a transient followed by a sustained rise in [Ca(2+)](i). The transient rise in [Ca(2+)](i) was partially inhibited by nifedipine, whereas the nifedipine-insensitive transient rise in [Ca(2+)](i) was inhibited by KB-R7943, a selective inhibitor of reverse mode Na(+)/Ca(2+) exchanger (NCX). The nifedipine-insensitive sustained rise in [Ca(2+)](i) was inhibited by SKF-96365, Ni(2+), La(3+), and Gd(3+). In addition, store depletion or hypoxia increased the rate of Mn(2+) quench of fura 2 fluorescence that was also inhibited by these blockers, exhibiting pharmacological properties characteristic of CCE. We conclude that cell culture of canine PASMCs reorganizes IP(3) and ryanodine receptors into a common intracellular Ca(2+) compartment, and depletion of this store or hypoxia activates voltage-operated Ca(2+) entry, reverse mode NCX, and CCE.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio , Calcio/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Actinas/metabolismo , Animales , Cafeína/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/efectos de los fármacos , ATPasas Transportadoras de Calcio/antagonistas & inhibidores , ATPasas Transportadoras de Calcio/metabolismo , Hipoxia de la Célula , Células Cultivadas , Perros , Inhibidores Enzimáticos/farmacología , Imidazoles/farmacología , Indoles/farmacología , Inositol 1,4,5-Trifosfato/metabolismo , Contracción Muscular , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/enzimología , Nifedipino/farmacología , Oxígeno/metabolismo , Arteria Pulmonar/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Serotonina/metabolismo , Intercambiador de Sodio-Calcio/antagonistas & inhibidores , Intercambiador de Sodio-Calcio/metabolismo , Tiourea/análogos & derivados , Tiourea/farmacología
10.
J Pharmacol Exp Ther ; 323(1): 381-90, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17640951

RESUMEN

Ryanodine is a selective ryanodine receptor (RyR) blocker, with binding dependent on RyR opening. In whole-cell studies, ryanodine binding can lock the RyR in an open-conductance state, short-circuiting the sarcoplasmic reticulum, which restricts studies of inositol-1,4,5-trisphosphate receptor (InsP3R) activity. Other RyR blockers have nonselective effects that also limit their utility. 4-(2-aminopropyl)-3,5-dichloro-N,N-dimethylaniline (FLA 365) blocks RyR-elicited Ca2+ increases in skeletal and cardiac muscle; yet, its actions on smooth muscle are unknown. Canine pulmonary arterial smooth muscle cells (PASMCs) express both RyRs and InsP3Rs; thus, we tested the ability of FLA 365 to block RyR- and serotonin-mediated InsP3R-elicited Ca2+ release by imaging fura-2-loaded PASMCs. Acute exposure to 10 mM caffeine, a selective RyR activator, induced Ca2+ increases that were reversibly reduced by FLA 365, with an estimated IC50 of approximately 1 to 1.5 microM, and inhibited by 10 microM ryanodine or 10 microM cyclopiazonic acid. FLA 365 also blocked L-type Ca2+ channel activity, with 10 microM reducing Ba2+ current amplitude in patch voltage-clamp studies to 54 +/- 6% of control and 100 microM FLA 365 reducing membrane current to 21 +/- 6%. InsP3R-mediated Ca2+ responses elicited by 10 microM 5-hydroxytryptamine (serotonin) in canine PASMCs and 100 microM carbachol in human embryonic kidney (HEK)-293 cells were not reduced by 2 microM FLA 365, but they were reduced by 20 microM FLA 365 to 76 +/- 9% of control in canine PASMCs and 52 +/- 1% in HEK-293 cells. Thus, FLA 365 preferentially blocks RyRs with limited inhibition of L-type Ca2+ channels or InsP3R in canine PASMCs.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Fenetilaminas/farmacología , Arteria Pulmonar/efectos de los fármacos , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Bario/metabolismo , Cafeína/farmacología , Señalización del Calcio/efectos de los fármacos , Línea Celular , Perros , Relación Dosis-Respuesta a Droga , Electrofisiología , Femenino , Indoles/farmacología , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/citología , Arteria Pulmonar/metabolismo , Rianodina/farmacología
11.
J Biol Chem ; 282(23): 16871-7, 2007 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-17442672

RESUMEN

Short ClC3 isoform (sClC3) functions as a volume-sensitive outwardly rectifying anion channel (VSOAC) in some cell types. In previous studies, we have shown that the hypotonic activation of sClC3 is linked to cell swelling-mediated remodeling of the actin cytoskeleton. In the present study, we have tested the hypothesis that the cytosolic tails of sClC3 bind to actin directly and that binding modulates the hypotonic activation of the channel. Co-sedimentation assays in vitro demonstrated a strong binding between the glutathione S-transferase-fused cytosolic C terminus of sClC3 (GST-sClC3-CT) to filamentous actin (F-actin) but not to globular monomeric actin (G-actin). The GST-fused N terminus (GST-sClC3-NT) exhibited low binding affinity to both G- and F-actin. Co-sedimentation experiments with progressively truncated GST-sClC3-CT indicated that the F-actin binding region is located between amino acids 690 and 760 of sClC3. Two synthetic peptides mapping basic clusters of the cytosolic sClC3-CT (CTP2, isoleucine 716 to leucine 734; and CTP3, proline 688 to proline 709) prevented binding of GST-sClC3-CT to F-actin in vitro. Dialysis into NIH/3T3 cells of these two peptides (but not of synthetic peptide CTP1 (isoleucine 737 to glutamine 748)) reduced the maximal current density by 60 and 38%, respectively. Based on these results, we have concluded that, by direct interaction with subcortical actin filaments, sClC3 contributes to the hypotonic stress-induced VSOACs in NIH/3T3 cells.


Asunto(s)
Actinas/metabolismo , Canales de Cloruro/metabolismo , Citosol/metabolismo , Animales , Western Blotting , Glutatión Transferasa/metabolismo , Humanos , Ratones , Células 3T3 NIH , Concentración Osmolar , Unión Proteica , Proteínas Recombinantes de Fusión/metabolismo
12.
Heart Rhythm ; 3(10): 1210-20, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17018354

RESUMEN

BACKGROUND: An osmotic challenge activates volume-regulated chloride currents (I(Cl,vol)), resulting in depolarization of the resting membrane potential and shortening of action potential duration (APD). I(Cl,vol) is activated in ischemia/reperfusion, but the effects of osmotic challenges and I(Cl,vol) on ventricular fibrillation (VF) are unknown. OBJECTIVES: The purpose of this study was to investigate the influence of hypo-osmotic and hypotonic stress and I(Cl,vol) activation on VF dynamics. METHODS: Guinea pig hearts were isolated, stained with di-4 ANEPPS to optically map action potentials (APs) from epicardium using a photodiode array, and perfused with iso-osmotic (low NaCl Ringer plus 45 mM mannitol) or hypo-osmotic (low NaCl Ringer) solution. RESULTS: Hypo-osmotic solution shortened APDs (143 +/- 5 ms --> 115 +/- 10 ms) and increased APD gradients between right and left ventricles (21 +/- 7 ms --> 41 +/- 10 ms, n = 4). In VF induced by burst stimulation, switching to hypo-osmotic solution increased VF frequencies (15.3 +/- 1.2 Hz to 28.9 +/- 3.6 Hz, n = 11), transforming complex fast Fourier transformation spectra to a single dominant high frequency on the left but not the right ventricle. Perfusion with the I(Cl,vol) blocker indanyloxyacetic acid-94 (10 muM) reversed organized VF to complex VF with lower (13.5 +/- 3.7 Hz in left ventricle) frequencies (n = 8), indicating that I(Cl,vol) underlies the changes in VF dynamics. Consistent with this interpretation, the levels of ClC-3 channel protein were 27% greater on left than right ventricles (n = 10), and computer simulations showed that insertion of I(Cl,vol) transformed complex VF to a stable spiral. CONCLUSION: Activation of I(Cl,vol) by decreasing osmolarity (45 mOsm) has a major impact on VF dynamics by transforming random multiple wavelets to a highly organized VF with a single dominant frequency.


Asunto(s)
Diuréticos Osmóticos/farmacología , Frecuencia Cardíaca/fisiología , Manitol/farmacología , Fibrilación Ventricular/tratamiento farmacológico , Animales , Simulación por Computador , Modelos Animales de Enfermedad , Cobayas , Frecuencia Cardíaca/efectos de los fármacos , Técnicas In Vitro , Soluciones Isotónicas/farmacología , Masculino , Concentración Osmolar , Perfusión , Solución de Ringer , Fibrilación Ventricular/metabolismo , Fibrilación Ventricular/fisiopatología , Función Ventricular/efectos de los fármacos
13.
J Physiol ; 568(Pt 2): 423-43, 2005 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16081489

RESUMEN

The K(v)4.2/4.3 channels are the primary subunits that contribute to the fast-inactivating, voltage-dependent transient outward K(+) current (I(to,fast)) in the heart. I(to,fast) is the critical determinant of the early repolarization of the cardiac action potential and plays an important role in the adaptive remodelling of cardiac myocytes, which usually causes cell volume changes, during myocardial ischaemia, hypertrophy and heart failure. It is not known, however, whether I(to,fast) is regulated by cell volume changes. In this study we investigated the molecular mechanism for cell volume regulation of I(to,fast) in native mouse left ventricular myocytes. Hyposmotic cell swelling caused a marked increase in densities of the peak I(to,fast) and a significant shortening in phase 1 repolarization of the action potential duration. The voltage-dependent gating properties of I(to,fast) were, however, not altered by changes in cell volume. In the presence of either protein kinase C (PKC) activator (12,13-dibutyrate) or phosphatase inhibitors (calyculin A and okadaic acid), hyposmotic cell swelling failed to further up-regulate I(to,fast). When expressed in NIH/3T3 cells, both K(v)4.2 and K(v)4.3 channels were also strongly regulated by cell volume in the same voltage-independent but PKC- and phosphatase-dependent manner as seen in I(to,fast) in the native cardiac myocytes. We conclude that K(v)4.2/4.3 channels in the heart are regulated by cell volume through a phosphorylation/dephosphorylation pathway mediated by PKC and serine/threonine phosphatase(s). These findings suggest a novel role of K(v)4.2/4.3 channels in the adaptive electrical and structural remodelling of cardiac myocytes in response to myocardial hypertrophy, ischaemia and reperfusion.


Asunto(s)
Tamaño de la Célula , Miocitos Cardíacos/metabolismo , Canales de Potasio Shal/metabolismo , Potenciales de Acción , Animales , Activación Enzimática , Ventrículos Cardíacos/metabolismo , Soluciones Hipotónicas , Activación del Canal Iónico/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/citología , Células 3T3 NIH , Presión Osmótica , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Canales de Potasio Shal/genética , Factores de Tiempo , Transfección
14.
J Biol Chem ; 280(27): 25871-80, 2005 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-15883157

RESUMEN

A novel volume-regulated hyperpolarization-activated chloride inward rectifier channel (Cl.ir) was identified in mammalian heart. To investigate whether ClC-2 is the gene encoding Cl.ir channels in heart, ClC-2 cDNAs cloned from rat (rClC-2) and guinea pig (gpClC-2) hearts were functionally characterized. When expressed in NIH/3T3 cells, full-length rClC-2 yielded inwardly rectifying whole-cell currents with very slow activation kinetics (time constants > 1.7 s) upon hyperpolarization under hypotonic condition. The single-channel rClC-2 currents had a unitary slope conductance of 3.9 +/- 0.2 picosiemens. A novel variant with an in-frame deletion at the beginning of exon 15 that leads to a deletion of 45 bp (corresponding to 15 amino acids in alpha-helices O and P, rClC-2(Delta509-523)) was identified in rat heart. The relative transcriptional expression levels of full-length rClC-2 and rClC-2(Delta509-523) in rat heart were 0.018 +/- 0.003 and 0.028 +/- 0.006 arbitrary units, respectively, relative to glyceraldehyde-3-phosphate dehydrogenase (n = 5, p = nonsignificant). A similar partial exon 15 skipping with a deletion of 105 bp (35 amino acids in alpha-helices O-Q, gpClC-2(Delta509-543)) was also identified in guinea pig heart. Expression of both rClC-2(Delta509-523) and gpClC-2(Delta509-543) resulted in functional channels with phenotypic activation kinetics and many properties identical to those of endogenous Cl.ir channels in native rat and guinea pig cardiac myocytes, respectively. Intracellular dialysis of anti-ClC-2 antibody inhibited expressed ClC-2 channels and endogenous Cl.ir currents in native rat and guinea pig cardiac myocytes. These results demonstrate that novel deletion variants of ClC-2 due to partial exon 15 skipping may be expressed normally in heart and contribute to the formation of endogenous Cl.ir channels in native cardiac cells.


Asunto(s)
Empalme Alternativo/fisiología , Canales de Cloruro/genética , Activación del Canal Iónico/fisiología , Miocitos Cardíacos/fisiología , Secuencia de Aminoácidos , Animales , Anticuerpos/farmacología , Canales de Cloruro CLC-2 , Canales de Cloruro/química , Canales de Cloruro/inmunología , Clonación Molecular , Cobayas , Ventrículos Cardíacos/citología , Masculino , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley
15.
Acta Pharmacol Sin ; 26(3): 265-78, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15715921

RESUMEN

In comparison to cation (K+, Na+, and Ca2+) channels, much less is currently known about the functional role of anion (Cl-) channels in cardiovascular physiology and pathophysiology. Over the past 15 years, various types of Cl- currents have been recorded in cardiac cells from different species including humans. All cardiac Cl- channels described to date may be encoded by five different Cl- channel genes: the PKA- and PKC-activated cystic fibrosis tansmembrane conductance regulator (CFTR), the volume-regulated ClC-2 and ClC-3, and the Ca2+-activated CLCA or Bestrophin. Recent studies using multiple approaches to examine the functional role of Cl- channels in the context of health and disease have demonstrated that Cl- channels might contribute to: 1) arrhythmogenesis in myocardial injury; 2) cardiac ischemic preconditioning; and 3) the adaptive remodeling of the heart during myocardial hypertrophy and heart failure. Therefore, anion channels represent very attractive novel targets for therapeutic approaches to the treatment of heart diseases. Recent evidence suggests that Cl- channels, like cation channels, might function as a multiprotein complex or functional module. In the post-genome era, the emergence of functional proteomics has necessitated a new paradigm shift to the structural and functional assessment of integrated Cl- channel multiprotein complexes in the heart, which could provide new insight into our understanding of the underlying mechanisms responsible for heart disease and protection.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Cardiomegalia/fisiopatología , Canales de Cloruro/fisiología , Precondicionamiento Isquémico , Animales , Arritmias Cardíacas/genética , Cardiomegalia/genética , Cardiomegalia/metabolismo , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Humanos
16.
Br J Pharmacol ; 145(1): 5-14, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15723096

RESUMEN

Cl- channels have been implicated in essential cellular functions including volume regulation, progression of cell cycle, cell proliferation and contraction, but the physiological functions of the ClC-3 channel are controversial. We tested the hypothesis that the ClC-3 gene (ClCn-3) is upregulated in hypertensive pulmonary arteries of monocrotaline-treated rats, and upregulated ClC-3 channel aids viability of pulmonary artery smooth muscle cells (PASMCs). Experimental pulmonary hypertension was induced in rats by a single subcutaneous administration of monocrotaline (60 mg kg(-1)). Injected animals developed characteristic features of pulmonary hypertension including medial hypertrophy of pulmonary arteries and right ventricular hypertrophy. Reverse transcriptase-polymerase chain reaction (RT-PCR), immunohistochemistry and Western immunoblot analysis indicated that histopathological alterations were associated with upregulation of the ClC-3 mRNA and protein expression in both smooth muscle cells of hypertensive pulmonary arteries and in cardiac myocytes. RT-PCR analysis of mRNA, extracted from canine cultured PASMCs, indicated that incubation with the inflammatory mediators endothelin-1 (ET-1), platelet-derived growth factor (PDGF), interleukin-1beta (IL-1beta) and tumor necrosis factor alpha (TNF alpha), but not transforming growth factor beta (TGFbeta), upregulated ClC-3 mRNA. Adenovirus-mediated delivery and overexpression of ClC-3 in canine PASMCs improved cell viability against increasing concentrations of hydrogen peroxide (H2O2, range 50-250 microM). In conclusion, upregulation of ClC-3 in rat hypertensive lung and heart is a novel observation. Our functional data suggest that upregulation of ClC-3 is an adaptive response of inflamed pulmonary artery, which enhances the viability of PASMCs against reactive oxygen species.


Asunto(s)
Canales de Cloruro/metabolismo , Músculo Liso Vascular/metabolismo , Arteria Pulmonar/metabolismo , Animales , Arteritis/metabolismo , Perros , Femenino , Hipertensión/metabolismo , Hipertrofia/metabolismo , Masculino , Músculo Liso Vascular/patología , Estrés Oxidativo , Arteria Pulmonar/patología , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
17.
Br J Pharmacol ; 144(2): 252-64, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15655514

RESUMEN

1. Measurements of artery contraction, cytosolic [Ca(2+)], and Ca(2+) permeability were made to examine contractile and cytosolic [Ca(2+)] responses of canine pulmonary arteries and isolated cells to 5-hydroxytryptamine (5-HT), and to determine the roles of intracellular Ca(2+) release and extracellular Ca(2+) entry in 5-HT responses. 2. The EC(50) for 5-HT-mediated contractions and cytosolic [Ca(2+)] increases was approximately 10(-7) M and responses were inhibited by ketanserin, a 5-HT(2A)-receptor antagonist. 3. 5-HT induced cytosolic [Ca(2+)] increases were blocked by 20 microM Xestospongin-C and by 2-APB (IC(50)=32 microM inhibitors of InsP(3) receptor activation. 4. 5-HT-mediated contractions were reliant on release of InsP(3) but not ryanodine-sensitive Ca(2+) stores. 5. 5-HT-mediated contractions and cytosolic [Ca(2+)] increases were partially inhibited by 10 microM nisoldipine, a voltage-dependent Ca(2+) channel blocker. 6. Extracellular Ca(2+) removal reduced 5-HT-mediated contractions further than nisoldipine and ablated cytosolic [Ca(2+)] increases and [Ca(2+)] oscillations. Similar to Ca(2+) removal, Ni(2+) reduced cytosolic [Ca(2+)] and [Ca(2+)] oscillations. 7. Mn(2+) quench of fura-2 and voltage-clamp experiments showed that 5-HT failed to activate any significant voltage-independent Ca(2+) entry pathways, including store-operated and receptor-activated nonselective cation channels. Ni(2+) but not nisoldipine or Gd(3+) blocked basal Mn(2+) entry. 8. Voltage-clamp experiments showed that simultaneous depletion of both InsP(3) and ryanodine-sensitive intracellular Ca(2+) stores activates a current with linear voltage dependence and a reversal potential consistent with it being a nonselective cation channel. 5-HT did not activate this current. 9. Basal Ca(2+) entry, rather than CCE, is important to maintain 5-HT-induced cytosolic [Ca(2+)] responses and contraction in canine pulmonary artery.


Asunto(s)
Calcio/metabolismo , Líquido Extracelular/metabolismo , Arteria Pulmonar/metabolismo , Serotonina/farmacología , Vasoconstricción/fisiología , Animales , Calcio/fisiología , Perros , Relación Dosis-Respuesta a Droga , Femenino , Técnicas In Vitro , Masculino , Arteria Pulmonar/efectos de los fármacos , Vasoconstricción/efectos de los fármacos
18.
J Physiol ; 563(Pt 2): 409-19, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15613369

RESUMEN

Capacitative Ca2+ entry (CCE) has been speculated to contribute to Ca2+ influx during hypoxic pulmonary vasoconstriction (HPV). The aim of the present study was to directly test if acute hypoxia causes intracellular Ca2+ concentration ([Ca2+]i) rises through CCE in canine pulmonary artery smooth muscle cells (PASMCs). In PASMCs loaded with fura-2, hypoxia produced a transient rise in [Ca2+]i in Ca2+-free solution, indicating Ca2+ release from the intracellular Ca2+ stores. Subsequent addition of 2 mm Ca2+ in hypoxia elicited a sustained rise in [Ca2+]i, which was partially inhibited by 10 microm nisoldipine. The dihydropyridine-insensitive rise in [Ca2+]i was due to increased Ca2+ influx, because it was abolished in Ca2+-free solution and hypoxia was shown to significantly enhance the rate of Mn2+ quench of fura-2 fluorescence. The dihyropyridine-insensitive rise in [Ca2+]i and the increased rate of Mn2+ quench of fura-2 fluorescence were inhibited by 50 microm SKF 96365 and 500 microm Ni2+, but not by 100 microm La3+ or 100 microm Gd3+, exhibiting pharmacological properties characteristic of CCE. In addition, predepletion of the intracellular Ca2+ stores inhibited the rise in [Ca2+]i induced by hypoxia. These results provide the first direct evidence that acute hypoxia, by causing Ca2+ release from the intracellular stores, activates CCE in isolated canine PASMCs, which may contribute to HPV.


Asunto(s)
Calcio/fisiología , Hipoxia de la Célula/fisiología , Músculo Liso Vascular/fisiología , Arteria Pulmonar/fisiología , Retículo Sarcoplasmático/fisiología , Animales , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Perros , Femenino , Técnicas In Vitro , Ionomicina/farmacología , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Nisoldipino/farmacología , Arteria Pulmonar/citología , Vasoconstricción/fisiología
19.
Am J Physiol Heart Circ Physiol ; 287(2): H533-44, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15277197

RESUMEN

The serum- and glucocorticoid-inducible kinase (SGK) is a serine/threonine protein kinase (PK) transcriptionally regulated by corticoids, serum, and cell volume. SGK regulates cell volume of various cells by effects on Na(+) and K(+) transport through membrane channels. We hypothesized a role for SGK in the activation of volume-sensitive osmolyte and anion channels (VSOACs) in cultured canine pulmonary artery smooth muscle cells (PASMCs). Intracellular dialysis through the patch electrode of recombinant active SGK, but not kinase-dead Delta60-SGK-K127M, heat-inactivated SGK, or active Akt1, partially activated VSOACs under isotonic conditions. Dialysis of active SGK before cell exposure to hypotonic medium significantly accelerated the activation kinetics and increased the maximal density of VSOAC current. Exposure of PASMCs to hypotonic medium (230 mosM) activated phosphatidylinositol 3-kinases (PI3Ks) and their downstream targets Akt/PKB and SGK but not PKC-epsilon. Inhibition of PI3Ks with wortmannin reduced the activation rate and maximal amplitude of VSOACs. Immunoprecipitated ClC-3 channels were phosphorylated by PKC-epsilon but not by SGK in vitro, suggesting that SGK may activate VSOACs indirectly. These data indicate that the PI3K-SGK cascade is activated on hypotonic swelling of PASMCs and, in turn, affects downstream signaling molecules linked to activation of VSOACs.


Asunto(s)
Canales de Cloruro/fisiología , Soluciones Hipotónicas/farmacología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Nucleares , Proteínas Serina-Treonina Quinasas/fisiología , Arteria Pulmonar/metabolismo , Animales , Células Cultivadas , Canales de Cloruro/metabolismo , Perros , Conductividad Eléctrica , Electrofisiología , Proteínas Inmediatas-Precoces , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteína Quinasa C/metabolismo , Proteína Quinasa C-epsilon , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-akt , Arteria Pulmonar/citología
20.
J Physiol ; 557(Pt 2): 439-56, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15020697

RESUMEN

ClC-3, a member of the large superfamily of ClC voltage-dependent Cl(-) channels, has been proposed as a molecular candidate responsible for volume-sensitive osmolyte and anion channels (VSOACs) in some cells, including heart and vascular smooth muscle. However, the reported presence of native VSOACs in at least two cell types from transgenic ClC-3 disrupted (Clcn3(-/-)) mice casts considerable doubt on this proposed role for ClC-3. We compared several properties of native VSOACs and examined mRNA transcripts and membrane protein expression profiles in cardiac and pulmonary arterial smooth muscle cells from Clcn3(+/+) and Clcn3(-/-) mice to: (1) test the hypothesis that native VSOACs are unaltered in cells from Clcn3(-/-) mice, and (2) test the possibility that targeted inactivation of the Clcn3 gene using a conventional murine global knock-out approach may result in compensatory changes in expression of other membrane proteins. Our experiments demonstrate that VSOAC currents in myocytes from Clcn3(+/+) and Clcn3(-/-) mice are remarkably similar in terms of activation and inactivation kinetics, steady-state current densities, rectification, anion selectivity (I(-) > Cl(-)>> Asp(-)) and sensitivity to block by glibenclamide, niflumic acid, DIDS and extracellular ATP. However, additional experiments revealed several significant differences in other fundamental properties of native VSOACs recorded from atrial and smooth muscle cells from Clcn3(-/-) mice, including: differences in regulation by endogenous protein kinase C, differential sensitivity to block by anti-ClC-3 antibodies, and differential sensitivities to [ATP](i) and free [Mg(2+)](i). These results suggest that in response to Clcn3 gene deletion, there may be compensatory changes in expression of other proteins that alter VSOAC channel subunit composition or associated regulatory subunits that give rise to VSOACs with different properties. Consistent with this hypothesis, in atria from Clcn3(-/-) mice compared to Clcn3(+/+) mice, quantitative analysis of ClC mRNA expression levels revealed significant increases in transcripts for ClC-1, ClC-2, and ClC-3, and protein expression profiles obtained using two-dimensional polyacrylamide gel electrophoresis revealed complex changes in at least 35 different unidentified membrane proteins in cells from Clcn3(-/-) mice. These findings emphasize that caution needs to be exercised in simple attempts to interpret the phenotypic consequences of conventional global Clcn3 gene inactivation.


Asunto(s)
Canales de Cloruro/fisiología , Canales Iónicos/fisiología , Proteínas de la Membrana/biosíntesis , Miocitos Cardíacos/metabolismo , Miocitos del Músculo Liso/metabolismo , Adenosina Trifosfato/farmacología , Animales , Anticuerpos/farmacología , Encéfalo/metabolismo , Canales de Cloruro/deficiencia , Canales de Cloruro/genética , Atrios Cardíacos/metabolismo , Canales Iónicos/química , Magnesio/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/química , Miocitos Cardíacos/inmunología , Miocitos del Músculo Liso/química , Miocitos del Músculo Liso/inmunología , Proteína Quinasa C/farmacología , Arteria Pulmonar/metabolismo , ARN Mensajero/análisis , ARN Mensajero/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA