Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Am Surg ; 85(4): 335-341, 2019 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-31043191

RESUMEN

The ACS NSQIP Surgical Risk Calculator (SRC) is an evidence-based clinical tool commonly used for evaluating postoperative risk. The goal of this study was to validate SRC-predicted complications by comparing them with observed outcomes in the acute care surgical setting. In this study, pre- and postoperative data from 1693 acute care surgeries (hernia repair, enterolysis, intestinal incision/excision and enterectomy, gastrectomy, debridement, colectomy, appendectomy, cholecystectomy, gastrorrhaphy, and incision and drainage of soft tissue, breast abscesses, and removal of foreign bodies) performed at a Level I trauma center over a five-year time period were abstracted. Predictions for any and serious complications were based on SRC were compared with observed outcomes using various measures of diagnostic. When evaluated as one group, the SRC had good discriminative power for predicting any and serious complications after acute care surgeries (Area Under the Curve (AUC) 0.79, 0.81). In addition, the SRC met Brier score requirements for an informative model overall. However, the predictive accuracy of the SRC varied for various procedures within the acute care patient population. For serious complications, the diagnostic measures ranged from an AUC of 0.61 and negative likelihood ratio of 0.716 for incision & drainage soft tissue to AUC of 0.91 and negative likelihood ratio of 0.064 for gastrorrhaphy. Length of stay was significantly underestimated by the SRC overall (8.56 days, P < 0.01) and for individual procedures. The SRC performs well at predicting complications after acute care surgeries overall; however, there is great variability in performance between procedure types. Further refinements in risk stratification may improve SRC predictions.


Asunto(s)
Cuidados Críticos , Técnicas de Apoyo para la Decisión , Complicaciones Posoperatorias/diagnóstico , Humanos , Complicaciones Posoperatorias/etiología , Estudios Retrospectivos , Medición de Riesgo/métodos , Factores de Riesgo , Sensibilidad y Especificidad
2.
Int J Cardiol ; 270: 228-236, 2018 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-30220377

RESUMEN

BACKGROUND: Excessive cardiac fibrosis due to maladaptive remodeling leads to progression of cardiac dysfunction and is modulated by TGF-ß1-activated intracellular phospho-SMAD signaling effectors and transcription regulators. SMAD2/3 phosphorylation, regulated by protein-phosphatases, has been studied in different cell types, but its role in human ventricular fibroblasts (hVFs) is not defined as a target to reduce cytokine-mediated excessive fibrotic response and adverse cardiac remodeling. Statins are a class of drugs reported to reduce cardiac fibrosis, although underlying mechanisms are not completely understood. We aimed to assess whether simvastatin-mediated reduction in TGF-ß1-augmented profibrotic response involves reduction in phospho-SMAD2/3 owing to activation of protein-phosphatase in hVFs. METHODS AND RESULTS: Cultures of hVFs were used. Effect of simvastatin on TGF-ß1-treated hVF proliferation, cytotoxicity, myofibroblast differentiation/activation, profibrotic gene expression and protein-phosphatase activity was assessed. Simvastatin (1 µM) reduced effect of TGF-ß1 (5 ng/mL) on hVF proliferation, myofibroblast differentiation (reduced α-smooth muscle actin [α-SMA-expression]) and activation (decreased procollagen-peptide release). Simvastatin also reduced TGF-ß1-stimulated time-dependent increases in SMAD2/3 phosphorylation and nuclear translocation, mediated through catalytic activation of protein-phosphatases PPM1A and PP2A, which physically interact with SMAD2/3, thereby promoting their dephosphorylation. Effect of simvastatin on TGF-ß1-induced fibroblast activation was annulled by okadaic acid, an inhibitor of protein-phosphatase. CONCLUSIONS: This proof-of-concept study using an in vitro experimental cell culture model identifies the protective role of simvastatin against TGF-ß1-induced hVF transformation into activated myofibroblasts through activation of protein phosphatase, a novel target that can be therapeutically modulated to curb excessive cardiac fibrosis associated with maladaptive cardiac remodeling.


Asunto(s)
Fibroblastos/metabolismo , Proteína Fosfatasa 2C/metabolismo , Proteína Fosfatasa 2/metabolismo , Simvastatina/farmacología , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/toxicidad , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Fibroblastos/efectos de los fármacos , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/metabolismo , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2C/antagonistas & inhibidores
3.
Am J Physiol Cell Physiol ; 311(2): C330-9, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27335167

RESUMEN

Fibroblasts, the most abundant cells in the heart, contribute to cardiac fibrosis, the substrate for the development of arrythmogenesis, and therefore are potential targets for preventing arrhythmic cardiac remodeling. A chamber-specific difference in the responsiveness of fibroblasts from the atria and ventricles toward cytokine and growth factors has been described in animal models, but it is unclear whether similar differences exist in human cardiac fibroblasts (HCFs) and whether drugs affect their proliferation differentially. Using cardiac fibroblasts from humans, differences between atrial and ventricular fibroblasts in serum-induced proliferation, DNA synthesis, cell cycle progression, cyclin gene expression, and their inhibition by simvastatin were determined. The serum-induced proliferation rate of human atrial fibroblasts was more than threefold greater than ventricular fibroblasts with faster DNA synthesis and higher mRNA levels of cyclin genes. Simvastatin predominantly decreased the rate of proliferation of atrial fibroblasts, with inhibition of cell cycle progression and an increase in the G0/G1 phase in atrial fibroblasts with a higher sensitivity toward inhibition compared with ventricular fibroblasts. The DNA synthesis and mRNA levels of cyclin A, D, and E were significantly reduced by simvastatin in atrial but not in ventricular fibroblasts. The inhibitory effect of simvastatin on atrial fibroblasts was abrogated by mevalonic acid (500 µM) that bypasses 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibition. Chamber-specific differences exist in the human heart because atrial fibroblasts have a higher proliferative capacity and are more sensitive to simvastatin-mediated inhibition through HMG-CoA reductase pathway. This mechanism may be useful in selectively preventing excessive atrial fibrosis without inhibiting adaptive ventricular remodeling during cardiac injury.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Simvastatina/farmacología , Acilcoenzima A/metabolismo , Células Cultivadas , Ciclinas/metabolismo , Fibroblastos/metabolismo , Fase G1/efectos de los fármacos , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/metabolismo , Atrios Cardíacos/fisiopatología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Ácido Mevalónico/farmacología , ARN Mensajero/metabolismo , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA