Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochim Biophys Acta ; 1862(4): 611-621, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26775030

RESUMEN

We identified murine miR-322, orthologous to human miR-424, as a new regulator of insulin receptor, IGF-1 receptor and sirtuin 4 mRNA in vitro and in vivo in the heart and found that miR-322/424 is highly expressed in the heart of mice. C57Bl/6N mice fed 10weeks of high fat diet (HFD) presented signs of cardiomyopathy and a stable miR-322 cardiac level while cardiac function was slightly affected in 11week-old ob/ob which overexpressed miR-322. We thus hypothesized that mmu-miR-322 could be protective against cardiac consequences of hyperinsulinemia and hyperlipidemia. We overexpressed or knocked-down mmu-miR-322 using AAV9 and monitored cardiac function in wild-type C57Bl/6N mice fed a control diet (CD) or a HFD and in ob/ob mice. The fractional shortening progressively declined while the left ventricle systolic diameter increased in HFD mice infected with an AAVcontrol or with an AAVsponge (decreasing miR-322 bioavailability) but also in ob/ob mice infected with AAVsponge. Similar observations were also found in CD-fed mice infected with AAVsponge. On the contrary over-expressing miR-322 with AAVmiR-322 was efficient in protecting the heart from HFD effects in C57Bl/6N mice. This cardioprotection could be associated with the regulation of identified targets IGF1R, INSR and CD1, a decrease in insulin signaling pathway and an enrichment of genes involved in mitochondrial function and fatty acid oxidation as demonstrated by transcriptome analysis. Altogether, these results emphasize miR-322 as a new potential therapeutic target against cardiac consequences of metabolic syndrome, which represents an increasing burden in the western countries.


Asunto(s)
Cardiopatías/metabolismo , Insulina/metabolismo , Síndrome Metabólico/metabolismo , MicroARNs/biosíntesis , Transducción de Señal , Animales , Dependovirus , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/farmacología , Vectores Genéticos , Cardiopatías/genética , Cardiopatías/patología , Cardiopatías/terapia , Humanos , Hiperinsulinismo/genética , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patología , Hiperinsulinismo/terapia , Hiperlipidemias/genética , Hiperlipidemias/metabolismo , Hiperlipidemias/patología , Hiperlipidemias/terapia , Insulina/genética , Masculino , Síndrome Metabólico/genética , Síndrome Metabólico/patología , Síndrome Metabólico/terapia , Ratones , Ratones Obesos , MicroARNs/genética , Ratas , Ratas Wistar , Transducción Genética
2.
Blood ; 126(15): 1823-30, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26316625

RESUMEN

Molecules that reduce the level of cyclic adenosine 5'-monophosphate (cAMP) in the platelet cytosol, such as adenosine 5'-diphosphate (ADP) secreted from dense granules, trigger platelet activation. Therefore, any change in the distribution and/or availability of cyclic nucleotides or ADP may interfere with platelet reactivity. In this study, we evaluated the role of multidrug resistance protein 4 (MRP4, or ABCC4), a nucleotide transporter, in platelet functions in vivo and in vitro by investigating MRP4-deficient mice. MRP4 deletion resulted in a slight increase in platelet count but had no impact on platelet ultrastructure. In MRP4-deficient mice, the arterial occlusion was delayed and the tail bleeding time was prolonged. In a model of platelet depletion and transfusion mimicking a platelet-specific knockout, mice injected with MRP4(-/-) platelets also showed a significant increase in blood loss compared with mice injected with wild-type platelets. Defective thrombus formation and platelet activation were confirmed in vitro by studying platelet adhesion to collagen in flow conditions, integrin αIIbß3 activation, washed platelet secretion, and aggregation induced by low concentrations of proteinase-activated receptor 4-activating peptide, U46619, or ADP. We found no role of MRP4 in ADP dense-granule storage, but MRP4 redistributed cAMP from the cytosol to dense granules, as confirmed by increased vasodilator-stimulated phosphoprotein phosphorylation in MRP4-deficient platelets. These data suggest that MRP4 promotes platelet aggregation by modulating the cAMP-protein kinase A signaling pathway, suggesting that MRP4 might serve as a target for novel antiplatelet agents.


Asunto(s)
Plaquetas/patología , AMP Cíclico/metabolismo , Homeostasis/fisiología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/fisiología , Activación Plaquetaria , Trombosis/patología , Animales , Transporte Biológico , Plaquetas/metabolismo , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ratones , Ratones Noqueados , Fosforilación , Agregación Plaquetaria , Pruebas de Función Plaquetaria , Transducción de Señal , Trombosis/metabolismo
3.
Circulation ; 129(7): 773-85, 2014 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-24249716

RESUMEN

BACKGROUND: Phenotypic modulation or switching of vascular smooth muscle cells from a contractile/quiescent to a proliferative/synthetic phenotype plays a key role in vascular proliferative disorders such as atherosclerosis and restenosis. Although several calcium handling proteins that control differentiation of smooth muscle cells have been identified, the role of protein phosphatase inhibitor 1 (I-1) in the acquisition or maintenance of the contractile phenotype modulation remains unknown. METHODS AND RESULTS: In human coronary arteries, I-1 and sarco/endoplasmic reticulum Ca2+ -ATPase expression is specific to contractile vascular smooth muscle cells. In synthetic cultured human coronary artery smooth muscle cells, protein phosphatase inhibitor 1 (I-1 target) is highly expressed, leading to a decrease in phospholamban phosphorylation, sarco/endoplasmic reticulum Ca2+ -ATPase, and cAMP-responsive element binding activity. I-1 knockout mice lack phospholamban phosphorylation and exhibit vascular smooth muscle cell arrest in the synthetic state with excessive neointimal proliferation after carotid injury, as well as significant modifications of contractile properties and relaxant response to acetylcholine of femoral artery in vivo. Constitutively active I-1 gene transfer decreased neointimal formation in an angioplasty rat model by preventing vascular smooth muscle cell contractile to synthetic phenotype change. CONCLUSIONS: I-1 and sarco/endoplasmic reticulum Ca2+ -ATPase synergistically induce the vascular smooth muscle cell contractile phenotype. Gene transfer of constitutively active I-1 is a promising therapeutic strategy for preventing vascular proliferative disorders.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Músculo Liso Vascular/metabolismo , Proteína Fosfatasa 1/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Vasoconstricción/fisiología , Animales , Aorta Torácica/citología , Aorta Torácica/fisiología , Señalización del Calcio/fisiología , Vasos Coronarios/citología , Vasos Coronarios/fisiología , Arteria Femoral/citología , Arteria Femoral/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Arterias Mamarias/citología , Arterias Mamarias/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Fenotipo , Proteína Fosfatasa 1/genética , Proteínas/genética , Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Enfermedades Vasculares/genética , Enfermedades Vasculares/metabolismo
4.
Proc Natl Acad Sci U S A ; 108(32): 13258-63, 2011 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-21788490

RESUMEN

Myocardial ischemic disease is the major cause of death worldwide. After myocardial infarction, reperfusion of infracted heart has been an important objective of strategies to improve outcomes. However, cardiac ischemia/reperfusion (I/R) is characterized by inflammation, arrhythmias, cardiomyocyte damage, and, at the cellular level, disturbance in Ca(2+) and redox homeostasis. In this study, we sought to determine how acute inflammatory response contributes to reperfusion injury and Ca(2+) homeostasis disturbance after acute ischemia. Using a rat model of I/R, we show that circulating levels of TNF-α and cardiac caspase-8 activity were increased within 6 h of reperfusion, leading to myocardial nitric oxide and mitochondrial ROS production. At 1 and 15 d after reperfusion, caspase-8 activation resulted in S-nitrosylation of the RyR2 and depletion of calstabin2 from the RyR2 complex, resulting in diastolic sarcoplasmic reticulum (SR) Ca(2+) leak. Pharmacological inhibition of caspase-8 before reperfusion with Q-LETD-OPh or prevention of calstabin2 depletion from the RyR2 complex with the Ca(2+) channel stabilizer S107 ("rycal") inhibited the SR Ca(2+) leak, reduced ventricular arrhythmias, infarct size, and left ventricular remodeling after 15 d of reperfusion. TNF-α-induced caspase-8 activation leads to leaky RyR2 channels that contribute to myocardial remodeling after I/R. Thus, early prevention of SR Ca(2+) leak trough normalization of RyR2 function is cardioprotective.


Asunto(s)
Caspasa 8/metabolismo , Ventrículos Cardíacos/patología , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/patología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Activación Enzimática , Fluorescencia , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Fenantridinas/metabolismo , Ratas , Ratas Endogámicas WKY , Factor de Necrosis Tumoral alfa/sangre , Remodelación Ventricular
5.
FASEB J ; 26(3): 1009-17, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22090316

RESUMEN

Recent studies indicate that members of the multidrug-resistance protein (MRP) family belonging to ATP binding cassette type C (ABCC) membrane proteins extrude cyclic nucleotides from various cell types. This study aimed to determine whether MRP proteins regulate cardiac cAMP homeostasis. Here, we demonstrate that MRP4 is the predominant isoform present at the plasma membrane of cardiacmyocytes and that it mediates the efflux of cAMP in these cells. MRP4-deficient mice displayed enhanced cardiac myocyte cAMP formation, contractility, and cardiac hypertrophy at 9 mo of age, an effect that was compensated transiently by increased phosphodiesterase expression at young age. These findings suggest that cAMP extrusion via MRP4 acts together with phosphodiesterases to control cAMP levels in cardiac myocytes.


Asunto(s)
AMP Cíclico/metabolismo , Homeostasis , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Miocitos Cardíacos/metabolismo , 1-Metil-3-Isobutilxantina/farmacología , Animales , Western Blotting , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/genética , Cardiomegalia/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Ecocardiografía , Femenino , Regulación Enzimológica de la Expresión Génica , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Ratones , Ratones Noqueados , Microscopía Confocal , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Contracción Miocárdica/genética , Contracción Miocárdica/fisiología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Inhibidores de Fosfodiesterasa/farmacología , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
6.
J Mol Cell Cardiol ; 53(6): 801-8, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22884737

RESUMEN

Cardiac gene transfer is a powerful molecular tool to improve our understanding of the role of new proteins and mutants in cardiac pathophysiology. There is a need for a simple efficient myocardial gene delivery technique in order to study the physiological role of proteins in their native environment. Here we tested a new method of myocardial nonviral gene delivery, by using the combination of ultrasound energy (USE), liposomes and high pressure injections to the rat heart. Wistar rats were subjected to intra-myocardial injections of liposomes-DNA or siRNA mix. The heart was exposed after an inter-costal incision, and then injections were conducted between two sets of USE heart exposure. Ultrasound application resulted in much higher transfection efficiency (2% of left ventricle) than the liposomes-DNA alone (0.12% of left ventricle) as shown by the beta-galactosidase staining. The ultrasonic based liposomes-DNA delivery resulted in low inflammatory response, as well as in low cardiac fibrosis as shown by total collagen staining. Quantitative real time polymerase chain reaction (PCR) showed that the ultrasonic delivery resulted in cardiac specific transduction. Moreover, 23,906±2197 and 71,883±4065 calcium tolerant transfected cardiac myocytes were isolated following the delivery of a GFP plasmid or tagged siRNA, respectively. This was sufficient to perform single cell physiological measurements and biochemical experiments on homogenates. We developed an interesting safe method for local gene transfer in the heart using ultrasound and liposomes gene delivery. This method is particularly useful to study the effect of gene transfer on cardiac myocytes maintained in their normal environment in animal models.


Asunto(s)
Técnicas de Transferencia de Gen , Miocardio/metabolismo , Ultrasonido/métodos , Animales , Técnicas de Transferencia de Gen/instrumentación , Liposomas , Hígado/metabolismo , Pulmón/metabolismo , Ratones , Contracción Miocárdica , Miocardio/citología , Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Plásmidos/genética , Ratas , Bazo/metabolismo , Transfección/métodos , Ultrasonido/instrumentación , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
7.
Am J Physiol Cell Physiol ; 303(10): C1104-14, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23015549

RESUMEN

Plasticity-related gene-1 (PRG-1) protects neuronal cells from lysophosphatidic acid (LPA) effects. In vascular smooth muscle cells (VSMCs), LPA was shown to induce phenotypic modulation in vitro and vascular remodeling in vivo. Thus we explored the role of PRG-1 in modulating VSMC response to LPA. PCR, Western blot, and immunofluorescence experiments showed that PRG-1 is expressed in rat and human vascular media. PRG-1 expression was strongly inhibited in proliferating compared with quiescent VSMCs both in vitro and in vivo (medial vs. neointimal VSMCs), suggesting that PRG-1 expression is dependent on the cell phenotype. In vitro, adenovirus-mediated overexpression of PRG-1 specifically inhibited LPA-induced rat VSMC proliferation and migration but not platelet-derived growth factor-induced proliferation. This effect was abolished by mutation of a conserved histidine in the lipid phosphate phosphatase family that is essential for interaction with lipid phosphates. In vivo, balloon-induced neointimal formation in rat carotid was significantly decreased in vessels infected with PRG-1 adenovirus compared with ß-galactosidase adenovirus (-71%; P < 0.05). PRG-1 overexpression abolished the activation of the p42/p44 signaling pathway in LPA-stimulated rat VSMCs in culture and in balloon-injured rat carotids. Taken together, these findings provide the first evidence of a protective role of PRG-1 in the vascular media under pathophysiological conditions.


Asunto(s)
Proteínas de Unión a Calmodulina/metabolismo , Movimiento Celular/efectos de los fármacos , Lisofosfolípidos/farmacología , Músculo Liso Vascular/fisiología , Monoéster Fosfórico Hidrolasas/metabolismo , Adenoviridae , Animales , Proteínas de Unión a Calmodulina/genética , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Regulación de la Expresión Génica/fisiología , Vectores Genéticos , Humanos , Masculino , Músculo Liso Vascular/efectos de los fármacos , Neointima/inducido químicamente , Monoéster Fosfórico Hidrolasas/genética , Ratas , Ratas Wistar
8.
Circulation ; 124(7): 796-805, 2011 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-21810664

RESUMEN

BACKGROUND: Cardiomyocytes use Ca2+ not only in excitation-contraction coupling but also as a signaling molecule promoting, for example, cardiac hypertrophy. It is largely unclear how Ca2+ triggers signaling in cardiomyocytes in the presence of the rapid and large Ca2+ fluctuations that occur during excitation-contraction coupling. A potential route is store-operated Ca2+ entry, a drug-inducible mechanism for Ca2+ signaling that requires stromal interaction molecule 1 (STIM1). Store-operated Ca2+ entry can also be induced in cardiomyocytes, which prompted us to study STIM1-dependent Ca2+ entry with respect to cardiac hypertrophy in vitro and in vivo. METHODS AND RESULTS: Consistent with earlier reports, we found drug-inducible store-operated Ca2+ entry in neonatal rat cardiomyocytes, which was dependent on STIM1. Although this STIM1-dependent, drug-inducible store-operated Ca2+ entry was only marginal in adult cardiomyocytes isolated from control hearts, it increased significantly in cardiomyocytes isolated from adult rats that had developed compensated cardiac hypertrophy after abdominal aortic banding. Moreover, we detected an inwardly rectifying current in hypertrophic cardiomyocytes that occurs under native conditions (i.e., in the absence of drug-induced store depletion) and is dependent on STIM1. By manipulating its expression, we found STIM1 to be both sufficient and necessary for cardiomyocyte hypertrophy in vitro and in the adult heart in vivo. Stim1 silencing by adeno-associated viruses of serotype 9-mediated gene transfer protected rats from pressure overload-induced cardiac hypertrophy. CONCLUSION: By controlling a previously unrecognized sarcolemmal current, STIM1 promotes cardiac hypertrophy.


Asunto(s)
Señalización del Calcio/fisiología , Cardiomegalia/fisiopatología , Glicoproteínas de Membrana/fisiología , Miocitos Cardíacos/fisiología , Adenoviridae/genética , Factores de Edad , Animales , Animales Recién Nacidos , Cafeína/farmacología , Calcio/metabolismo , Canales de Calcio , Señalización del Calcio/efectos de los fármacos , Cardiomegalia/metabolismo , Cardiomegalia/patología , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Silenciador del Gen , Técnicas de Transferencia de Gen , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Transgénicos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Técnicas de Placa-Clamp , Inhibidores de Fosfodiesterasa/farmacología , Ratas , Sarcolema/metabolismo , Molécula de Interacción Estromal 1 , Tapsigargina/farmacología
9.
Adv Exp Med Biol ; 740: 795-810, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22453970

RESUMEN

Cyclic variations in calcium (Ca(2+)) concentrations, through a process called excitation-contraction coupling, allow regulation of vascular smooth muscle cells contractility and thus modulation of vascular tone and blood pressure. As a second messenger, Ca(2+) also activates signaling cascades leading to transcription factors activation in a process called excitation-transcription coupling. Furthermore, recent evidences indicate an interaction between post-transcriptional regulation by microRNAs (miRNAs) and Ca(2+) signaling. All these actors, which are frequently altered in vascular diseases, will be reviewed here.


Asunto(s)
Señalización del Calcio/fisiología , Músculo Liso Vascular/metabolismo , Animales , Calcio/metabolismo , Canales de Calcio/fisiología , Acoplamiento Excitación-Contracción , Humanos , Retículo Sarcoplasmático/metabolismo , Enfermedades Vasculares/metabolismo
10.
J Mol Cell Cardiol ; 50(4): 621-33, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21195084

RESUMEN

In blood vessels, tone is maintained by agonist-induced cytosolic Ca(2+) oscillations of quiescent/contractile vascular smooth muscle cells (VSMCs). However, in synthetic/proliferative VSMCs, Gq/phosphoinositide receptor-coupled agonists trigger a steady-state increase in cytosolic Ca(2+) followed by a Store Operated Calcium Entry (SOCE) which translates into activation of the proliferation-associated transcription factor NFAT. Here, we report that in human coronary artery smooth muscle cells (hCASMCs), the sarco/endoplasmic reticulum calcium ATPase type 2a (SERCA2a) expressed in the contractile form of the hCASMCs, controls the nature of the agonist-induced Ca(2+) transient and the resulting down-stream signaling pathway. Indeed, restoring SERCA2a expression by gene transfer in synthetic hCASMCs 1) increased Ca(2+) storage capacity; 2) modified agonist-induced IP(3)R Ca(2+) release from steady-state to oscillatory mode (the frequency of agonist-induced IP(3)R Ca(2+) signal was 11.66 ± 1.40/100 s in SERCA2a-expressing cells (n=39) vs 1.37 ± 0.20/100 s in control cells (n=45), p<0.01); 3) suppressed SOCE by preventing interactions between SR calcium sensor STIM1 and pore forming unit ORAI1; 4) inhibited calcium regulated transcription factor NFAT and its down-stream physiological function such as proliferation and migration. This study provides evidence for the first time that oscillatory and steady-state patterns of Ca(2+) transients have different effects on calcium-dependent physiological functions in smooth muscle cells.


Asunto(s)
Señalización del Calcio/fisiología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Factores de Transcripción NFATC/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Western Blotting , Calcio/metabolismo , Señalización del Calcio/genética , Movimiento Celular/genética , Movimiento Celular/fisiología , Proliferación Celular , Células Cultivadas , Vasos Coronarios/citología , Ciclina D1/genética , Ciclina D1/metabolismo , Humanos , Inmunoprecipitación , Microscopía Confocal , Modelos Biológicos , Factores de Transcripción NFATC/genética , Reacción en Cadena de la Polimerasa , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
11.
J Clin Invest ; 118(8): 2747-57, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18636120

RESUMEN

The second messengers cAMP and cGMP can be degraded by specific members of the phosphodiesterase superfamily or by active efflux transporters, namely the multidrug resistance-associated proteins (MRPs) MRP4 and MRP5. To determine the role of MRP4 and MRP5 in cell signaling, we studied arterial SMCs, in which the effects of cyclic nucleotide levels on SMC proliferation have been well established. We found that MRP4, but not MRP5, was upregulated during proliferation of isolated human coronary artery SMCs and following injury of rat carotid arteries in vivo. MRP4 inhibition significantly increased intracellular cAMP and cGMP levels and was sufficient to block proliferation and to prevent neointimal growth in injured rat carotid arteries. The antiproliferative effect of MRP4 inhibition was related to PKA/CREB pathway activation. Here we provide what we believe to be the first evidence that MRP4 acts as an independent endogenous regulator of intracellular cyclic nucleotide levels and as a mediator of cAMP-dependent signal transduction to the nucleus. We also identify MRP4 inhibition as a potentially new way of preventing abnormal VSMC proliferation.


Asunto(s)
Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Músculo Liso Vascular/metabolismo , Transducción de Señal , Animales , Proliferación Celular , Células Cultivadas , Vasos Coronarios/citología , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Humanos , Masculino , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Músculo Liso Vascular/citología , Ratas , Ratas Wistar
12.
J Pharmacol Exp Ther ; 336(3): 643-51, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21139058

RESUMEN

Wound healing after angioplasty or stenting is associated with increased production of thrombin and the activation of protease-activated receptor 1 (PAR1). The aim of the present study was to examine the effects of a new selective PAR1 antagonist, 2-[5-oxo-5-(4-pyridin-2-ylpiperazin-1-yl)-penta-1,3-dienyl]-benzonitrile (F 16618), in restenosis and vascular smooth muscle cell (SMC) proliferation and migration using both in vivo and in vitro approaches. Daily oral administration of F 16618 inhibited the restenosis induced by balloon angioplasty on rat carotid artery in a dose-dependent manner. Furthermore, single intravenous administration of F 16618 during the angioplasty procedure was sufficient to protect the carotid artery against restenosis. In vitro, F 16618 inhibited the growth of human aortic SMCs in a concentration-dependent manner with maximal effects at 10 µM. At that concentration, F 16618 also prevented thrombin-mediated SMC migration. In vivo, oral and intravenous F 16618 treatments reduced by 30 and 50% the expression of the inflammatory cytokine tumor necrosis factor α (TNFα) 24 h after angioplasty. However, only acute intravenous administration prevented the induction of matrix metalloproteinase 7 expression. In contrast, F 16618 treatments had no effect on early SMC de-differentiation and transcription of monocyte chemoattractant protein-1 and interleukin-6 and late re-endothelialization of injured arteries. Furthermore, F 16618 compensated for the carotid endothelium loss by inhibiting PAR1-mediated contraction. Altogether, these data demonstrate that PAR1 antagonists such as F 16618 are a highly effective treatment of restenosis after vascular injury, by inhibition of TNFα, matrix metalloproteinase 7, and SMC migration and proliferation in addition to an antithrombotic effect.


Asunto(s)
Estenosis Carotídea/prevención & control , Movimiento Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Inhibidores de la Metaloproteinasa de la Matriz , Músculo Liso Vascular/efectos de los fármacos , Piperazinas/farmacología , Piridinas/farmacología , Receptor PAR-1/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Estenosis Carotídea/metabolismo , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Reestenosis Coronaria/tratamiento farmacológico , Reestenosis Coronaria/metabolismo , Regulación hacia Abajo/fisiología , Regulación Enzimológica de la Expresión Génica , Humanos , Masculino , Metaloproteinasa 7 de la Matriz/biosíntesis , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/metabolismo , Piperazinas/uso terapéutico , Piridinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptor PAR-1/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
13.
J Pathol ; 221(3): 331-42, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20527026

RESUMEN

Recently, we discovered on primary cell cultures that adenylyl cyclase type 8 (AC8) was involved in the transition of rat vascular smooth muscle cells (VSMCs) to an inflammatory phenotype. Here we demonstrate, in human vessels displaying early or advanced atherosclerotic lesions, that: (a) only intimal VSMCs strongly express AC8; and (b) very few AC8-positive VSMCs were detected in the medial layer, either in atherosclerotic or healthy arteries. Furthermore, over-expressing AC8 in primary rat VSMC cultures triggered the recolonization of a wounded zone and similar results were obtained in the presence of mitomycin, a potent inhibitor of proliferation. This phenomenon was prevented by silencing AC8. Indeed, in IL-1 beta-treated cells, AC8 silencing halted migration and decreased the matrix-metalloproteinases 2/9 secretion, known to be involved in VSMC migration. In vivo, we showed: (a) a pronounced up-regulation of AC8 expression in highly migrating VSMCs of the injured rat carotid artery; (b) an undetectable AC8 labelling in re-endothelized vessels where neo-intimal thickening had stopped. From our data, we conclude that AC8 expression appears closely linked to the properties developed by VSMCs in atherosclerosis and post-angioplasty neo-intima formation leading to restenosis. In addition, it reinforces the idea that VSMC responses to their cell environment greatly depend on the AC isoforms expressed and attributes a new role for AC8 in these pathological vascular processes.


Asunto(s)
Adenilil Ciclasas/fisiología , Aterosclerosis/enzimología , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Adenilil Ciclasas/genética , Adenilil Ciclasas/metabolismo , Animales , Aterosclerosis/patología , Traumatismos de las Arterias Carótidas/enzimología , Movimiento Celular/fisiología , Proliferación Celular , Células Cultivadas , Humanos , Interleucina-1beta/farmacología , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Persona de Mediana Edad , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , ARN Interferente Pequeño/genética , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
14.
Mol Ther ; 18(7): 1284-92, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20461063

RESUMEN

Congestive heart failure (HF) is associated with impaired endothelium-dependent nitric oxide-mediated vasodilatation. The aim of this study was to examine the effects of sarco/endoplasmic reticulum (ER) Ca(2+)-ATPase 2a (SERCA2a) gene transfer on endothelial function in a swine HF model. Two months after the creation of mitral regurgitation to induce HF, the animals underwent intracoronary injection of adeno-associated virus (AAV) carrying SERCA2a (n = 7) or saline (n = 6). At 4 months, coronary flow (CF) was measured in the mid-portion of the left anterior descending (LAD) artery. In the failing animals, CF was decreased significantly; SERCA2a gene transfer rescued CF to levels observed in sham-group [ml/min/g, 0.47 +/- 0.064 saline versus 0.89 +/- 0.116, SERCA2a; P < 0.05; 1.00 +/- 0. 185 sham P = NS (nonsignificant)]. In coronary arteries from HF animals, SERCA2a and endothelial isoform of nitric oxide synthase (eNOS) protein expression were decreased, but restored to normal levels by SERCA2a gene transfer. In human coronary artery endothelial cells (HCAECs), SERCA2a overexpression increased eNOS expression, phosphorylation, eNOS promoter activity, Ca(2+) storage capacity, and enhanced histamine-induced calcium oscillations, eNOS activity, and cyclic guanosine monophosphate (cGMP) production. Thus, SERCA2a gene transfer increases eNOS expression and activity by modulating calcium homeostasis to improve CF. These findings suggest that SERCA2a gene transfer improves vascular reactivity in the setting of HF.


Asunto(s)
Células Endoteliales/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Animales , Western Blotting , Línea Celular , Vasos Coronarios/citología , Femenino , Técnicas de Transferencia de Gen , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/terapia , Humanos , Inmunoprecipitación , Microscopía Confocal , Óxido Nítrico Sintasa de Tipo III/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Porcinos
15.
Mol Ther ; 17(3): 455-62, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19107116

RESUMEN

Our objective was to study the expression and function of stromal interaction molecule 1 (STIM1), an endoplasmic reticulum protein recently identified as the calcium sensor that regulated Ca(2+)-released activated channels in T cells. STIM1 was found to be upregulated in serum-induced proliferating human coronary artery smooth muscle cells (hCASMCs) as well as in the neointima of injured rat carotid arteries. Growth factors-induced proliferation was significantly lower in hCASMC transfected with STIM1 siRNA than in those transfected with scrambled siRNA (increase relative to 0.1% S: 116 +/- 12% and 184 +/- 16%, respectively, P < 0.01). To assess the role of STIM1 in preventing vascular smooth muscle cells (VSMCs) proliferation in vivo, we infected balloon-injured rat carotid arteries with an adenoviral vector expressing a short hairpin (sh) RNA against rat STIM1 mRNA (Ad-shSTIM1). Intima/media ratios reflecting the degree of restenosis were significantly lower in Ad-shSTIM1- infected arteries than in Ad-shLuciferase-infected arteries (0.34 +/- 0.02 vs. 0.92 +/- 0.11, P < 0.006). Finally, we demonstrated that silencing STIM1 prevents activation of the transcription factor NFAT (nuclear factor of activated T cell). In conclusion, STIM1 appears as a major regulator of in vitro and in vivo VSMC proliferation, representing a novel and original pharmacological target for prominent vascular proliferative diseases.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/deficiencia , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Proteínas de Neoplasias/deficiencia , ARN Interferente Pequeño/genética , Túnica Íntima/citología , Túnica Íntima/metabolismo , Transporte Activo de Núcleo Celular , Adenoviridae/genética , Animales , Secuencia de Bases , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Vasos Coronarios/citología , Vasos Coronarios/lesiones , Vasos Coronarios/metabolismo , Vectores Genéticos/genética , Humanos , Masculino , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Factores de Transcripción NFATC/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Ratas , Ratas Wistar , Molécula de Interacción Estromal 1 , Canales Catiónicos TRPC/metabolismo , Técnicas de Cultivo de Tejidos , Regulación hacia Arriba
16.
Curr Vasc Pharmacol ; 18(5): 507-516, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31284864

RESUMEN

OBJECTIVE: Restenosis is a frequent complication of angioplasty. It consists of a neointimal hyperplasia resulting from progression and migration of vascular smooth muscle cells (VSMC) into the vessel lumen. microRNA miR-223 has recently been shown to be involved in cardiovascular diseases including atherosclerosis, vascular calcification and arterial thrombosis. In this study, our aim was to assess the impact of miR-223 modulation on restenosis in a rat model of carotid artery after balloon injury. METHODS: The over and down-expression of miR-223 was induced by adenoviral vectors, containing either a pre-miR-223 sequence allowing artificial miR-223 expression or a sponge sequence, trapping the native microRNA, respectively. Restenosis was quantified on stained rat carotid sections. RESULTS: In vitro, three mRNA (Myocyte Enhancer Factor 2C (MEF2C), Ras homolog gene family, member B (RhoB) and Nuclear factor 1 A-type (NFIA)) reported as miR-223 direct targets and known to be implicated in VSMC differentiation and contractility were studied by RT-qPCR. Our findings showed that down-expression of miR-223 significantly reduced neointimal hyperplasia by 44% in carotids, and was associated with a 2-3-fold overexpression of MEF2C, RhoB and NFIA in a murine monocyte macrophage cell line, RAW 264.7 cells. CONCLUSION: Down-regulating miR-223 could be a potential therapeutic approach to prevent restenosis after angioplasty.


Asunto(s)
Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/terapia , Estenosis Carotídea/prevención & control , Terapia Genética , MicroARNs/metabolismo , Angioplastia de Balón , Animales , Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Estenosis Carotídea/genética , Estenosis Carotídea/metabolismo , Estenosis Carotídea/patología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Masculino , Ratones , MicroARNs/genética , Neointima , Células RAW 264.7 , Ratas Wistar , Transducción de Señal
17.
Sci Rep ; 9(1): 6047, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30988334

RESUMEN

Early adaptive cardiac hypertrophy (EACH) is initially a compensatory process to optimize pump function. We reported the emergence of Orai3 activity during EACH. This study aimed to characterize how inflammation regulates store-independent activation of Orai3-calcium influx and to evaluate the functional role of this influx. Isoproterenol infusion or abdominal aortic banding triggered EACH. TNFα or conditioned medium from cardiac CD11b/c cells activated either in vivo [isolated from rats displaying EACH], or in vitro [isolated from normal rats and activated with lipopolysaccharide], were added to adult cardiomyocytes before measuring calcium entry, cell hypertrophy and cell injury. Using intramyocardial injection of siRNA, Orai3 was in vivo knockdown during EACH to evaluate its protective activity in heart failure. Inflammatory CD11b/c cells trigger a store-independent calcium influx in hypertrophied cardiomyocytes, that is mimicked by TNFα. Pharmacological or molecular (siRNA) approaches demonstrate that this calcium influx, depends on TNFR2, is Orai3-driven, and elicits cardiomyocyte hypertrophy and resistance to oxidative stress. Neutralization of Orai3 inhibits protective GSK3ß phosphorylation, impairs EACH and accelerates heart failure. Orai3 exerts a pathophysiological protective impact in EACH promoting hypertrophy and resistance to oxidative stress. We highlight inflammation arising from CD11b/c cells as a potential trigger of TNFR2- and Orai3-dependent signaling pathways.


Asunto(s)
Canales de Calcio/metabolismo , Cardiomegalia/inmunología , Insuficiencia Cardíaca/inmunología , Miocitos Cardíacos/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Animales , Antígeno CD11b/metabolismo , Antígeno CD11c/metabolismo , Calcio/metabolismo , Cardiomegalia/inducido químicamente , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Medios de Cultivo Condicionados/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Isoproterenol/toxicidad , Masculino , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/inmunología , Fosforilación/inmunología , ARN Interferente Pequeño/metabolismo , Ratas , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
18.
Circ Res ; 97(12): 1296-304, 2005 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-16269655

RESUMEN

cAMP is one of the most important second messenger in the heart. The discovery of Epac as a guanine exchange factor (GEF), which is directly activated by cAMP, raises the question of the role of this protein in cardiac cells. Here we show that Epac activation leads to morphological changes and induces expression of cardiac hypertrophic markers. This process is associated with a Ca2+-dependent activation of the small GTPase, Rac. In addition, we found that Epac activates a prohypertrophic signaling pathway, which involves the Ca2+ sensitive phosphatase, calcineurin, and its primary downstream effector, NFAT. Rac is involved in Epac-induced NFAT dependent cardiomyocyte hypertrophy. Blockade of either calcineurin or Rac activity blunts the hypertrophic response elicited by Epac indicating these signaling molecules coordinately regulate cardiac gene expression and cellular growth. Our results thus open new insights into the signaling pathways by which cAMP may mediate its biological effects and identify Epac as a new positive regulator of cardiac growth.


Asunto(s)
Cardiomegalia/etiología , Factores de Intercambio de Guanina Nucleótido/fisiología , Transporte Activo de Núcleo Celular , Adenoviridae/genética , Animales , Factor Natriurético Atrial/genética , Calcineurina/fisiología , Calcio/metabolismo , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Genes fos , Factores de Transcripción NFATC/metabolismo , Regiones Promotoras Genéticas , Ratas , Transducción de Señal , Tionucleótidos/farmacología , Proteínas de Unión al GTP rac/metabolismo
19.
Circ Res ; 97(5): 488-95, 2005 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-16081870

RESUMEN

Proliferation of vascular smooth muscle cells (VSMC) is a primary cause of vascular disorders and is associated with major alterations in Ca2+ handling supported by loss of the sarco/endoplasmic reticulum calcium ATPase, SERCA2a. To determine the importance of SERCA2a in neointima formation, we have prevented loss of its expression by adenoviral gene transfer in a model of balloon injury of the rat carotid artery. Two weeks after injury, the intima/media ratio was significantly lower in SERCA2a-infected than in injured noninfected or injured beta-galactosidase-infected carotids (0.29+/-0.04 versus 0.89+/-0.19 and 0.72+/-0.14, respectively; P<0.05), and was comparable to that observed in control carotids (0.21+/-0.03). The pathways leading to proliferation were analyzed in serum-stimulated VSMC. Forced expression of SERCA2a arrested cell cycle at the G1 phase and prevented apoptosis. SERCA2a inhibits proliferation through inactivation of calcineurin (PP2B) and its target transcription factor NFAT (nuclear factor of activated T-cells) resulting in lowering of cyclin D1 and pRb levels. By using NFAT-competing peptide VIVIT, we showed that NFAT activity is strongly required to promote VSMC proliferation. In conclusion, we provide the first evidence that increasing SERCA2a activity inhibits VSMC proliferation and balloon injury-induced neointima formation.


Asunto(s)
ATPasas Transportadoras de Calcio/fisiología , Músculo Liso Vascular/citología , Túnica Íntima/patología , Adenosina Trifosfato/farmacología , Animales , Apoptosis , Inhibidores de la Calcineurina , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , ATPasas Transportadoras de Calcio/genética , Enfermedades de las Arterias Carótidas/prevención & control , Proliferación Celular , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Transferencia de Gen Horizontal , Terapia Genética , Masculino , Factores de Transcripción NFATC , Proteínas Nucleares/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico , Factores de Transcripción/metabolismo
20.
Biochem J ; 394(Pt 1): 27-33, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16250893

RESUMEN

Histamine, known to induce Ca2+ oscillations in endothelial cells, was used to alter Ca2+ cycling. Treatment of HUVEC (human umbilical-vein endothelial cell)-derived EA.hy926 cells with histamine for 1-3 days increased the levels of SERCA (sarcoplasmic/endoplasmic reticulum Ca2+-ATPase) 3, but not of SERCA 2b, transcripts and proteins. Promoter-reporter gene assays demonstrated that this increase in expression was due to activation of SERCA 3 gene transcription. The effect of histamine was abolished by mepyramine, but not by cimetidine, indicating that the H1 receptor, but not the H2 receptor, was involved. The histamine-induced up-regulation of SERCA 3 was abolished by cyclosporin A and by VIVIT, a peptide that prevents calcineurin and NFAT (nuclear factor of activated T-cells) from interacting, indicating involvement of the calcineurin/NFAT pathway. Histamine also induced the nuclear translocation of NFAT. NFAT did not directly bind to the SERCA 3 promoter, but activated Ets-1 (E twenty-six-1), which drives the expression of the SERCA 3 gene. Finally, cells treated with histamine and loaded with fura 2 exhibited an improved capacity in eliminating high cytosolic Ca2+ concentrations, in accordance with an increase in activity of a low-affinity Ca2+-ATPase, like SERCA 3. Thus chronic treatment of endothelial cells with histamine up-regulates SERCA 3 transcription. The effect of histamine is mediated by the H1R (histamine 1 receptor) and involves activation of the calcineurin/NFAT pathway. By increasing the rate of Ca2+ sequestration, up-regulation of SERCA 3 counteracts the cytosolic increase in Ca2+ concentration.


Asunto(s)
Calcineurina/metabolismo , ATPasas Transportadoras de Calcio/genética , Células Endoteliales/metabolismo , Regulación Enzimológica de la Expresión Génica , Factores de Transcripción NFATC/metabolismo , Transducción de Señal , Transcripción Genética , Calcineurina/genética , Calcio/metabolismo , Línea Celular , Histamina , Antagonistas de los Receptores Histamínicos , Humanos , Factores de Transcripción NFATC/genética , Proteína Proto-Oncogénica c-ets-1/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA