Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Transplant ; 16(3): 773-82, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26780159

RESUMEN

The resuscitation of hearts donated after circulatory death (DCD) is gaining widespread interest; however, the method of initial reperfusion (IR) that optimizes functional recovery has not been elucidated. We sought to determine the impact of IR temperature on the recovery of myocardial function during ex vivo heart perfusion (EVHP). Eighteen pigs were anesthetized, mechanical ventilation was discontinued, and cardiac arrest ensued. A 15-min standoff period was observed and then hearts were reperfused for 3 min at three different temperatures (5°C; N = 6, 25°C; N = 5, and 35°C; N = 7) with a normokalemic adenosine-lidocaine crystalloid cardioplegia. Hearts then underwent normothermic EVHP for 6 h during which time myocardial function was assessed in a working mode. We found that IR coronary blood flow differed among treatment groups (5°C = 483 ± 53, 25°C = 722 ± 60, 35°C = 906 ± 36 mL/min, p < 0.01). During subsequent EVHP, less myocardial injury (troponin I: 5°C = 91 ± 6, 25°C = 64 ± 16, 35°C = 57 ± 7 pg/mL/g, p = 0.04) and greater preservation of endothelial cell integrity (electron microscopy injury score: 5°C = 3.2 ± 0.5, 25°C = 1.8 ± 0.2, 35°C = 1.7 ± 0.3, p = 0.01) were evident in hearts initially reperfused at warmer temperatures. IR under profoundly hypothermic conditions impaired the recovery of myocardial function (cardiac index: 5°C = 3.9 ± 0.8, 25°C = 6.2 ± 0.4, 35°C = 6.5 ± 0.6 mL/minute/g, p = 0.03) during EVHP. We conclude that the avoidance of profound hypothermia during IR minimizes injury and improves the functional recovery of DCD hearts.


Asunto(s)
Corazón/fisiología , Hipotermia/prevención & control , Isquemia Miocárdica/terapia , Reperfusión Miocárdica/métodos , Preservación de Órganos/métodos , Recuperación de la Función , Recolección de Tejidos y Órganos/métodos , Animales , Paro Cardíaco Inducido , Trasplante de Corazón , Porcinos
2.
Am J Transplant ; 16(3): 783-93, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26663659

RESUMEN

Hearts donated following circulatory death (DCD) may represent an additional source of organs for transplantation; however, the impact of donor extubation on the DCD heart has not been well characterized. We sought to describe the physiologic changes that occur following withdrawal of life-sustaining therapy (WLST) in a porcine model of DCD. Physiologic changes were monitored continuously for 20 min following WLST. Ventricular pressure, volume, and function were recorded using a conductance catheter placed into the right (N = 8) and left (N = 8) ventricles, and using magnetic resonance imaging (MRI, N = 3). Hypoxic pulmonary vasoconstriction occurred following WLST, and was associated with distension of the right ventricle (RV) and reduced cardiac output. A 120-fold increase in epinephrine was subsequently observed that produced a transient hyperdynamic phase; however, progressive RV distension developed during this time. Circulatory arrest occurred 7.6±0.3 min following WLST, at which time MRI demonstrated an 18±7% increase in RV volume and a 12±9% decrease in left ventricular volume compared to baseline. We conclude that hypoxic pulmonary vasoconstriction and a profound catecholamine surge occur following WLST that result in distension of the RV. These changes have important implications on the resuscitation, preservation, and evaluation of DCD hearts prior to transplantation.


Asunto(s)
Paro Cardíaco , Trasplante de Corazón , Ventrículos Cardíacos/patología , Corazón/fisiopatología , Respiración Artificial/efectos adversos , Vasoconstricción , Animales , Modelos Animales , Porcinos , Donantes de Tejidos , Supervivencia Tisular
3.
Cell Death Dis ; 6: e1696, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25789971

RESUMEN

Transforming growth factor-ß(1) (TGF-ß(1)) is an important regulator of fibrogenesis in heart disease. In many other cellular systems, TGF-ß(1) may also induce autophagy, but a link between its fibrogenic and autophagic effects is unknown. Thus we tested whether or not TGF-ß(1)-induced autophagy has a regulatory function on fibrosis in human atrial myofibroblasts (hATMyofbs). Primary hATMyofbs were treated with TGF-ß(1) to assess for fibrogenic and autophagic responses. Using immunoblotting, immunofluorescence and transmission electron microscopic analyses, we found that TGF-ß(1) promoted collagen type Iα2 and fibronectin synthesis in hATMyofbs and that this was paralleled by an increase in autophagic activation in these cells. Pharmacological inhibition of autophagy by bafilomycin-A1 and 3-methyladenine decreased the fibrotic response in hATMyofb cells. ATG7 knockdown in hATMyofbs and ATG5 knockout (mouse embryonic fibroblast) fibroblasts decreased the fibrotic effect of TGF-ß(1) in experimental versus control cells. Furthermore, using a coronary artery ligation model of myocardial infarction in rats, we observed increases in the levels of protein markers of fibrosis, autophagy and Smad2 phosphorylation in whole scar tissue lysates. Immunohistochemistry for LC3ß indicated the localization of punctate LC3ß with vimentin (a mesenchymal-derived cell marker), ED-A fibronectin and phosphorylated Smad2. These results support the hypothesis that TGF-ß(1)-induced autophagy is required for the fibrogenic response in hATMyofbs.


Asunto(s)
Autofagia/genética , Fibrosis/genética , Atrios Cardíacos/metabolismo , Miofibroblastos/metabolismo , Factor de Crecimiento Transformador beta1/biosíntesis , Adenina/administración & dosificación , Adenina/análogos & derivados , Animales , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia , Proteína 7 Relacionada con la Autofagia , Proliferación Celular/efectos de los fármacos , Colágeno Tipo I/metabolismo , Fibronectinas/biosíntesis , Fibrosis/patología , Atrios Cardíacos/patología , Humanos , Macrólidos/administración & dosificación , Ratones , Proteínas Asociadas a Microtúbulos/genética , Miofibroblastos/patología , Cultivo Primario de Células , Ratas , Transducción de Señal/efectos de los fármacos , Proteína Smad2/biosíntesis , Proteína Smad2/genética , Factor de Crecimiento Transformador beta1/genética
4.
Curr Mol Med ; 14(5): 616-29, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24894175

RESUMEN

Survival of myocytes and mesenchymal cells in the heart is tightly regulated by a number of adaptive processes that are invoked with the changes that occur within the parenchyma and stroma. Autophagy is implicated in cellular housekeeping duties and maintenance of the integrity of the intracellular milieu by removal of protein aggregates and damaged organelles, whereas under pathophysiological conditions, the chronic up-regulation of autophagy may lead to significant disturbance of homeostatic conditions. Nonetheless, the role of autophagy in heart disease in the context of cardiac ischemia-reperfusion injury is currently unclear. This review will focus upon the role of autophagy as it pertains to ischemia reperfusion damage in the heart.


Asunto(s)
Autofagia/fisiología , Cardiopatías/metabolismo , Cardiopatías/patología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Humanos , Modelos Biológicos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología
5.
Cell Death Dis ; 3: e330, 2012 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-22717585

RESUMEN

3-hydroxy-3-methyl-glutaryl-CoA reductase inhibitors (statins) are cholesterol-lowering drugs that exert other cellular effects and underlie their beneficial health effects, including those associated with myocardial remodeling. We recently demonstrated that statins induces apoptosis and autophagy in human lung mesenchymal cells. Here, we extend our knowledge showing that statins simultaneously induces activation of the apoptosis, autophagy and the unfolded protein response (UPR) in primary human atrial fibroblasts (hATF). Thus we tested the degree to which coordination exists between signaling from mitochondria, endoplasmic reticulum and lysosomes during response to simvastatin exposure. Pharmacologic blockade of the activation of ER-dependent cysteine-dependent aspartate-directed protease (caspase)-4 and lysosomal cathepsin-B and -L significantly decreased simvastatin-induced cell death. Simvastatin altered total abundance and the mitochondrial fraction of proapoptotic and antiapoptotic proteins, while c-Jun N-terminal kinase/stress-activated protein kinase mediated effects on B-cell lymphoma 2 expression. Chemical inhibition of autophagy flux with bafilomycin-A1 augmented simvastatin-induced caspase activation, UPR and cell death. In mouse embryonic fibroblasts that are deficient in autophagy protein 5 and refractory to autophagy induction, caspase-7 and UPR were hyper-induced upon treatment with simvastatin. These data demonstrate that mevalonate cascade inhibition-induced death of hATF manifests from a complex mechanism involving co-regulation of apoptosis, autophagy and UPR. Furthermore, autophagy has a crucial role in determining the extent of ER stress, UPR and permissiveness of hATF to cell death induced by statins.


Asunto(s)
Apoptosis , Autofagia , Muerte Celular , Estrés del Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Fibroblastos/efectos de los fármacos , Ácido Mevalónico/metabolismo , Miocardio/citología , Caspasa 7/metabolismo , Inhibidores de Caspasas/farmacología , Caspasas Iniciadoras/metabolismo , Células Cultivadas , Activación Enzimática , Fibroblastos/metabolismo , Atrios Cardíacos/citología , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Ácido Mevalónico/farmacología , Transducción de Señal , Simvastatina/farmacología , Respuesta de Proteína Desplegada/efectos de los fármacos
6.
Am J Physiol Heart Circ Physiol ; 288(6): H2931-9, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15653752

RESUMEN

Despite the important roles played by ventricular fibroblasts and myofibroblasts in the formation and maintenance of the extracellular matrix, neither the ionic basis for membrane potential nor the effect of modulating membrane potential on function has been analyzed in detail. In this study, whole cell patch-clamp experiments were done using ventricular fibroblasts and myofibroblasts. Time- and voltage-dependent outward K(+) currents were recorded at depolarized potentials, and an inwardly rectifying K(+) (Kir) current was recorded near the resting membrane potential (RMP) and at more hyperpolarized potentials. The apparent reversal potential of Kir currents shifted to more positive potentials as the external K(+) concentration ([K(+)](o)) was raised, and this Kir current was blocked by 100-300 muM Ba(2+). RT-PCR measurements showed that mRNA for Kir2.1 was expressed. Accordingly, we conclude that Kir current is a primary determinant of RMP in both fibroblasts and myofibroblasts. Changes in [K(+)](o) influenced fibroblast membrane potential as well as proliferation and contractile functions. Recordings made with a voltage-sensitive dye, DiBAC(3)(4), showed that 1.5 mM [K(+)](o) resulted in a hyperpolarization, whereas 20 mM [K(+)](o) produced a depolarization. Low [K(+)](o) (1.5 mM) enhanced myofibroblast number relative to control (5.4 mM [K(+)](o)). In contrast, 20 mM [K(+)](o) resulted in a significant reduction in myofibroblast number. In separate assays, 20 mM [K(+)](o) significantly enhanced contraction of collagen I gels seeded with myofibroblasts compared with control mechanical activity in 5.4 mM [K(+)](o). In combination, these results show that ventricular fibroblasts and myofibroblasts express a variety of K(+) channel alpha-subunits and demonstrate that Kir current can modulate RMP and alter essential physiological functions.


Asunto(s)
Fibroblastos/fisiología , Potenciales de la Membrana/fisiología , Contracción Miocárdica/fisiología , Canales de Potasio/fisiología , Función Ventricular , Animales , División Celular , Técnicas Electrofisiológicas Cardíacas/métodos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Células Musculares/fisiología , Contracción Miocárdica/efectos de los fármacos , Técnicas de Placa-Clamp , Potasio/farmacología , Canales de Potasio/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
J Cell Mol Med ; 8(1): 85-92, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15090263

RESUMEN

Post-myocardial infarction (MI) remodeling of cardiac myocytes and the myocardial interstitium results in alteration of gross ventricular geometry and ventricular dysfunction. To investigate the mechanisms of the remodeling process of the heart after large MI, the expression of various genes in viable left ventricle and infarct scar tissue were examined at 16 weeks post-MI. Steady-state expression of Na(+)-K+ ATPase alpha-1 and -2, phospholamban (PLB), alpha-myosin heavy chain (alpha-MHC), ryanodine receptor (Rya) and Ca2+ ATPase (Serca2) mRNAs were decreased in the infarct scar vs noninfarcted sham-operated controls (P < 0.05). On the other hand, Gialpha2 and beta-MHC mRNAs were upregulated (P < 0.05, respectively) in the infarct scar whereas Na(+)-K+ ATPase-beta, Na(+)-Ca2+ exchanger and Gs mRNAs were not altered vs control values. In viable left ventricle, the alpha-1 subunit of Na(+)-K+ ATPase, alpha-3, beta-isoforms, Rya, beta-MHC, Gialpha2, Gs and Na(+)-Ca2+ exchanger were significantly elevated while expression of the alpha-2 subunit of Na(+)-K+ ATPase, PLB and Serca2 were significantly decreased compared to controls. Expression of CK2alpha mRNA was elevated in noninfarcted heart (145 +/- 15%) and diminished in the infarct scar (66 +/- 13%) vs controls. Expression of beta-MHC mRNA was elevated in both viable and infarct scar tissues of experimental hearts (140 +/- 31% and 183 +/- 30% vs. controls, respectively). These results suggest that cardiac genes in the infarcted tissue and viable left ventricle following MI are differentially regulated.


Asunto(s)
Cicatriz/patología , Regulación de la Expresión Génica , Ventrículos Cardíacos/patología , Infarto del Miocardio/patología , Infarto del Miocardio/cirugía , Miocardio/patología , Animales , Northern Blotting , Quinasa de la Caseína II , ADN Complementario/metabolismo , Proteínas de Unión al GTP/metabolismo , Humanos , Hipertrofia Ventricular Izquierda , Masculino , Miocardio/metabolismo , Cadenas Pesadas de Miosina/biosíntesis , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Serina-Treonina Quinasas/biosíntesis , ARN/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Intercambiador de Sodio-Calcio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/biosíntesis , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA