Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Am J Physiol Lung Cell Mol Physiol ; 314(4): L670-L685, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29351439

RESUMEN

Application of H2S ("sulfide") elicits a complex contraction in rat pulmonary arteries (PAs) comprising a small transient contraction (phase 1; Ph1) followed by relaxation and then a second, larger, and more sustained contraction (phase 2; Ph2). We investigated the mechanisms causing this response using isometric myography in rat second-order PAs, with Na2S as a sulfide donor. Both phases of contraction to 1,000 µM Na2S were attenuated by the pan-PKC inhibitor Gö6983 (3 µM) and by 50 µM ryanodine; the Ca2+ channel blocker nifedipine (1 µM) was without effect. Ph2 was attenuated by the mitochondrial complex III blocker myxothiazol (1 µM), the NADPH oxidase (NOX) blocker VAS2870 (10 µM), and the antioxidant TEMPOL (3 mM) but was unaffected by the complex I blocker rotenone (1 µM). The bath sulfide concentration, measured using an amperometric sensor, decreased rapidly following Na2S application, and the peak of Ph2 occurred when this had fallen to ~50 µM. Sulfide caused a transient increase in NAD(P)H autofluorescence, the offset of which coincided with development of the Ph2 contraction. Sulfide also caused a brief mitochondrial hyperpolarization (assessed using tetramethylrhodamine ethyl ester), followed immediately by depolarization and then a second more prolonged hyperpolarization, the onset of which was temporally correlated with the Ph2 contraction. Sulfide application to cultured PA smooth muscle cells increased reactive oxygen species (ROS) production (recorded using L012); this was absent when the mitochondrial flavoprotein sulfide-quinone oxoreductase (SQR) was knocked down using small interfering RNA. We propose that the Ph2 contraction is largely caused by SQR-mediated sulfide metabolism, which, by donating electrons to ubiquinone, increases electron production by complex III and thereby ROS production.


Asunto(s)
Benzoquinonas/química , Sulfuro de Hidrógeno/farmacología , Músculo Liso Vascular/fisiología , Oxidorreductasas/metabolismo , Arteria Pulmonar/fisiología , Especies Reactivas de Oxígeno/metabolismo , Sulfuros/química , Animales , Calcio/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Músculo Liso Vascular/citología , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , Ratas , Ratas Wistar
2.
Proc Natl Acad Sci U S A ; 106(26): 10775-80, 2009 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-19541629

RESUMEN

Phenotypic modulation of airway smooth muscle (ASM) is an important feature of airway remodeling in asthma that is characterized by enhanced proliferation and secretion of pro-inflammatory chemokines. These activities are regulated by the concentration of free Ca(2+) in the cytosol ([Ca(2+)](i)). A rise in [Ca(2+)](i) is normalized by rapid reuptake of Ca(2+) into sarcoplasmic reticulum (SR) stores by the sarco/endoplasmic reticulum Ca(2+) (SERCA) pump. We examined whether increased proliferative and secretory responses of ASM from asthmatics result from reduced SERCA expression. ASM cells were cultured from subjects with and without asthma. SERCA expression was evaluated by western blot, immunohistochemistry and real-time PCR. Changes in [Ca(2+)](i), cell spreading, cellular proliferation, and eotaxin-1 release were measured. Compared with control cells from healthy subjects, SERCA2 mRNA and protein expression was reduced in ASM cells from subjects with moderately severe asthma. SERCA2 expression was similarly reduced in ASM in vivo in subjects with moderate/severe asthma. Rises in [Ca(2+)](i) following cell surface receptor-induced SR activation, or inhibition of SERCA-mediated Ca(2+) re-uptake, were attenuated in ASM cells from asthmatics. Likewise, the return to baseline of [Ca](i) after stimulation by bradykinin was delayed by approximately 50% in ASM cells from asthmatics. siRNA-mediated knockdown of SERCA2 in ASM from healthy subjects increased cell spreading, eotaxin-1 release and proliferation. Our findings implicate a deficiency in SERCA2 in ASM in asthma that contributes to its secretory and hyperproliferative phenotype in asthma, and which may play a key role in mechanisms of airway remodeling.


Asunto(s)
Asma/metabolismo , Bronquios/metabolismo , Retículo Sarcoplasmático/enzimología , Asma/patología , Asma/fisiopatología , Western Blotting , Bronquios/patología , Bronquios/fisiopatología , Calcio/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Quimiocina CCL11/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica , Homeostasis , Humanos , Inmunohistoquímica , Interleucina-13/farmacología , Masculino , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico
3.
Cardiovasc Res ; 77(3): 570-9, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18032393

RESUMEN

AIMS: We investigated the role of src family kinases (srcFK) in agonist-mediated Ca2+-sensitization in pulmonary artery and whether this involves interaction with the rho/rho-kinase pathway. METHODS AND RESULTS: Intra-pulmonary arteries (IPAs) and cultured pulmonary artery smooth muscle cells (PASMC) were obtained from rat. Expression of srcFK was determined at the mRNA and protein levels. Ca2+-sensitization was induced by prostaglandin F(2 alpha) (PGF(2 alpha)) in alpha-toxin-permeabilized IPAs. Phosphorylation of the regulatory subunit of myosin phosphatase (MYPT-1) and of myosin light-chain-20 (MLC20) and translocation of rho-kinase in response to PGF(2 alpha) were also determined. Nine srcFK were expressed at the mRNA level, including src, fyn, and yes, and PGF(2 alpha) enhanced phosphorylation of three srcFK proteins at tyr-416. In alpha-toxin-permeabilized IPAs, PGF(2 alpha) enhanced the Ca2+-induced contraction (pCa 6.9) approximately three-fold. This enhancement was inhibited by the srcFK blockers SU6656 and PP2 and by the rho-kinase inhibitor Y27632. Y27632, but not SU6656 or PP2, also inhibited the underlying pCa 6.9 contraction. PGF(2 alpha) enhanced phosphorylation of MYPT-1 at thr-697 and thr-855 and of MLC20 at ser-19. This enhancement, but not the underlying basal phosphorylation, was inhibited by SU6656. Y27632 suppressed both basal and PGF(2 alpha)-mediated phosphorylation. The effects of SU6656 and Y27632, on both contraction and MYPT-1 and MLC20 phosphorylation, were not additive. PGF(2 alpha) triggered translocation of rho-kinase in PASMC, and this was inhibited by SU6656. CONCLUSIONS: srcFK are activated by PGF(2 alpha) in the rat pulmonary artery and may contribute to Ca2+-sensitization and contraction via rho-kinase translocation and phosphorylation of MYPT-1.


Asunto(s)
Calcio/metabolismo , Arteria Pulmonar/metabolismo , Quinasas Asociadas a rho/fisiología , Familia-src Quinasas/fisiología , Animales , Células Cultivadas , Dinoprost/farmacología , Masculino , Cadenas Ligeras de Miosina/metabolismo , Fosforilación , Proteína Fosfatasa 1/metabolismo , Ratas , Ratas Wistar , Tirosina/metabolismo
4.
Free Radic Biol Med ; 45(10): 1468-76, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18805479

RESUMEN

Reactive oxygen species are implicated in pulmonary hypertension and hypoxic pulmonary vasoconstriction. We examined the effects of low concentrations of peroxide on intrapulmonary arteries (IPA). IPAs from Wistar rats were mounted on a myograph for recording tension and estimating intracellular Ca2+ using Fura-PE3. Ca2+ sensitization was examined in alpha-toxin-permeabilized IPAs, and phosphorylation of MYPT-1 and MLC(20) was assayed by Western blot. Peroxide (30 microM) induced a vasoconstriction with transient and sustained components and equivalent elevations of intracellular Ca2+. The transient constriction was strongly suppressed by indomethacin, the TP-receptor antagonist SQ-29584, and the Rho kinase inhibitor Y-27632, whereas sustained constriction was unaffected. Neither vasoconstriction nor elevation of intracellular Ca2+ was affected by removal of extracellular Ca2+, whereas dantrolene suppressed the former and ryanodine abolished the latter. Peroxide-induced constriction of permeabilized IPAs was unaffected by Y-27632 but abolished by PKC inhibitors; these also suppressed constriction in intact IPAs. Peroxide caused translocation of PKCalpha, but had no significant effect on MYPT-1 or MLC(20) phosphorylation. We conclude that in IPAs peroxide causes transient release of vasoconstrictor prostanoids, but sustained constriction is associated with release of Ca2+ from ryanodine-sensitive stores and a PKC-dependent but Rho kinase- and MLC(20)-independent constrictor mechanism.


Asunto(s)
Calcio/metabolismo , Peróxido de Hidrógeno/farmacología , Proteína Quinasa C/metabolismo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/fisiología , Vasoconstricción/efectos de los fármacos , Animales , Masculino , Ratas , Ratas Wistar , Factores de Tiempo
5.
JCI Insight ; 3(19)2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30282816

RESUMEN

Physiological and premature aging are frequently associated with an accumulation of prelamin A, a precursor of lamin A, in the nuclear envelope of various cell types. Here, we aimed to underpin the hitherto unknown mechanisms by which prelamin A alters myonuclear organization and muscle fiber function. By experimentally studying membrane-permeabilized myofibers from various transgenic mouse lines, our results indicate that, in the presence of prelamin A, the abundance of nuclei and myosin content is markedly reduced within muscle fibers. This leads to a concept by which the remaining myonuclei are very distant from each other and are pushed to function beyond their maximum cytoplasmic capacity, ultimately inducing muscle fiber weakness.


Asunto(s)
Envejecimiento Prematuro/fisiopatología , Núcleo Celular/metabolismo , Lamina Tipo A/metabolismo , Fibras Musculares Esqueléticas/fisiología , Fuerza Muscular , Envejecimiento Prematuro/genética , Animales , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Lamina Tipo A/genética , Ratones , Ratones Noqueados , Fibras Musculares Esqueléticas/citología , Miosinas/metabolismo , Cultivo Primario de Células
6.
Cardiovasc Res ; 65(2): 505-12, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15639490

RESUMEN

OBJECTIVE: To examine whether altered PCO2 or HCO3- at normal pH potentiate agonist-induced vasoconstriction of small pulmonary arteries, and if so to determine the mechanism. METHODS: Small intrapulmonary arteries (IPA) from rats were mounted on a myograph and PGF2alpha (3 microM)-induced tension recorded before and 40 min after replacing normal bath solution (5% CO2, 24 mM [HCO3-], pH 7.4) with one containing either normal [HCO3-] (24 mM) gassed with 10% CO2 (pH 7.12; hypercapnic acidosis) or high [HCO3-] (48 mM) gassed with 10% CO2 (pH 7.4; euhydric hypercapnia). RESULTS: Hypercapnic acidosis had no significant effect on the response of IPA to PGF2alpha. Euhydric hypercapnia however caused a substantial approximately 5.5-fold potentiation of the response (n=17, p<0.001) in the majority of preparations, whilst 20% of IPA (11 of 58) developed a slow spontaneous vasoconstriction after approximately 20 min. No equivalent responses to euhydric hypercapnia were observed in either mesenteric or renal arteries. Both the potentiation of PGF2alpha-induced vasoconstriction and the spontaneous vasoconstriction in IPA were inhibited by the L-type channel blocker diltiazem (10 microM). The potentiation was also suppressed by DIDS, an inhibitor of anion transporters, removal of extracellular Na+, and anthracene-9-carboxylic acid (A9C; 200 microM), reported to inhibit Ca2+-activated Cl- channels. Inhibition of nitric oxide synthase with L-NAME (100 microM) did not prevent potentiation. Depolarisation with 20 mM [K+] mimicked the effect of euhydric hypercapnia in that it also potentiated the response to PGF(2alpha) (>sixfold, n=6). CONCLUSIONS: Euhydric hypercapnia increases vasoreactivity of IPA, but not mesenteric or renal arteries, via a mechanism involving Na+-dependent HCO3- transport, activation of Ca2+-dependent Cl- channels, and subsequent depolarisation. These results may have consequences for patients with CO2-retaining chronic respiratory disease where plasma [HCO3-] is raised following renal compensation, and could explain the increased propensity to pulmonary hypertension and increased mortality in such patients.


Asunto(s)
Bicarbonatos/metabolismo , Hipercapnia/metabolismo , Músculo Liso Vascular/metabolismo , Circulación Pulmonar , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , Acetazolamida/farmacología , Animales , Antracenos/farmacología , Arterias , Transporte Biológico , Canales de Cloruro/efectos de los fármacos , Diltiazem/farmacología , Dinoprostona/farmacología , Concentración de Iones de Hidrógeno , Técnicas In Vitro , Canales Iónicos/efectos de los fármacos , Masculino , Arterias Mesentéricas/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Ratas , Ratas Wistar , Arteria Renal/efectos de los fármacos , Sodio/metabolismo , Vasoconstrictores/farmacología
7.
Cardiovasc Res ; 68(1): 56-64, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15950201

RESUMEN

OBJECTIVE: Sphingosylphosphorylcholine (SPC) is an important lipid mediator that has been implicated in vascular disease. As it has not been studied in the pulmonary circulation, we examined its mechanisms of action in rat small intrapulmonary arteries (IPA). METHODS: IPA were mounted on a myograph for recording tension and intracellular Ca2+ concentration ([Ca2+]i). Ca2+ sensitisation was examined in alpha-toxin permeabilized IPA, and by Western blot analysis of MYPT1 phosphorylation. RESULTS: SPC induced a slow but powerful vasoconstriction in IPA associated with an elevation in [Ca2+]i, with an EC50 for vasoconstriction of 12+/-2 microM. Removal of extracellular Ca2+ increased the EC50 to 76+/-33 microM (p<0.01) and abolished the rise in [Ca2+]i. Endothelial denudation or inhibition of NO synthase with L-NAME enhanced vasoconstriction. Treatment with pertussis toxin or the PLC inhibitor U731223 had no effect on SPC-induced vasoconstriction. The Rho kinase inhibitor Y27632 reduced SPC-induced vasoconstriction by approximately 70% and abolished both SPC-induced Ca2+ sensitisation in permeabilized IPA and the associated increase in MYPT1 phosphorylation; Ca2+ sensitisation was substantially inhibited by GDPbetaS. La3+ and 2-APB, at concentrations previously shown to block capacitative Ca2+ entry in IPA, suppressed SPC-induced vasoconstriction to the same extent as removal of extracellular Ca2+; residual tension was abolished by Y27632. Diltiazem was relatively ineffective. 2-APB also abolished the SPC-induced rise in [Ca2+]i. However, treatment with thapsigargin to empty intracellular stores had no effect on the elevation of [Ca2+]i induced by SPC. CONCLUSION: We present evidence that SPC is a powerful vasoconstrictor of IPA and the novel finding that SPC-induced vasoconstriction in IPA is dependent on activation of a Ca2+ entry pathway with a similar sensitivity to La3+ and 2-APB as capacitative Ca2+ entry, although its activation is not dependent on emptying of PLC/IP3 or thapsigargin-sensitive intracellular stores.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Músculo Liso Vascular/metabolismo , Fosforilcolina/análogos & derivados , Arteria Pulmonar/metabolismo , Esfingosina/análogos & derivados , Vasoconstricción , Amidas/farmacología , Animales , Western Blotting/métodos , Calcio/antagonistas & inhibidores , Bloqueadores de los Canales de Calcio/farmacología , Relación Dosis-Respuesta a Droga , Quinasa 1 del Receptor Acoplado a Proteína-G/antagonistas & inhibidores , Técnicas In Vitro , Lantano/farmacología , Lisofosfolípidos/farmacología , Músculo Liso Vascular/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Fosforilcolina/farmacología , Arteria Pulmonar/efectos de los fármacos , Piridinas/farmacología , Ratas , Esfingosina/farmacología , Tapsigargina/farmacología , Fosfolipasas de Tipo C/farmacología
8.
Pharmacol Ther ; 104(3): 207-31, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15556675

RESUMEN

Hypoxic pulmonary vasoconstriction (HPV) is an adaptive mechanism that in the normal animal diverts blood away from poorly ventilated areas of the lung, thereby maintaining optimal ventilation-perfusion matching. In global hypoxia however, such as in respiratory disease or at altitude, it causes detrimental increases in pulmonary vascular resistance and pulmonary artery (PA) pressure. The precise intracellular pathways and mechanisms underlying HPV remain unclear, although it is now recognised that both an elevation in smooth muscle intracellular [Ca2+] and a concomitant increase in Ca2+ sensitivity are involved. Several key intracellular protein kinases have been proposed as components of the signal transduction pathways leading to development of HPV, specifically Rho kinase, non-receptor tyrosine kinases (NRTK), p38 mitogen activated protein (MAP) kinase, and protein kinase C (PKC). All of these have been implicated to a greater or lesser extent in pathways leading to Ca2+ sensitisation, and in some cases regulation of intracellular [Ca2+] as well. In this article, we review the role of these key protein kinases in the regulation of vascular smooth muscle (VSM) constriction, applying what is known in the systemic circulation to the pulmonary circulation and HPV. We conclude that the strongest evidence for direct involvement of protein kinases in the mechanisms of HPV concerns a central role for Rho kinase in Ca2+ sensitisation, and a potential role for Src-family kinases in both modulation of Ca2+ entry via capacitative Ca2+ entry (CCE) and activation of Rho kinase, though others are likely to have indirect or modulatory influences. In addition, we speculate that Src family kinases may provide a central interface between the proposed hypoxia-induced generation of reactive oxygen species by mitochondria and both the elevation in intracellular [Ca2+] and Rho kinase mediated Ca2+ sensitisation.


Asunto(s)
Hipoxia/fisiopatología , Pulmón/irrigación sanguínea , Tono Muscular/fisiología , Músculo Liso Vascular/fisiología , Proteínas Quinasas/fisiología , Vasoconstricción/fisiología , Animales , Humanos , Hipoxia/metabolismo , Pulmón/metabolismo , Pulmón/fisiopatología , Proteínas Quinasas/clasificación , Reino Unido
9.
Cardiovasc Res ; 106(1): 121-30, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25661082

RESUMEN

AIMS: Sphingosylphosphorylcholine (SPC) elicits vasoconstriction at micromolar concentrations. At lower concentrations (≤1 µmol/L), however, it does not constrict intrapulmonary arteries (IPAs), but strongly potentiates vasoreactivity. Our aim was to determine whether this also occurs in a systemic artery and to delineate the signalling pathway. METHODS AND RESULTS: Rat mesenteric arteries and IPAs mounted on a myograph were challenged with ∼25 mmol/L [K+] to induce a small vasoconstriction. SPC (1 µmol/L) dramatically potentiated this constriction in all arteries by ∼400%. The potentiation was greatly suppressed or abolished by inhibition of phospholipase C (PLC; U73122), PKCε (inhibitory peptide), Src (PP2), and NADPH oxidase (VAS2870), and also by Tempol (superoxide scavenger), but not by inhibition of Rho kinase (Y27632). Potentiation was lost in mesenteric arteries from p47(phox-/-), but not NOX2(-/-), mice. The intracellular superoxide generator LY83583 mimicked the effect of SPC. SPC elevated reactive oxygen species (ROS) in vascular smooth muscle cells, and this was blocked by PP2, VAS2870, and siRNA knockdown of PKCε. SPC (1 µmol/L) significantly reduced the EC50 for U46619-induced vasoconstriction, an action ablated by Tempol. In patch-clamped mesenteric artery cells, SPC (200 nmol/L) enhanced Ba2+ current through L-type Ca2+ channels, an action abolished by Tempol but mimicked by LY83583. CONCLUSION: Our results suggest that low concentrations of SPC activate a PLC-coupled and NOX1-mediated increase in ROS, with consequent enhancement of voltage-gated Ca2+ entry and thus vasoreactivity. We speculate that this pathway is not specific for SPC, but may also contribute to vasoconstriction elicited by other G-protein coupled receptor and PLC-coupled agonists.


Asunto(s)
Canales de Calcio/efectos de los fármacos , Arterias Mesentéricas/fisiología , NADH NADPH Oxidorreductasas/fisiología , Fosforilcolina/análogos & derivados , Arteria Pulmonar/fisiología , Especies Reactivas de Oxígeno/metabolismo , Esfingosina/análogos & derivados , Vasoconstricción/efectos de los fármacos , Animales , Canales de Calcio/fisiología , Óxidos N-Cíclicos/farmacología , Relación Dosis-Respuesta a Droga , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Arterias Mesentéricas/efectos de los fármacos , Ratones , Ratones Noqueados , Modelos Animales , NADPH Oxidasa 1 , NADPH Oxidasa 2 , NADPH Oxidasas/deficiencia , NADPH Oxidasas/genética , NADPH Oxidasas/farmacología , NADPH Oxidasas/fisiología , Fosforilcolina/farmacología , Proteína Quinasa C-epsilon/farmacología , Arteria Pulmonar/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Esfingosina/farmacología , Marcadores de Spin , Fosfolipasas de Tipo C/farmacología , Vasoconstricción/fisiología
10.
Cell Calcium ; 36(3-4): 209-20, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15261477

RESUMEN

A key event in hypoxic pulmonary vasoconstriction (HPV) is the elevation in smooth muscle intracellular Ca2+ concentration. However, there is controversy concerning the source of this Ca2+, the signal transduction pathways involved, and the identity of the oxygen sensor. Although there is wide support for the hypothesis that hypoxia elicits depolarisation via inhibition of K+ channels, and thus promotes Ca2+ entry through L-type channels, a significant number of studies are inconsistent with this mechanism being either the sole or even major means by which Ca2+ is elevated during HPV. There is strong evidence that intracellular Ca2+ stores play a critical role, and voltage-independent Ca2+ entry mechanisms including capacitative Ca2+ entry (CCE) have also been implicated. There is renewed interest in the role of mitochondria in HPV, both in terms of modulators of Ca2+ homeostasis per se and as oxygen sensors. There is however considerable uncertainty concerning the mechanisms involved in the latter, with proposals for changes in redox couples and both an increase and decrease in mitochondrial production of reactive oxygen species (ROS). In this article we review the evidence for and against involvement of such mechanisms in HPV, and propose a model for the regulation of intracellular [Ca2+] in pulmonary artery during hypoxia in which the mitochondria play a central role.


Asunto(s)
Calcio/fisiología , Mitocondrias/fisiología , Oxígeno/fisiología , Circulación Pulmonar/fisiología , Animales , Humanos , Hipoxia/metabolismo , Hipoxia/fisiopatología
11.
Br J Pharmacol ; 140(1): 97-106, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12967939

RESUMEN

(1) The effect of induction of capacitative Ca2+ entry (CCE) upon tone in small (i.d. 200-500 microm) intrapulmonary (IPA), mesenteric (MA), renal (RA), femoral (FA), and coronary arteries (CA) of the rat was examined. (2) Following incubation of IPA with 100 nm thapsigargin (Thg) in Ca2+-free physiological salt solution (PSS), a sustained contraction was observed upon reintroduction of 1.8 mm Ca2+, which was unaffected by either diltiazem (10 microm) or the reverse mode Na+/Ca2+ antiport inhibitor KB-R7943 (10 microm). An identical protocol failed to elicit contraction in MA, RA, or CA, while a small transient contraction was sometimes observed in FA. (3) The effect of this protocol on the intracellular Ca2+ concentration ([Ca2+]i) was assessed using Fura PE3-loaded IPA, MA, and FA. Reintroduction of Ca2+ into the bath solution following Thg treatment in Ca2+-free PSS caused a large, rapid, and sustained increase in [Ca2+]i in all the three types of artery. (4) 100 nm Thg induced a slowly developing noisy inward current in smooth muscle cells (SMC) isolated from IPA, which was due to an increase in the activity of single channels with a conductance of approximately 30 pS. The current had a reversal potential near 0 mV in normal PSS, and persisted when Ca2+-dependent K+ and Cl- currents were blocked; it was greatly inhibited by 1 microm La3+, 1 microm Gd3+, and the IP3 receptor antagonist 2-APB (75 microm), and by replacement of extracellular cations by NMDG+. (5) In conclusion, depletion of intracellular Ca2+ stores with Thg caused capacitative Ca2+ entry in rat small muscular IPA, MA, and FA. However, a corresponding contraction was observed only in IPA. CCE in IPA was associated with the development of a small La3+- and Gd3+-sensitive current, and an increased Mn2+ quench of Fura PE-3 fluorescence. These results suggest that although CCE occurs in a number of types of small arteries, its coupling to contraction appears to be of particular importance in pulmonary arteries.


Asunto(s)
Calcio/metabolismo , Músculo Liso Vascular/metabolismo , Arteria Pulmonar/metabolismo , Vasoconstricción/fisiología , Animales , Calcio/farmacología , Capacidad Eléctrica , Técnicas In Vitro , Masculino , Músculo Liso Vascular/efectos de los fármacos , Arteria Pulmonar/efectos de los fármacos , Ratas , Ratas Wistar , Vasoconstricción/efectos de los fármacos
12.
Respir Physiol Neurobiol ; 132(1): 55-67, 2002 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-12126695

RESUMEN

Vasomotor responses to hypoxia constitute a fundamental adaptation to a commonly encountered stress. It has long been suspected that changes in cellular energetics may modulate both hypoxic systemic artery vasodilatation (HSV) and hypoxic pulmonary artery vasoconstriction (HPV). Although limitation of energy has been shown to underlie hypoxic relaxation in some smooth muscles, the response to hypoxia in vascular smooth muscle does not appear to be a simple function of energy stores, but instead may involve perturbations of ATP or energy delivery to mechanisms controlling muscle force, and/or changes associated with anaerobic metabolism. Recent work in pulmonary vascular smooth muscle has demonstrated that energy stores are maintained during hypoxic pulmonary vasoconstriction, and that this is dependent on glucose availability and up-regulation of glycolysis. There is increasing evidence that glycolysis is preferentially coupled to a variety of membrane associated ATP dependent processes, including the Na(+) pump, Ca(2+)-ATPase, and possibly some protein kinases. These and other mechanisms may influence excitation-contraction coupling in both systemic and pulmonary arteries by effects on intracellular Ca(2+) and/or Ca(2+) sensitivity. Hypoxia has also been postulated to have major effects on other cytosolic second messenger systems including phosphatidylinositol pathways, cell redox state and mitochondrial reactive oxygen species production. This review examines the relationship between energy state, anaerobic respiration and hypoxic vasomotor tone, with a particular emphasis on hypoxic pulmonary vasoconstriction.


Asunto(s)
Metabolismo Energético/fisiología , Hipoxia/metabolismo , Músculo Liso Vascular/metabolismo , Circulación Pulmonar/fisiología , Animales , Tono Muscular/fisiología
13.
Cardiovasc Res ; 99(3): 404-11, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23708740

RESUMEN

AIMS: To determine the role of gap junctions (GJs) in hypoxic pulmonary vasoconstriction (HPV). METHODS AND RESULTS: Studies were performed in rat isolated intrapulmonary arteries (IPAs) mounted on a myograph and in anaesthetized rats. Hypoxia induced a biphasic HPV response in IPAs preconstricted with prostaglandin F2α (PGF2α, 3 µM) or 20 mM K⁺. The GJ inhibitors 18ß-glycyrrhetinic acid (18ß-GA, 30 µM), heptanol (3.5 mM), or 2-aminoethoxydiphenyl borate (2-APB) (75 µM) had little effect on the transient Phase 1 of HPV, but abolished the sustained Phase 2 which is associated with Ca²âº sensitization. The voltage-dependent Ca²âº channel blocker diltiazem (10 µM) had no effect on HPV, and did not alter the inhibitory action of 18ß-GA. Sustained HPV is enhanced by high glucose (15 mM) via potentiation of Ca²âº sensitization, in the presence of high glucose 18ß-GA still abolished sustained HPV. Simultaneous measurement of tension and intracellular Ca²âº using Fura PE-3 demonstrated that whilst 18ß-GA abolished tension development during sustained HPV, it did not affect the elevation of intracellular Ca²âº. Consistent with this, 18ß-GA abolished hypoxia-induced phosphorylation of the Rho kinase target MYPT-1. In anaesthetized rats hypoxia caused a biphasic increase in systolic right ventricular pressure. Treatment with oral 18ß-GA (25 mg/kg) abolished the sustained component of the hypoxic pressor response. CONCLUSION: These results imply that GJs are critically involved in the signalling pathways leading to Rho kinase-dependent Ca²âº sensitization during sustained HPV, but not elevation of intracellular Ca²âº, and may explain the dependence of the former on an intact endothelium.


Asunto(s)
Calcio/metabolismo , Uniones Comunicantes/metabolismo , Hipoxia/fisiopatología , Arteria Pulmonar/fisiopatología , Vasoconstricción/fisiología , Animales , Presión Sanguínea/efectos de los fármacos , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/metabolismo , Diltiazem/farmacología , Uniones Comunicantes/efectos de los fármacos , Ácido Glicirretínico/análogos & derivados , Ácido Glicirretínico/farmacología , Hipoxia/metabolismo , Masculino , Fosforilación , Proteína Fosfatasa 1/metabolismo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo
14.
Cardiovasc Res ; 89(1): 214-24, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20805095

RESUMEN

AIMS: the aim of this study was to determine the relative importance of Ca(2+) sensitization, ion channels, and intracellular Ca(2+) ([Ca(2+)](i)) in the mixed constrictor/relaxation actions of superoxide anion on systemic and pulmonary arteries. METHODS AND RESULTS: pulmonary and mesenteric arteries were obtained from rat. Superoxide was generated in arteries and cells with 6-anilino-5,8-quinolinequinone (LY83583). Following pre-constriction with U46619, 10 µmol/L LY83583 caused constriction in pulmonary and relaxation in mesenteric arteries. Both constrictor and relaxant actions of LY83583 were inhibited by superoxide dismutase and catalase. LY83583 caused Rho-kinase-dependent constriction in α-toxin-permeabilized pulmonary but not mesenteric arteries. Phosphorylation of myosin phosphatase-targeting subunit-1 (MYPT-1; as determined by western blot), was enhanced by LY83583 in pulmonary artery only. However, in both artery types, changes in tension were closely correlated with changes in phosphorylation of the 20 kDa myosin light chain as well as changes in [Ca(2+)](i) (as measured with Fura PE-3), with LY83583 causing increases in pulmonary and decreases in mesenteric arteries. When U46619 was replaced by 30 mmol/L K(+), all changes in [Ca(2+)](i) were abolished and LY83583 constricted both artery types. The K(V) channel inhibitor 4-aminopyridine abolished the LY83583-induced relaxation in mesenteric artery without affecting constriction in pulmonary artery. However, LY83583 caused a similar hyperpolarizing shift in the steady-state activation of K(V) current in isolated smooth muscle cells of both artery types. CONCLUSIONS: superoxide only causes Rho-kinase-dependent Ca(2+) sensitization in pulmonary artery, resulting in constriction, and whilst it opens K(V) channels in both artery types, this only results in relaxation in mesenteric.


Asunto(s)
Arterias Mesentéricas/fisiología , Arteria Pulmonar/fisiología , Superóxidos/metabolismo , Vasoconstricción/fisiología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , 4-Aminopiridina/farmacología , Aminoquinolinas/farmacología , Animales , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Guanilato Ciclasa/antagonistas & inhibidores , Técnicas In Vitro , Masculino , Arterias Mesentéricas/citología , Arterias Mesentéricas/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Oxadiazoles/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Canales de Potasio con Entrada de Voltaje/fisiología , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , Quinoxalinas/farmacología , Ratas , Ratas Wistar , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Quinasas Asociadas a rho/metabolismo
16.
Free Radic Biol Med ; 46(5): 633-42, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19103285

RESUMEN

Reactive oxygen species play a key role in vascular disease, pulmonary hypertension, and hypoxic pulmonary vasoconstriction. We investigated contractile responses, intracellular Ca(2+) ([Ca(2+)](i)), Rho-kinase translocation, and phosphorylation of the regulatory subunit of myosin phosphatase (MYPT-1) and of myosin light chain (MLC(20)) in response to LY83583, a generator of superoxide anion, in small intrapulmonary arteries (IPA) of rat. LY83583 caused concentration-dependent constrictions in IPA and greatly enhanced submaximal PGF(2alpha)-mediated preconstriction. In small femoral or mesenteric arteries of rat, LY83583 alone was without effect, but it relaxed a PGF(2)alpha-mediated preconstriction. Constrictions in IPA were inhibited by superoxide dismutase and tempol, but not catalase, and were endothelium and guanylate cyclase independent. Constrictions were also inhibited by the Rho-kinase inhibitor Y27632 and the Src-family kinase inhibitor SU6656. LY83583 did not raise [Ca(2+)](i), but caused a Y27632-sensitive constriction in alpha-toxin-permeabilized IPA. LY83583 triggered translocation of Rho-kinase from the nucleus to the cytosol in pulmonary artery smooth muscle cells and enhanced phosphorylation of MYPT-1 at Thr-855 and of MLC(20) at Ser-19 in IPA. This enhancement was inhibited by superoxide dismutase and abolished by Y27632. Hydrogen peroxide did not activate Rho-kinase. We conclude that in rat small pulmonary artery, superoxide triggers Rho-kinase-mediated Ca(2+) sensitization and vasoconstriction independent of hydrogen peroxide.


Asunto(s)
Broncoconstricción/fisiología , Hipertensión Pulmonar/enzimología , Miocitos del Músculo Liso/fisiología , Superóxidos/farmacología , Quinasas Asociadas a rho/fisiología , Familia-src Quinasas/fisiología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Amidas/farmacología , Aminoquinolinas/farmacología , Animales , Broncoconstricción/efectos de los fármacos , Señalización del Calcio/fisiología , Guanilato Ciclasa/antagonistas & inhibidores , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/prevención & control , Indoles/farmacología , Masculino , Miocitos del Músculo Liso/patología , Cadenas Ligeras de Miosina/metabolismo , Fosforilación , Proteína Fosfatasa 1/metabolismo , Arteria Pulmonar/patología , Piridinas/farmacología , Ratas , Ratas Wistar , Sulfonamidas/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Familia-src Quinasas/antagonistas & inhibidores
17.
Cardiovasc Res ; 80(3): 453-62, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18682436

RESUMEN

AIMS: We investigated the role of src-family kinases (srcFKs) in hypoxic pulmonary vasoconstriction (HPV) and how this relates to Rho-kinase-mediated Ca(2+) sensitization and changes in intracellular Ca(2+) concentration ([Ca(2+)](i)). METHODS AND RESULTS: Intra-pulmonary arteries (IPAs) were obtained from male Wistar rats. HPV was induced in myograph-mounted IPAs. Auto-phosphorylation of srcFKs and phosphorylation of the regulatory subunit of myosin phosphatase (MYPT-1) and myosin light-chain (MLC(20)) in response to hypoxia were determined by western blotting. Translocation of Rho-kinase and effects of siRNA knockdown of src and fyn were examined in cultured pulmonary artery smooth muscle cells (PASMCs). [Ca(2+)](i) was estimated in Fura-PE3-loaded IPA. HPV was inhibited by two blockers of srcFKs, SU6656 and PP2. Hypoxia enhanced phosphorylation of three srcFK proteins at Tyr-416 (60, 59, and 54 kDa, corresponding to src, fyn, and yes, respectively) and enhanced srcFK-dependent tyrosine phosphorylation of multiple target proteins. Hypoxia caused a complex, time-dependent enhancement of MYPT-1 and MLC(20) phosphorylation, both in the absence and presence of pre-constriction. The sustained component of this enhancement was blocked by SU6656 and the Rho-kinase inhibitor Y27632. In PASMCs, hypoxia caused translocation of Rho-kinase from the nucleus to the cytoplasm, and this was prevented by anti-src siRNA and to a lesser extent by anti-fyn siRNA. The biphasic increases in [Ca(2+)](i) that accompany HPV were also inhibited by PP2. CONCLUSION: Hypoxia activates srcFKs and triggers protein tyrosine phosphorylation in IPA. Hypoxia-mediated Rho-kinase activation, Ca(2+) sensitization, and [Ca(2+)](i) responses are depressed by srcFK inhibitors and/or siRNA knockdown, suggesting a central role of srcFKs in HPV.


Asunto(s)
Hipoxia/metabolismo , Arteria Pulmonar/metabolismo , Vasoconstricción/fisiología , Familia-src Quinasas/metabolismo , Amidas/farmacología , Animales , Calcio/metabolismo , Inhibidores Enzimáticos/farmacología , Indoles/farmacología , Masculino , Modelos Animales , Cadenas Ligeras de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosforilación , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , Piridinas/farmacología , Ratas , Ratas Wistar , Sulfonamidas/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo , Familia-src Quinasas/antagonistas & inhibidores
18.
Hypertension ; 51(2): 239-45, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18158336

RESUMEN

Sphingosylphosphorylcholine (SPC) is a powerful vasoconstrictor, but in vitro its EC(50) is approximately 100-fold more than plasma concentrations. We examined whether subcontractile concentrations of SPC (100 nmol/L of SPC, and independent of the endothelium, 2-aminoethoxydiphenylborane-sensitive Ca(2+) entry, and Rho kinase. It was abolished by the phospholipase C inhibitor U73122, the broad spectrum protein kinase C (PKC) inhibitor Ro31-8220, and the PKC delta inhibitor rottlerin, but not by Gö6976, which is ineffective against PKC delta. The potentiation could be attributed to enhancement of Ca(2+) entry. SPC also potentiated the responses to prostaglandin F(2 alpha) and U436619, which activate a 2-aminoethoxydiphenylborane sensitive nonselective cation channel in intrapulmonary arteries. In this case, potentiation was partially inhibited by diltiazem but abolished by 2-aminoethoxydiphenylborane, Ro31-8220, and rottlerin. SPC (1 micromol/L) caused translocation of PKC delta to the perinuclear region and cytoskeleton of cultured intrapulmonary artery smooth muscle cells. We present the novel finding that low, subcontractile concentrations of SPC potentiate Ca(2+) entry in intrapulmonary arteries through both voltage-dependent and independent pathways via a receptor-dependent mechanism involving PKC delta. This has implications for the physiological role of SPC, especially in cardiovascular disease, where SPC is reported to be elevated.


Asunto(s)
Calcio/metabolismo , Fosforilcolina/análogos & derivados , Proteína Quinasa C-delta/fisiología , Arteria Pulmonar/fisiología , Esfingosina/análogos & derivados , Sistema Vasomotor/efectos de los fármacos , Animales , Transporte Biológico/efectos de los fármacos , Bloqueadores de los Canales de Calcio/farmacología , Células Cultivadas , Diltiazem/farmacología , Dinoprost/farmacología , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Electrofisiología , Inhibidores Enzimáticos/farmacología , Membranas Intracelulares/metabolismo , Masculino , Miocitos del Músculo Liso/enzimología , Concentración Osmolar , Fosforilcolina/administración & dosificación , Fosforilcolina/farmacología , Potasio/farmacología , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/metabolismo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/enzimología , Arteria Pulmonar/metabolismo , Ratas , Ratas Wistar , Esfingosina/administración & dosificación , Esfingosina/farmacología , Vasoconstricción/efectos de los fármacos
19.
Am J Physiol Lung Cell Mol Physiol ; 293(4): L982-90, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17616643

RESUMEN

It has been proposed that a hypoxia-induced inhibition of the Na(+)-Ca(2+) exchanger (NCX) contributes to hypoxic pulmonary vasoconstriction (HPV). By recording isometric tension development in rat intrapulmonary arteries (IPA), we examined the effect on HPV of maneuvers that reduce the ability of NCX to regulate intracellular Ca(2+) concentration ([Ca(2+)](i)). In some experiments, fura pentakis(acetoxymethyl) ester-3 (fura PE-3) was also used to monitor [Ca(2+)](i). HPV was elicited in IPA that were pretreated with 10 microM diltiazem and slightly preconstricted with PGF(2alpha), which enhances the hypoxic response. Substitution of Na(+) with Li(+) increased HPV and the associated rise in [Ca(2+)](i). Pretreatment with ouabain (100 microM) to diminish the Na(+) gradient or with the reverse-mode NCX inhibitor KB-R7943 (3 or 10 microM) had no significant effect on HPV. Combined treatment with ouabain and low-[Na(+)] (24 mM) solution enhanced HPV strongly. The role of NCX in Ca(2+) extrusion was examined by assessing the decrease in [Ca(2+)](i) in Ca(2+)-free physiological saline solution either containing or lacking Na(+) following a high K(+)-induced loading of cellular [Ca(2+)]. Although the large initial rapid fall in [Ca(2+)] was Na(+) independent, final recovery of [Ca(2+)] to its basal level was delayed in the absence of Na(+). Therefore, HPV persisted or was increased under conditions in which forward-mode NCX was already attenuated or prevented, demonstrating that inhibition of NCX by hypoxia is unlikely to initiate HPV. Instead, NCX appears to act to inhibit HPV as would be expected if it is functioning to extrude Ca(2+).


Asunto(s)
Hipoxia/fisiopatología , Arteria Pulmonar/fisiopatología , Intercambiador de Sodio-Calcio/metabolismo , Vasoconstricción , Animales , Calcio/metabolismo , Membranas Intracelulares/metabolismo , Contracción Isométrica , Litio/farmacología , Masculino , Concentración Osmolar , Potasio/farmacología , Arteria Pulmonar/metabolismo , Ratas , Ratas Wistar , Sodio/farmacología , Vasoconstricción/efectos de los fármacos
20.
J Physiol ; 571(Pt 1): 147-63, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16357015

RESUMEN

The mechanisms by which prostaglandin F(2alpha) (PGF(2alpha)) increases intracellular Ca2+ concentration [Ca2+]i in vascular smooth muscle remain unclear. We examined the role of store-, receptor- and voltage-operated Ca2+ influx pathways in rat intrapulmonary arteries (IPA) loaded with Fura PE-3. Low concentrations (0.01-1 microM) of PGF(2alpha) caused a transient followed by a plateau rise in [Ca2+]i. Both responses became maximal at 0.1 microM PGF(2alpha). At higher concentrations of PGF(2alpha), a further slower rise in [Ca2+]i was superimposed on the plateau. The [Ca2+]i response to 0.1 microM PGF(2alpha) was mimicked by the FP receptor agonist fluprostenol, whilst the effect of 10 microM PGF(2alpha) was mimicked by the TP receptor agonist U-46619. The plateau rise in [Ca2+]i in response to 0.1 microM PGF(2alpha) was insensitive to diltiazem, and was abolished in Ca2+-free physiological salt solution, and by pretreatment with La3+, 2-APB, thapsigargin or U-73122. The rises in [Ca2+]i in response to 10 microM PGF(2alpha) and 0.01 microM U-46619 were partially inhibited by diltiazem. The diltiazem-resistant components of both of these responses were inhibited by 2-APB and La3+ to an extent which was significantly less than that seen for the response to 0.1 microM PGF(2alpha), and were also much less sensitive to U-73122. The U-46619 response was also relatively insensitive to thapsigargin. When Ca2+ was replaced with Sr2+, the sustained increase in the Fura PE-3 signal to 0.1 microM PGF(2alpha) was abolished, whereas 10 microM PGF(2alpha) and 0.05 microM U-46619 still caused substantial increases. These results suggest that low concentrations of PGF(2alpha) act via FP receptors to cause IP3-dependent Ca2+ release and store operated Ca2+ entry (SOCE). U-46619 and 10-100 microM PGF(2alpha) cause a TP receptor-mediated Ca2+ influx involving both L-type Ca2+ channels and a receptor operated pathway, which differs from SOCE in its susceptibility to La3+, 2-APB and thapsigargin, does not require phospholipase C activation, and is Sr2+ permeable.


Asunto(s)
Calcio/análisis , Calcio/metabolismo , Dinoprost/farmacología , Arteria Pulmonar/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Animales , Compuestos de Boro/farmacología , Canales de Calcio Tipo L/fisiología , Fármacos Cardiovasculares/farmacología , Diltiazem/farmacología , Inositol 1,4,5-Trifosfato/fisiología , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Arteria Pulmonar/química , Ratas , Ratas Wistar , Receptores de Prostaglandina/efectos de los fármacos , Receptores de Prostaglandina/fisiología , Receptores de Tromboxanos/efectos de los fármacos , Receptores de Tromboxanos/fisiología , Transducción de Señal/fisiología , Fosfolipasas de Tipo C/farmacología , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA