Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(13)2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-39000009

RESUMEN

Yohimbine (YHB) has been reported to possess anti-inflammatory, anticancer, and cardiac function-enhancing properties. Additionally, it has been reported to inhibit the proliferation, migration, and neointimal formation of vascular smooth muscle cells (VSMCs) induced by platelet-derived growth factor (PDGF) stimulation by suppressing the phospholipase C-gamma 1 pathway. However, the transcriptional regulatory mechanism of YHB controlling the behavior of VSMCs is not fully understood. In this study, YHB downregulated the expression of cell cycle regulatory proteins, such as proliferating cell nuclear antigen (PCNA), cyclin D1, cyclin-dependent kinase 4 (CDK4), and cyclin E, by modulating the transcription factor FOXO3a in VSMCs induced by PDGF. Furthermore, YHB decreased p-38 and mTOR phosphorylation in a dose-dependent manner. Notably, YHB significantly reduced the phosphorylation at Y397 and Y925 sites of focal adhesion kinase (FAK), and this effect was greater at the Y925 site than Y397. In addition, the expression of paxillin, a FAK-associated protein known to bind to the Y925 site of FAK, was significantly reduced by YHB treatment in a dose-dependent manner. A pronounced reduction in the migration and proliferation of VSMCs was observed following co-treatment of YHB with mTOR or p38 inhibitors. In conclusion, this study shows that YHB inhibits the PDGF-induced proliferation and migration of VSMCs by regulating the transcription factor FOXO3a and the mTOR/p38/FAK signaling pathway. Therefore, YHB may be a potential therapeutic candidate for preventing and treating cardiovascular diseases such as atherosclerosis and vascular restenosis.


Asunto(s)
Movimiento Celular , Proliferación Celular , Proteína Forkhead Box O3 , Músculo Liso Vascular , Miocitos del Músculo Liso , Factor de Crecimiento Derivado de Plaquetas , Yohimbina , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Proteína Forkhead Box O3/metabolismo , Proliferación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Animales , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Fosforilación/efectos de los fármacos , Yohimbina/farmacología , Ratas , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Células Cultivadas , Paxillin/metabolismo , Ratas Sprague-Dawley , Masculino
2.
Int J Mol Sci ; 24(12)2023 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-37373052

RESUMEN

Abnormal communication between endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) promotes vascular diseases, including atherogenesis. ETS variant transcription factor 2 (ETV2) plays a substantial role in pathological angiogenesis and the reprogramming of ECs; however, the role of ETV2 in the communication between ECs and VSMCs has not been revealed. To investigate the interactive role of ETV2 in the EC to VSMC phenotype, we first showed that treatment with a conditioned medium from ETV2-overexpressed ECs (Ad-ETV2 CM) significantly increased VSMC migration. The cytokine array showed altered levels of several cytokines in Ad-ETV2 CM compared with those in normal CM. We found that C-X-C motif chemokine 5 (CXCL5) promoted VSMC migration using the Boyden chamber and wound healing assays. In addition, an inhibitor of C-X-C motif chemokine receptor 2 (CXCR2) (the receptor for CXCL5) significantly inhibited this process. Gelatin zymography showed that the activities of matrix metalloproteinase (MMP)-2 and MMP-9 increased in the media of VSMCs treated with Ad-ETV2 CM. Western blotting revealed a positive correlation between Akt/p38/c-Jun phosphorylation and CXCL5 concentration. The inhibition of Akt and p38-c-Jun effectively blocked CXCL5-induced VSMC migration. In conclusion, CXCL5 from ECs induced by ETV2 promotes VSMC migration via MMP upregulation and the activation of Akt and p38/c-Jun.


Asunto(s)
Músculo Liso Vascular , Proteínas Proto-Oncogénicas c-akt , Células Cultivadas , Células Endoteliales , Movimiento Celular , Citocinas/farmacología , Miocitos del Músculo Liso
3.
Part Fibre Toxicol ; 19(1): 29, 2022 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-35449013

RESUMEN

BACKGROUND: Epidemiological studies have suggested that elevated concentrations of particulate matter (PM) are strongly associated with the incidence of atherosclerosis, however, the underlying cellular and molecular mechanisms of atherosclerosis by PM exposure and the components that are mainly responsible for this adverse effect remain to be established. In this investigation, we evaluated the effects of ambient PM on vascular smooth muscle cell (VSMC) behavior. Furthermore, the effects of polycyclic aromatic hydrocarbons (PAHs), major components of PM, on VSMC migration and the underlying mechanisms were examined. RESULTS: VSMC migration was significantly increased by treatment with organic matters extracted from ambient PM. The total amount of PAHs contained in WPM was higher than that in SPM, leading to higher ROS generation and VSMC migration. The increased migration was successfully inhibited by treatment with the anti-oxidant, N-acetyl-cysteine (NAC). The levels of matrix metalloproteinase (MMP) 2 and 9 were significantly increased in ambient PM-treated VSMCs, with MMP9 levels being significantly higher in WPM-treated VSMCs than in those treated with SPM. As expected, migration was significantly increased in all tested PAHs (anthracene, ANT; benz(a)anthracene, BaA) and their oxygenated derivatives (9,10-Anthraquinone, AQ; 7,12-benz(a)anthraquinone, BAQ, respectively). The phosphorylated levels of focal adhesion kinase (FAK) and formation of the focal adhesion complex were significantly increased in ambient PM or PAH-treated VSMCs, and these effects were blocked by administration of NAC or α-NF, an inhibitor of AhR, the receptor that allows PAH uptake. Subsequently, the levels of phosphorylated Src and NRF, the downstream targets of FAK, were altered with a pattern similar to that of p-FAK. CONCLUSIONS: PAHs, including oxy-PAHs, in ambient PM may have dual effects that lead to an increase in VSMC migration. One is the generation of oxidative stress followed by MMP upregulation, and the other is actin reorganization that results from the activation of the focal adhesion complex.


Asunto(s)
Contaminantes Atmosféricos , Aterosclerosis , Hidrocarburos Policíclicos Aromáticos , Actinas , Contaminantes Atmosféricos/análisis , Movimiento Celular , Humanos , Metaloproteinasas de la Matriz , Músculo Liso Vascular/química , Estrés Oxidativo , Material Particulado/análisis , Material Particulado/toxicidad , Hidrocarburos Policíclicos Aromáticos/análisis , Hidrocarburos Policíclicos Aromáticos/toxicidad , Regulación hacia Arriba
4.
J Med Food ; 25(5): 503-512, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35483086

RESUMEN

The abnormal change of vascular smooth muscle cell (VSMC) behavior is an important cellular event leading to neointimal hyperplasia in atherosclerosis and restenosis. Plantamajoside (PMS), a phenylethanoid glycoside compound of the Plantago asiatica, has been reported to have anti-inflammatory, antioxidative, and anticancer activities. In this study, the protective effects of PMS against intimal hyperplasia and the mechanisms underlying the regulation of VSMC behavior were investigated. MTT and BrdU assays were performed to evaluate the cytotoxicity and cell proliferative activity of PMS, respectively. Rat aortic VSMC migrations after treatment with the determined concentration of PMS (50 and 150 µM) were evaluated using wound healing and Boyden chamber assays. The inhibitory effects of PMS on intimal hyperplasia were evaluated in balloon-injured (BI) rat carotid artery. PMS suppressed the proliferation in platelet-derived growth factor-BB-induced VSMC, as confirmed from the decrease in cyclin-dependent kinase (CDK)-2, CDK-4, cyclin D1, and proliferating cell nuclear antigen levels. PMS also inhibited VSMC migration, consistent with the downregulated expression and zymolytic activities of matrix metalloproteinase (MMP)2, MMP9, and MMP13. PMS specifically regulated MMP expression through p38 mitogen-activated protein kinase and focal adhesion kinase pathways. Tissue inhibitor of metalloproteinase (TIMP)1 and TIMP2 levels were upregulated via Smad1. TIMPs inhibited the conversion of pro-MMPs to active MMPs. PMS significantly inhibited neointimal formation in BI rat carotid arteries. In conclusion, PMS inhibits VSMC proliferation and migration by upregulating TIMP1 and TIMP2 expression. Therefore, PMS could be a potential therapeutic agent for vascular atherosclerosis and restenosis treatment.


Asunto(s)
Aterosclerosis , Neointima , Animales , Aterosclerosis/metabolismo , Catecoles , Movimiento Celular , Proliferación Celular , Células Cultivadas , Glucósidos , Hiperplasia/tratamiento farmacológico , Hiperplasia/metabolismo , Hiperplasia/patología , Músculo Liso Vascular , Miocitos del Músculo Liso , Neointima/tratamiento farmacológico , Neointima/metabolismo , Neointima/patología , Ratas , Ratas Sprague-Dawley , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Inhibidores Tisulares de Metaloproteinasas/farmacología , Inhibidores Tisulares de Metaloproteinasas/uso terapéutico , Regulación hacia Arriba
5.
Int J Mol Sci ; 22(12)2021 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-34205318

RESUMEN

Low-density lipoprotein receptor-related protein 5 (LRP5) has been studied as a co-receptor for Wnt/ß-catenin signaling. However, its role in the ischemic myocardium is largely unknown. Here, we show that LRP5 may act as a negative regulator of ischemic heart injury via its interaction with prolyl hydroxylase 2 (PHD2), resulting in hypoxia-inducible factor-1α (HIF-1α) degradation. Overexpression of LRP5 in cardiomyocytes promoted hypoxia-induced apoptotic cell death, whereas LRP5-silenced cardiomyocytes were protected from hypoxic insult. Gene expression analysis (mRNA-seq) demonstrated that overexpression of LRP5 limited the expression of HIF-1α target genes. LRP5 promoted HIF-1α degradation, as evidenced by the increased hydroxylation and shorter stability of HIF-1α under hypoxic conditions through the interaction between LRP5 and PHD2. Moreover, the specific phosphorylation of LRP5 at T1492 and S1503 is responsible for enhancing the hydroxylation activity of PHD2, resulting in HIF-1α degradation, which is independent of Wnt/ß-catenin signaling. Importantly, direct myocardial delivery of adenoviral constructs, silencing LRP5 in vivo, significantly improved cardiac function in infarcted rat hearts, suggesting the potential value of LRP5 as a new target for ischemic injury treatment.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Isquemia Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Regulación de la Expresión Génica , Hidroxilación , Hipoxia/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Cultivo Primario de Células , Ratas , Vía de Señalización Wnt
6.
Part Fibre Toxicol ; 17(1): 25, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32527278

RESUMEN

BACKGROUND: Epidemiologic studies have suggested that elevated concentrations of particulate matter (PM) are strongly associated with an increased risk of developing cardiovascular diseases, including arrhythmia. However, the cellular and molecular mechanisms by which PM exposure causes arrhythmia and the component that is mainly responsible for this adverse effect remains to be established. In this study, the arrhythmogenicity of mobilized organic matter from two different types of PM collected during summer (SPM) and winter (WPM) seasons in the Seoul metropolitan area was evaluated. In addition, differential effects between polycyclic aromatic hydrocarbons (PAHs) and oxygenated PAHs (oxy-PAHs) on the induction of electrophysiological instability were examined. RESULTS: We extracted the bioavailable organic contents of ambient PM, measuring 10 µm or less in diameter, collected from the Seoul metropolitan area using a high-volume air sampler. Significant alterations in all factors tested for association with electrophysiological instability, such as intracellular Ca2+ levels, reactive oxygen species (ROS) generation, and mRNA levels of the Ca2+-regulating proteins, sarcoplasmic reticulum Ca2+ATPase (SERCA2a), Ca2+/calmodulin-dependent protein kinase II (CaMK II), and ryanodine receptor 2 (RyR2) were observed in cardiomyocytes treated with PM. Moreover, the alterations were higher in WPM-treated cardiomyocytes than in SPM-treated cardiomyocytes. Three-fold more oxy-PAH concentrations were observed in WPM than SPM. As expected, electrophysiological instability was induced higher in oxy-PAHs (9,10-anthraquinone, AQ or 7,12-benz(a) anthraquinone, BAQ)-treated cardiomyocytes than in PAHs (anthracene, ANT or benz(a) anthracene, BaA)-treated cardiomyocytes; oxy-PAHs infusion of cells mediated by aryl hydrocarbon receptor (AhR) was faster than PAHs infusion. In addition, ROS formation and expression of calcium-related genes were markedly more altered in cells treated with oxy-PAHs compared to those treated with PAHs. CONCLUSIONS: The concentrations of oxy-PAHs in PM were found to be higher in winter than in summer, which might lead to greater electrophysiological instability through the ROS generation and disruption of calcium regulation.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Contaminantes Atmosféricos/toxicidad , Miocitos Cardíacos/efectos de los fármacos , Oxígeno/química , Material Particulado/toxicidad , Hidrocarburos Policíclicos Aromáticos/toxicidad , Contaminantes Atmosféricos/química , Animales , Animales Recién Nacidos , Calcio/metabolismo , Células Cultivadas , Peroxidación de Lípido/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Tamaño de la Partícula , Material Particulado/química , Técnicas de Placa-Clamp , Hidrocarburos Policíclicos Aromáticos/química , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Estaciones del Año , Seúl
7.
Int J Cardiol ; 299: 235-242, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31350035

RESUMEN

BACKGROUND: Cardiac hypertrophy is associated with functional changes in cardiomyocytes, which often results in heart failure. The low-density lipoprotein receptor-related protein 1 (LRP1) is a large multifunctional endocytic receptor involved in many physiological and pathological processes. However, its function in the development of cardiac hypertrophy remains largely unclear. METHODS: Adenoviral constructs were used for either overexpression or silencing of LRP1 in both in vitro and in vivo experiments. Cardiac function was measured using the Millar catheter. RESULTS: LRP1 expression was upregulated in both transverse aortic constriction (TAC)-induced hypertrophic myocardium and catecholamine (phenylephrine (PE) and norepinephrine (NE))- and angiotensin II (AngII)-induced hypertrophic cardiomyocytes. In addition, cell surface area, protein/DNA ratio, and the mRNA levels of hypertrophic markers were significantly increased in LRP1-overexpressing cardiomyocytes without catecholamine stimulation. Conversely, LRP1 inhibition by LRP1-specific siRNA or a specific ligand-binding antagonist (RAP) significantly rescued hypertrophic effects in PE, NE, or AngII-induced cardiomyocytes. LRP1 overexpression induced PKCα, then activated ERK, resulting in cardiac hypertrophy with the downregulation of SERCA2a and calcium accumulation, which was successfully restored in both LRP1-silenced cardiomyocytes and TAC-induced hearts. CONCLUSIONS: LRP1 regulates cardiac hypertrophy via the PKCα-ERK dependent signaling pathway resulting in the alteration of intracellular calcium levels, demonstrating that LRP1 might be a potential therapeutic target for cardiac hypertrophy.


Asunto(s)
Cardiomegalia/metabolismo , Cardiomegalia/patología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/biosíntesis , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Animales , Humanos , Masculino , Ratas , Ratas Sprague-Dawley
8.
J Nanosci Nanotechnol ; 15(1): 112-5, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26328312

RESUMEN

Phytoncide, nanochemicals extracted from Chamaecyparis obtusa (C. obtusa), is reported to possess many pharmacological activities including immunological stimulating, anti-cancer, antioxidant, and antiinflammatory activities. However, the effect of phytoncide in vascuar diseases, especially on the behavior of vascular smooth muscle cells, has not yet been clearly elucidated. Therefore, in the present study, we investigated the effects of 15 kinds of phytoncide by various extraction conditions from C. obtusa on the proliferation and migration in rat aortic smooth muscle cells (RAoSMCs). First of all, we determined the concentration of each extracts not having cytotoxicity by MTT assay. We observed that the proliferation rate measured using BrdU assay was significantly reduced by supercritical fluid, steam distillation, Me-OH, and hexane extraction fraction in order with higher extent, respectively. Moreover, the treatment of above nanofractions inhibit the migration of RAoSMCs by 40%, 60%, and 30%, respectively, both in 2-D wound healing assay and 3-D boyden chamber assay. Immunoblot revealed that the phosphorylated levels of Akt and ERK were significantly reduced in nanofractions treated RAoSMCs. Taken together, these data suggest that phytoncide extracted from C. obtusa inhibits proliferation and migration in RAoSMCs via the modulation of phosphorylated levels of Akt and ERK. Therefore, phytoncide nanomolecules might be a potential therapeutic approach to prevent or treat atheroscrelosis and restenosis.


Asunto(s)
Chamaecyparis/química , Fitoquímicos/farmacología , Extractos Vegetales/farmacología , Cicatrización de Heridas/efectos de los fármacos , Animales , Aorta/citología , Aorta/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Fitoquímicos/química , Extractos Vegetales/química , Ratas , Ratas Sprague-Dawley
9.
J Nanosci Nanotechnol ; 15(1): 116-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26328313

RESUMEN

The plant Dendropanax morbifera Léveille (D. morbifera), a subtropical broad-leaved evergreen tree, have been used in folk medicine for the treatment of infectious diseases, skin diseases, and other maladies. However, the effect of extracts from D. morbifera in vascuar diseases has not yet been reported. In this study, BrdU assay revealed that extracts from D. morbifera inhibit significantly the proliferation rate of Rat Aortic Smooth Muscle Cells (RAoSMCs) by -40% in treated samples compared to controls. Notably, 2-D wound healing assay and 3-D boyden chamber assay showed the significant reduction of RAoSMCs migration induced by serum in nano extracts treated groups by -50%. We further observed that the phosphorylated levels of Akt and ERK were significantly reduced by 70% in extracts treated RAoSMCs. Moreover, the expression levels of matrix metalloproteinase (MMP) 2 and 9 were significantly reduced by extracts from D. morbifera. Our results suggest that extracts from D. morbifera inhibit proliferation and migration in RAoSMCs via the modulation of phosphorylated levels of Akt and ERK. Subsequently, the reduced MMP2 and 9 expression might result to reduced migration of RAoSMCs.


Asunto(s)
Araliaceae/química , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Extractos Vegetales/química , Extractos Vegetales/farmacología , Animales , Células Cultivadas , Músculo Liso Vascular/citología , Hojas de la Planta/química , Tallos de la Planta/química , Ratas , Ratas Sprague-Dawley
10.
Tissue Cell ; 47(1): 115-21, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25595313

RESUMEN

Migration of vascular smooth muscle cells (VSMCs) is an early event of atherosclerosis, which is mediated mainly by matrix metalloproteinase (MMP) 2 and 9. Because MMP13 is associated with tumor cells migration, we hypothesized that MMP13 participates in VSMC migration induced by certain stimuli such as platelet-derived growth factor (PDGF) and angiotensin II (Ang II). We found that the mRNA level of MMP13 in rat aortic smooth muscle cells (RAoSMCs) was increased by both PDGF and Ang II. We observed the significant decrease of migration in PDGF- or Ang II-treated RAoSMCs by MMP13 specific inhibitor treatment. Silencing of MMP13 by a specific small interfering RNA (siRNA) significantly decreased expression of the active form of MMP13, which is followed by the decreased migration of PDGF- or Ang II-treated RAoSMCs. Interestingly, we observed synergistic inhibitory effects on migration by treatment with MMP2 and 13 or MMP9 and 13 inhibitors compared with that in single treatments. Moreover, we found that cordycepin, a known inhibitor of VSMC migration, caused significant downregulation of MMP2, 9, and 13 expression in PDGF-treated RAoSMCs. We further show that the expression level of MMP13 was significantly decreased by the treatment of Akt or ERK specific inhibitor in PDGF-treated RAoSMCs. Together, our data strongly suggest that MMP13 involves VSMCs migration via an Akt and ERK-dependent regulation [corrected].


Asunto(s)
Movimiento Celular/efectos de los fármacos , Metaloproteinasa 13 de la Matriz/genética , Músculo Liso Vascular/efectos de los fármacos , Angiotensina II/administración & dosificación , Animales , Aorta/metabolismo , Aorta/patología , Desoxiadenosinas/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metaloproteinasa 13 de la Matriz/biosíntesis , Factor de Crecimiento Derivado de Plaquetas/administración & dosificación , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/biosíntesis , Ratas
11.
Heart Vessels ; 30(6): 728-33, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25048679

RESUMEN

The aim of this study was to evaluate the prognostic value of serum total bilirubin on the development of adverse outcomes after percutaneous coronary intervention (PCI) besides high-sensitivity cardiac troponin T (hs-cTnT) and N-terminal pro-B type natriuretic peptide (NT-proBNP). Serum total bilirubin, hs-cTnT, and NT-proBNP were analyzed in 372 patients who underwent PCI. The primary endpoint was cardiac death. There were 21 events of cardiac death during a mean of 25.8 months of follow-up. When the serum total bilirubin cut-off level (median value) was set to 0.58 mg/dL using the receiver operating characteristic curve, the sensitivity was 95.2 % and the specificity was 51.0 % for differentiating between the group with cardiac death and the group without cardiac death. Kaplan-Meier analysis revealed that the lower serum total bilirubin group (<0.58 mg/dL) had a significantly higher cardiac death rate than the higher serum total bilirubin group (≥0.58 mg/dL) (10.4 vs. 0.6 %, log-rank: P = 0.0001). In conclusion, low serum total bilirubin is a predictive marker for cardiac death after PCI.


Asunto(s)
Bilirrubina/sangre , Muerte , Péptido Natriurético Encefálico/sangre , Fragmentos de Péptidos/sangre , Intervención Coronaria Percutánea/efectos adversos , Troponina T/sangre , Anciano , Biomarcadores/sangre , Femenino , Estudios de Seguimiento , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Pronóstico , Modelos de Riesgos Proporcionales , Curva ROC , República de Corea , Estudios Retrospectivos , Factores de Riesgo , Resultado del Tratamiento
12.
Int J Cardiol ; 175(3): 554-9, 2014 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-25015023

RESUMEN

OBJECTIVE: The aim of this study was to determine the associations of brachial-ankle pulse wave velocity (baPWV), high-sensitivity cardiac troponin T (hs-cTnT) and N-terminal pro-B type natriuretic peptide (NT-proBNP) with the development of adverse outcomes after percutaneous coronary intervention (PCI). METHODS: The baPWV, hs-cTnT and NT-proBNP were analyzed in 372 patients who underwent PCI. The primary endpoint was cardiac death. RESULTS: There were 21 events of cardiac death during a mean of 25.8 months of follow-up. When the baPWV cut-off level was set to 1672 cm/s using the receiver operating characteristic curve, the sensitivity was 85.7% and the specificity was 60.1% for differentiating between the group with cardiac death and the group without cardiac death. Kaplan-Meier analysis revealed that the higher baPWV group (≥1672 cm/s) had a significantly higher cardiac death rate than the lower baPWV group (<1672 cm/s) (11.4% vs. 1.4%, log-rank: P<0.0001). This value was more useful in patients with myocardial injury (hs-cTnT≥0.1 ng/mL) or heart failure (NT-proBNP≥450 pg/mL). CONCLUSIONS: The results of this study show that high baPWV is a predictive marker for cardiac death after PCI.


Asunto(s)
Índice Tobillo Braquial/mortalidad , Índice Tobillo Braquial/tendencias , Muerte , Intervención Coronaria Percutánea/mortalidad , Intervención Coronaria Percutánea/tendencias , Flujo Pulsátil/fisiología , Anciano , Velocidad del Flujo Sanguíneo/fisiología , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Intervención Coronaria Percutánea/efectos adversos , Valor Predictivo de las Pruebas , República de Corea/epidemiología , Estudios Retrospectivos , Factores de Tiempo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA