Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 38(1): e23379, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38133921

RESUMEN

Dynamin-related protein 1 (Drp1) is a cytosolic GTPase protein that when activated translocates to the mitochondria, meditating mitochondrial fission and increasing reactive oxygen species (ROS) in cardiomyocytes. Drp1 has shown promise as a therapeutic target for reducing cardiac ischemia/reperfusion (IR) injury; however, the lack of specificity of some small molecule Drp1 inhibitors and the reliance on the use of Drp1 haploinsufficient hearts from older mice have left the role of Drp1 in IR in question. Here, we address these concerns using two approaches, using: (a) short-term (3 weeks), conditional, cardiomyocyte-specific, Drp1 knockout (KO) and (b) a novel, highly specific Drp1 GTPase inhibitor, Drpitor1a. Short-term Drp1 KO mice exhibited preserved exercise capacity and cardiac contractility, and their isolated cardiac mitochondria demonstrated increased mitochondrial complex 1 activity, respiratory coupling, and calcium retention capacity compared to controls. When exposed to IR injury in a Langendorff perfusion system, Drp1 KO hearts had preserved contractility, decreased reactive oxygen species (ROS), enhanced mitochondrial calcium capacity, and increased resistance to mitochondrial permeability transition pore (MPTP) opening. Pharmacological inhibition of Drp1 with Drpitor1a following ischemia, but before reperfusion, was as protective as Drp1 KO for cardiac function and mitochondrial calcium homeostasis. In contrast to the benefits of short-term Drp1 inhibition, prolonged Drp1 ablation (6 weeks) resulted in cardiomyopathy. Drp1 KO hearts were also associated with decreased ryanodine receptor 2 (RyR2) protein expression and pharmacological inhibition of the RyR2 receptor decreased ROS in post-IR hearts suggesting that changes in RyR2 may have a role in Drp1 KO mediated cardioprotection. We conclude that Drp1-mediated increases in myocardial ROS production and impairment of mitochondrial calcium handling are key mechanisms of IR injury. Short-term inhibition of Drp1 is a promising strategy to limit early myocardial IR injury which is relevant for the therapy of acute myocardial infarction, cardiac arrest, and heart transplantation.


Asunto(s)
Dinaminas , Infarto del Miocardio , Daño por Reperfusión Miocárdica , Animales , Ratones , Calcio/metabolismo , Dinaminas/metabolismo , Homeostasis , Mitocondrias Cardíacas/metabolismo , Dinámicas Mitocondriales , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
2.
Neurocrit Care ; 36(1): 61-70, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34268646

RESUMEN

BACKGROUND: Neurological injury following successful resuscitation from sudden cardiac arrest (CA) is common. The pathophysiological basis of this injury remains poorly understood, and treatment options are limited. Microglial activation and neuroinflammation are established contributors to many neuropathologies, such as Alzheimer disease and traumatic brain injury, but their potential role in post-CA injury has only recently been recognized. Here, we hypothesize that microglial activation that occurs following brief asystolic CA is associated with neurological injury and represents a potential therapeutic target. METHODS: Adult C57BL/6 male and female mice were randomly assigned to 12-min, KCl-induced asystolic CA, under anesthesia and ventilation, followed by successful cardiopulmonary resuscitation (n = 19) or sham intervention (n = 11). Neurological assessments of mice were performed using standardized neurological scoring, video motion tracking, and sensory/motor testing. Mice were killed at 72 h for histological studies; neuronal degeneration was assessed using Fluoro-Jade C staining. Microglial characteristics were assessed by immunohistochemistry using the marker of ionized calcium binding adaptor molecule 1, followed by ImageJ analyses for cell integrity density and skeletal analyses. RESULTS: Neurological injury in post-cardiopulmonary-resuscitation mice vs. sham mice was evident by poorer neurological scores (difference of 3.626 ± 0.4921, 95% confidence interval 2.618-4.634), sensory and motor functions (worsened by sixfold and sevenfold, respectively, compared with baseline), and locomotion (75% slower with a 76% decrease in total distance traveled). Post-CA brains demonstrated evidence of neurodegeneration and neuroinflammatory microglial activation. CONCLUSIONS: Extensive microglial activation and neurodegeneration in the CA1 region and the dentate gyrus of the hippocampus are evident following brief asystolic CA and are associated with severe neurological injury.


Asunto(s)
Reanimación Cardiopulmonar , Paro Cardíaco , Animales , Modelos Animales de Enfermedad , Femenino , Paro Cardíaco/complicaciones , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo
3.
Neurocrit Care ; 34(1): 64-72, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32358767

RESUMEN

BACKGROUND: Cardiac arrest (CA) patients who survived by cardiopulmonary resuscitation (CPR) can present different levels of neurological deficits ranging from minor cognitive impairments to persistent vegetative state and brain death. The pathophysiology of the resulting brain injury is poorly understood, and whether changes in post-CA brain metabolism contribute to the injury are unknown. Here we utilized [18F]fluorodeoxyglucose (FDG)-Positron emission tomography (PET) to study in vivo cerebral glucose metabolism 72 h following CA in a murine CA model. METHODS: Anesthetized and ventilated adult C57BL/6 mice underwent 12-min KCl-induced CA followed by CPR. Seventy-two hours following CA, surviving mice were intraperitoneally injected with [18F]FDG (~ 186 µCi/200 µL) and imaged on Molecubes preclinical micro-PET/computed tomography (CT) imaging systems after a 30-min awake uptake period. Brain [18F]FDG uptake was determined by the VivoQuant software on fused PET/CT images with the 3D brain atlas. Upon completion of Positron emission tomography (PET) imaging, remaining [18F]FDG radioactivity in the brain, heart, and liver was determined using a gamma counter. RESULTS: Global increases in brain [18F]FDG uptake in post-CA mice were observed compared to shams and controls. The median standardized uptake value of [18F]FDG for CA animals was 1.79 versus sham 1.25 (p < 0.05) and control animals 0.78 (p < 0.01). This increased uptake was consistent throughout the 60-min imaging period and across all brain regions reaching statistical significance in the midbrain, pons, and medulla. Biodistribution analyses of various key organs yielded similar observations that the median [18F]FDG uptake for brain was 7.04%ID/g tissue for CA mice versus 5.537%ID/g tissue for sham animals, p < 0.05). CONCLUSIONS: This study has successfully applied [18F]FDG-PET/CT to measure changes in brain metabolism in a murine model of asystolic CA. Our results demonstrate increased [18F]FDG uptake in the brain 72 h following CA, suggesting increased metabolic demand in the case of severe neurological injury. Further study is warranted to determine the etiology of these changes.


Asunto(s)
Fluorodesoxiglucosa F18 , Paro Cardíaco , Animales , Encéfalo/diagnóstico por imagen , Glucosa , Paro Cardíaco/diagnóstico por imagen , Humanos , Ratones , Ratones Endogámicos C57BL , Tomografía Computarizada por Tomografía de Emisión de Positrones , Tomografía de Emisión de Positrones , Radiofármacos , Distribución Tisular
4.
Crit Care Med ; 48(2): e133-e140, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31939812

RESUMEN

OBJECTIVES: Cardiogenic shock following cardiopulmonary resuscitation for sudden cardiac arrest is common, occurring even in the absence of acute coronary artery occlusion, and contributes to high rates of postcardiopulmonary resuscitation mortality. The pathophysiology of this shock is unclear, and effective therapies for improving clinical outcomes are lacking. DESIGN: Laboratory investigation. SETTING: University laboratory. SUBJECTS: C57BL/6 adult female mice. INTERVENTIONS: Anesthetized and ventilated adult female C57BL/6 wild-type mice underwent a 4, 8, 12, or 16-minute potassium chloride-induced cardiac arrest followed by 90 seconds of cardiopulmonary resuscitation. Mice were then blindly randomized to a single IV injection of vehicle (phosphate-buffered saline) or suppressor of site IQ electron leak, an inhibitor of superoxide production by complex I of the mitochondrial electron transport chain. Suppressor of site IQ electron leak and vehicle were administered during cardiopulmonary resuscitation. MEASUREMENTS AND MAIN RESULTS: Using a murine model of asystolic cardiac arrest, we discovered that duration of cardiac arrest prior to cardiopulmonary resuscitation determined postresuscitation success rates, degree of neurologic injury, and severity of myocardial dysfunction. Post-cardiopulmonary resuscitation cardiac dysfunction was not associated with myocardial necrosis, apoptosis, inflammation, or mitochondrial permeability transition pore opening. Furthermore, left ventricular function recovered within 72 hours of cardiopulmonary resuscitation, indicative of myocardial stunning. Postcardiopulmonary resuscitation, the myocardium exhibited increased reactive oxygen species and evidence of mitochondrial injury, specifically reperfusion-induced reactive oxygen species generation at electron transport chain complex I. Suppressor of site IQ electron leak, which inhibits complex I-dependent reactive oxygen species generation by suppression of site IQ electron leak, decreased myocardial reactive oxygen species generation and improved postcardiopulmonary resuscitation myocardial function, neurologic outcomes, and survival. CONCLUSIONS: The severity of cardiogenic shock following asystolic cardiac arrest is dependent on the length of cardiac arrest prior to cardiopulmonary resuscitation and is mediated by myocardial stunning resulting from mitochondrial electron transport chain complex I dysfunction. A novel pharmacologic agent targeting this mechanism, suppressor of site IQ electron leak, represents a potential, practical therapy for improving sudden cardiac arrest resuscitation outcomes.


Asunto(s)
Complejo I de Transporte de Electrón/antagonistas & inhibidores , Paro Cardíaco/terapia , Peróxido de Hidrógeno/antagonistas & inhibidores , Mitocondrias/efectos de los fármacos , Aturdimiento Miocárdico/prevención & control , Superóxidos/antagonistas & inhibidores , Animales , Reanimación Cardiopulmonar , Femenino , Paro Cardíaco/fisiopatología , Ratones , Ratones Endogámicos C57BL , Aturdimiento Miocárdico/fisiopatología , Distribución Aleatoria , Especies Reactivas de Oxígeno/metabolismo
5.
Am J Physiol Heart Circ Physiol ; 310(8): H962-72, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26873966

RESUMEN

Doxorubicin (Doxo) is a chemotherapeutic drug widely used to treat variety of cancers. One of the most serious side effects of Doxo is its dose-dependent and delayed toxicity to the heart. Doxo is known to induce cardiac mitochondrial damage. Recently, the mitochondrial sirtuin SIRT3 has been shown to protect mitochondria from oxidative stress. Here we show that overexpression of SIRT3 protects the heart from toxicity of Doxo by preventing the drug-induced mitochondrial DNA (mtDNA) damage. Doxo treatment caused depletion of Sirt3 levels both in primary cultures of cardiomyocytes and in mouse hearts, which led to massive acetylation of mitochondrial proteins. Doxo-induced toxicity to cardiomyocytes was associated with increased reactive oxygen species (ROS) production, mitochondrial fragmentation, and cell death. Overexpression of SIRT3 helped to attenuate Doxo-induced ROS levels and cardiomyocyte death. Sirt3 knockout (Sirt3.KO) mice could not endure the full dose of Doxo treatment, developed exacerbated cardiac hypertrophy, and died during the course of treatment, whereas Sirt3 transgenic (Sirt3.tg) mice were protected against Doxo-induced cardiotoxicity. Along with Sirt3, we also observed a concomitant decrease in levels of oxoguanine-DNA glycosylase-1 (OGG1), a major DNA glycosylase that hydrolyzes oxidized-guanine (8-oxo-dG) to guanine. Depletion of OGG1 levels was associated with increased mtDNA damage. Sirt3.KO mice and Doxo-treated mice showed increased 8-oxo-dG adducts in DNA and corresponding increase in mtDNA damage, whereas, 8-oxo-dG adducts and mtDNA damage were markedly reduced in Sirt3 overexpressing transgenic mice hearts. These results thus demonstrated that Sirt3 activation protects the heart from Doxo-induced cardiotoxicity by maintaining OGG1 levels and protecting mitochondria from DNA damage.


Asunto(s)
Cardiomiopatías/prevención & control , Daño del ADN , ADN Mitocondrial/metabolismo , Doxorrubicina , Mitocondrias Cardíacas/enzimología , Miocitos Cardíacos/enzimología , Sirtuina 3/metabolismo , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/enzimología , Cardiomegalia/genética , Cardiomegalia/patología , Cardiomiopatías/inducido químicamente , Cardiomiopatías/enzimología , Cardiomiopatías/genética , Cardiomiopatías/patología , Muerte Celular , Células Cultivadas , Aductos de ADN/metabolismo , ADN Glicosilasas/metabolismo , ADN Mitocondrial/genética , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Modelos Animales de Enfermedad , Femenino , Fibroblastos/enzimología , Fibroblastos/patología , Hidrólisis , Masculino , Ratones Noqueados , Mitocondrias Cardíacas/patología , Miocitos Cardíacos/patología , Estrés Oxidativo , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Sirtuina 3/deficiencia , Sirtuina 3/genética , Sirtuinas/metabolismo , Factores de Tiempo
6.
Crit Care Med ; 43(2): e38-47, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25599491

RESUMEN

OBJECTIVES: Survival following sudden cardiac arrest is poor despite advances in cardiopulmonary resuscitation and the use of therapeutic hypothermia. Dynamin-related protein 1, a regulator of mitochondrial fission, is an important determinant of reactive oxygen species generation, myocardial necrosis, and left ventricular function following ischemia/reperfusion injury, but its role in cardiac arrest is unknown. We hypothesized that dynamin-related protein 1 inhibition would improve survival, cardiac hemodynamics, and mitochondrial function in an in vivo model of cardiac arrest. DESIGN: Laboratory investigation. SETTING: University laboratory. INTERVENTIONS: Anesthetized and ventilated adult female C57BL/6 wild-type mice underwent an 8-minute KCl-induced cardiac arrest followed by 90 seconds of cardiopulmonary resuscitation. Mice were then blindly randomized to a single IV injection of Mdivi-1 (0.24 mg/kg), a small molecule dynamin-related protein 1 inhibitor or vehicle (dimethyl sulfoxide). MEASUREMENTS AND MAIN RESULTS: Following resuscitation from cardiac arrest, mitochondrial fission was evidenced by dynamin-related protein 1 translocation to the mitochondrial membrane and a decrease in mitochondrial size. Mitochondrial fission was associated with increased lactate and evidence of oxidative damage. Mdivi-1 administration during cardiopulmonary resuscitation inhibited dynamin-related protein 1 activation, preserved mitochondrial morphology, and decreased oxidative damage. Mdivi-1 also reduced the time to return of spontaneous circulation (116 ± 4 vs 143 ± 7 s; p < 0.001) during cardiopulmonary resuscitation and enhanced myocardial performance post-return of spontaneous circulation. These improvements were associated with significant increases in survival (65% vs 33%) and improved neurological scores up to 72 hours post cardiac arrest. CONCLUSIONS: Post-cardiac arrest inhibition of dynamin-related protein 1 improves time to return of spontaneous circulation and myocardial hemodynamics, resulting in improved survival and neurological outcomes in a murine model of cardiac arrest. Pharmacological targeting of mitochondrial fission may be a promising therapy for cardiac arrest.


Asunto(s)
Dinaminas/antagonistas & inhibidores , Paro Cardíaco/metabolismo , Dinámicas Mitocondriales/efectos de los fármacos , Dinámicas Mitocondriales/fisiología , Quinazolinonas/farmacología , Aconitato Hidratasa/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Distribución Aleatoria
7.
FASEB J ; 28(1): 316-26, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24076965

RESUMEN

Mitochondrial fission, regulated by dynamin-related protein-1 (Drp1), is a newly recognized determinant of mitochondrial function, but its contribution to left ventricular (LV) impairment following ischemia-reperfusion (IR) injury is unknown. We report that Drp1 activation during IR results in LV dysfunction and that Drp1 inhibition is beneficial. In both isolated neonatal murine cardiomyocytes and adult rat hearts (Langendorff preparation) mitochondrial fragmentation and swelling occurred within 30 min of IR. Drp1-S637 (serine 637) dephosphorylation resulted in Drp1 mitochondrial translocation and increased mitochondrial fission. The Drp1 inhibitor Mdivi-1 preserved mitochondrial morphology, reduced cytosolic calcium, and prevented cell death. Drp1 siRNA similarly preserved mitochondrial morphology. In Langendorff hearts, Mdivi-1 reduced mitochondrial reactive oxygen species, improved LV developed pressure (92±5 vs. 28±10 mmHg, P<0.001), and lowered LV end diastolic pressure (10±1 vs. 86±13 mmHg, P<0.001) following IR. Mdivi-1 was protective if administered prior to or following ischemia. Because Drp1-S637 dephosphorylation is calcineurin sensitive, we assessed the effects of a calcineurin inhibitor, FK506. FK506 treatment prior to IR prevented Drp1-S637 dephosphorylation and preserved cardiac function. Likewise, therapeutic hypothermia (30°C) inhibited Drp1-S637 dephosphorylation and preserved mitochondrial morphology and myocardial function. Drp1 inhibition is a novel strategy to improve myocardial function following IR.


Asunto(s)
Dinaminas/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Animales , Inhibidores de la Calcineurina , Células Cultivadas , Dinaminas/genética , Immunoblotting , Inmunosupresores/farmacología , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Dinámicas Mitocondriales/efectos de los fármacos , Dinámicas Mitocondriales/genética , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/genética , Quinazolinonas/farmacología , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tacrolimus/farmacología
8.
Circ Res ; 110(11): 1484-97, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22511751

RESUMEN

RATIONALE: Pulmonary arterial hypertension (PAH) is a lethal syndrome characterized by pulmonary vascular obstruction caused, in part, by pulmonary artery smooth muscle cell (PASMC) hyperproliferation. Mitochondrial fragmentation and normoxic activation of hypoxia-inducible factor-1α (HIF-1α) have been observed in PAH PASMCs; however, their relationship and relevance to the development of PAH are unknown. Dynamin-related protein-1 (DRP1) is a GTPase that, when activated by kinases that phosphorylate serine 616, causes mitochondrial fission. It is, however, unknown whether mitochondrial fission is a prerequisite for proliferation. OBJECTIVE: We hypothesize that DRP1 activation is responsible for increased mitochondrial fission in PAH PASMCs and that DRP1 inhibition may slow proliferation and have therapeutic potential. METHODS AND RESULTS: Experiments were conducted using human control and PAH lungs (n=5) and PASMCs in culture. Parallel experiments were performed in rat lung sections and PASMCs and in rodent PAH models induced by the HIF-1α activator, cobalt, chronic hypoxia, and monocrotaline. HIF-1α activation in human PAH leads to mitochondrial fission by cyclin B1/CDK1-dependent phosphorylation of DRP1 at serine 616. In normal PASMCs, HIF-1α activation by CoCl(2) or desferrioxamine causes DRP1-mediated fission. HIF-1α inhibition reduces DRP1 activation, prevents fission, and reduces PASMC proliferation. Both the DRP1 inhibitor Mdivi-1 and siDRP1 prevent mitotic fission and arrest PAH PASMCs at the G2/M interphase. Mdivi-1 is antiproliferative in human PAH PASMCs and in rodent models. Mdivi-1 improves exercise capacity, right ventricular function, and hemodynamics in experimental PAH. CONCLUSIONS: DRP-1-mediated mitotic fission is a cell-cycle checkpoint that can be therapeutically targeted in hyperproliferative disorders such as PAH.


Asunto(s)
Proliferación Celular , Dinaminas/metabolismo , GTP Fosfohidrolasas/metabolismo , Hipertensión Pulmonar/enzimología , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias Musculares/enzimología , Proteínas Mitocondriales/metabolismo , Mitosis , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Animales , Antihipertensivos/farmacología , Proteína Quinasa CDC2/metabolismo , Estudios de Casos y Controles , Puntos de Control del Ciclo Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cobalto , Ciclina B1/metabolismo , Modelos Animales de Enfermedad , Dinaminas/genética , Activación Enzimática , Hipertensión Pulmonar Primaria Familiar , GTP Fosfohidrolasas/genética , Terapia Genética/métodos , Glucólisis , Humanos , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/terapia , Hipoxia/complicaciones , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Proteínas Asociadas a Microtúbulos/genética , Mitocondrias Musculares/efectos de los fármacos , Mitocondrias Musculares/patología , Proteínas Mitocondriales/genética , Mitosis/efectos de los fármacos , Monocrotalina , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Fosforilación , Arteria Pulmonar/enzimología , Arteria Pulmonar/patología , Quinazolinonas/farmacología , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Serina , Factores de Tiempo , Transfección
9.
Am J Respir Crit Care Med ; 187(8): 865-78, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23449689

RESUMEN

RATIONALE: Pulmonary arterial hypertension (PAH) is a lethal, female-predominant, vascular disease. Pathologic changes in PA smooth muscle cells (PASMC) include excessive proliferation, apoptosis-resistance, and mitochondrial fragmentation. Activation of dynamin-related protein increases mitotic fission and promotes this proliferation-apoptosis imbalance. The contribution of decreased fusion and reduced mitofusin-2 (MFN2) expression to PAH is unknown. OBJECTIVES: We hypothesize that decreased MFN2 expression promotes mitochondrial fragmentation, increases proliferation, and impairs apoptosis. The role of MFN2's transcriptional coactivator, peroxisome proliferator-activated receptor γ coactivator 1-α (PGC1α), was assessed. MFN2 therapy was tested in PAH PASMC and in models of PAH. METHODS: Fusion and fission mediators were measured in lungs and PASMC from patients with PAH and female rats with monocrotaline or chronic hypoxia+Sugen-5416 (CH+SU) PAH. The effects of adenoviral mitofusin-2 (Ad-MFN2) overexpression were measured in vitro and in vivo. MEASUREMENTS AND MAIN RESULTS: In normal PASMC, siMFN2 reduced expression of MFN2 and PGC1α; conversely, siPGC1α reduced PGC1α and MFN2 expression. Both interventions caused mitochondrial fragmentation. siMFN2 increased proliferation. In rodent and human PAH PASMC, MFN2 and PGC1α were decreased and mitochondria were fragmented. Ad-MFN2 increased fusion, reduced proliferation, and increased apoptosis in human PAH and CH+SU. In CH+SU, Ad-MFN2 improved walking distance (381 ± 35 vs. 245 ± 39 m; P < 0.05); decreased pulmonary vascular resistance (0.18 ± 0.02 vs. 0.38 ± 0.14 mm Hg/ml/min; P < 0.05); and decreased PA medial thickness (14.5 ± 0.8 vs. 19 ± 1.7%; P < 0.05). Lung vascularity was increased by MFN2. CONCLUSIONS: Decreased expression of MFN2 and PGC1α contribute to mitochondrial fragmentation and a proliferation-apoptosis imbalance in human and experimental PAH. Augmenting MFN2 has therapeutic benefit in human and experimental PAH.


Asunto(s)
GTP Fosfohidrolasas/deficiencia , Proteínas de Choque Térmico/deficiencia , Hipertensión Pulmonar/fisiopatología , Dinámicas Mitocondriales/fisiología , Proteínas Mitocondriales/deficiencia , Factores de Transcripción/deficiencia , Animales , Apoptosis/fisiología , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Tolerancia al Ejercicio/efectos de los fármacos , Hipertensión Pulmonar Primaria Familiar , Femenino , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Pulmón/citología , Pulmón/patología , Proteínas de la Membrana/administración & dosificación , Proteínas de la Membrana/deficiencia , Dinámicas Mitocondriales/genética , Proteínas Mitocondriales/administración & dosificación , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/fisiología , Atrofia Óptica Autosómica Dominante/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Ratas , Ratas Sprague-Dawley
10.
Circulation ; 126(24): 2859-69, 2012 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-23124027

RESUMEN

BACKGROUND: The cause and consequences of impaired adrenergic signaling in right ventricular failure/hypertrophy (RVH) are poorly understood. We hypothesized that G protein-coupled receptor kinase-2 (GRK2)-mediated uncoupling of ß-adrenergic receptor signaling impairs inotropic reserve. The implications of right ventricular (RV) adrenergic remodeling for inotrope selection and the therapeutic benefit of interrupting Gßγ-GRK2 interaction, using gallein, were tested. METHODS AND RESULTS: Chamber-specificity and cellular localization of adrenergic remodeling were compared in rodent RVH associated with pulmonary arterial hypertension (PAH-RVH; SU5416+chronic-hypoxia or Monocrotaline) versus pulmonary artery banding-induced RVH (PAB-RVH). Results were corroborated in RV arrays from 10 PAH patients versus controls. Inotropic reserve was assessed in RV- and left ventricular-Langendorff models and in vivo. Gallein therapy (1.8 mg/kg/day ×2-weeks) was assessed. Despite similar RVH, cardiac output (58.3±4.9 versus 82.9±4.8 mL/min; P<0.001) and treadmill distance (41.5±11.6 versus 244.1±12.4 m; P<0.001) were lower in PAH-RVH versus PAB-RVH. In PAH-RVH versus PAB-RVH there was greater downregulation of ß1-, α1- and dopamine-1 receptors, more left ventricular involvement, and greater impairment of RV contractile reserve. RV GRK2 activity increased in parallel with a reduction in both adrenergic receptor expression and inotrope-stimulated cAMP levels (P<0.01). ß1-receptor downregulation also occurred in human PAH-RVH. Dobutamine was superior to dopamine as an RV inotrope, both ex vivo and in vivo. CONCLUSIONS: GRK2-mediated desensitization-downregulation of adrenergic and dopaminergic receptors impairs inotropic reserve in PAH-RVH. Acute inotropic support in RVH is best accomplished by dobutamine, reflecting its better coupling to adenylyl cyclase and the reliance of dopamine on dopamine-1-receptor signaling, which is impaired in RVH. Inhibiting Gßγ-GRK2 interactions has therapeutic benefit in RVH.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Hipertensión Pulmonar/tratamiento farmacológico , Hipertrofia Ventricular Derecha/tratamiento farmacológico , Receptores Adrenérgicos beta/metabolismo , Receptores de Dopamina D1/metabolismo , Xantenos/farmacología , Animales , Cardiotónicos/farmacología , Células Cultivadas , Dobutamina/farmacología , Dopamina/farmacología , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Femenino , Humanos , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Hipertrofia Ventricular Derecha/metabolismo , Hipertrofia Ventricular Derecha/patología , Masculino , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/fisiología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos beta/genética , Receptores de Dopamina D1/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
11.
Am J Respir Crit Care Med ; 183(8): 1080-91, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21148721

RESUMEN

RATIONALE: The etiology of hepatopulmonary syndrome (HPS), a common complication of cirrhosis, is unknown. Inflammation and macrophage accumulation occur in HPS; however, their importance is unclear. Common bile duct ligation (CBDL) creates an accepted model of HPS, allowing us to investigate the cause of HPS. OBJECTIVES: We hypothesized that macrophages are central to HPS and investigated the therapeutic potential of macrophage depletion. METHODS: Hemodynamics, alveolar-arterial gradient, vascular reactivity, and histology were assessed in CBDL versus sham rats (n = 21 per group). The effects of plasma on smooth muscle cell proliferation and endothelial tube formation were measured. Macrophage depletion was used to prevent (gadolinium) or regress (clodronate) HPS. CD68(+) macrophages and capillary density were measured in the lungs of patients with cirrhosis versus control patients (n = 10 per group). MEASUREMENTS AND MAIN RESULTS: CBDL increased cardiac output and alveolar-arterial gradient by causing capillary dilatation and arteriovenous malformations. Activated CD68(+)macrophages (nuclear factor-κB+) accumulated in HPS pulmonary arteries, drawn by elevated levels of plasma endotoxin and lung monocyte chemoattractant protein-1. These macrophages expressed inducible nitric oxide synthase, vascular endothelial growth factor, and platelet-derived growth factor. HPS plasma increased endothelial tube formation and pulmonary artery smooth muscle cell proliferation. Macrophage depletion prevented and reversed the histological and hemodynamic features of HPS. CBDL lungs demonstrated increased medial thickness and obstruction of small pulmonary arteries. Nitric oxide synthase inhibition unmasked exaggerated pulmonary vasoconstrictor responses in HPS. Patients with cirrhosis had increased pulmonary intravascular macrophage accumulation and capillary density. CONCLUSIONS: HPS results from intravascular accumulation of CD68(+)macrophages. An occult proliferative vasculopathy may explain the occasional transition to portopulmonary hypertension. Macrophage depletion may have therapeutic potential in HPS.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/inmunología , Síndrome Hepatopulmonar/inmunología , Macrófagos/inmunología , Animales , Antígenos CD/fisiología , Antígenos de Diferenciación Mielomonocítica/fisiología , Malformaciones Arteriovenosas/etiología , Malformaciones Arteriovenosas/fisiopatología , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Síndrome Hepatopulmonar/etiología , Humanos , Pulmón/irrigación sanguínea , Pulmón/citología , Pulmón/inmunología , Macrófagos/fisiología , Masculino , Músculo Liso Vascular/fisiopatología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/fisiología , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/fisiología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/fisiología
12.
Am J Physiol Lung Cell Mol Physiol ; 299(3): L401-12, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20581101

RESUMEN

High-frequency echocardiography and high-field-strength magnetic resonance imaging (MRI) are new noninvasive methods for quantifying pulmonary arterial hypertension (PAH) and right ventricular (RV) hypertrophy (RVH). We compared these noninvasive methods of assessing the pulmonary circulation to the gold standard, cardiac catheterization (micromanometer-tipped catheters), in rats with monocrotaline-induced PAH and normal controls. Closed-chest, Sprague-Dawley rats were anesthetized with inhaled isoflurane (25 monocrotaline, 6 age-matched controls). Noninvasive studies used 37.5-MHz ultrasound (Vevo 770; VisualSonics) or a 9.4-T MRI (Bruker BioSpin). Catheterization used a 1.4-F micromanometer-tipped Millar catheter and a thermodilution catheter to measure cardiac output (CO). We compared noninvasive measures of pulmonary artery (PA) pressure (PAP) using PA acceleration time (PAAT) and CO, using the geometric PA flow method and RV free wall (RVFW) thickness/mass with cardiac catheterization and/or autopsy. Blinded operators performed comparisons using each method within 2 days of another. In a subset of rats with monocrotaline PAH, weekly echocardiograms, catheterization, and autopsy data assessed disease progression. Heart rate was similar during all studies (>323 beats/min). PAAT shortened, and the PA flow envelope displayed systolic "notching," reflective of downstream vascular remodeling/stiffening, within 3 wk of monocrotaline. MRI and echocardiography measures of PAAT were highly correlated (r(2) = 0.87) and were inversely proportional to invasive mean PAP (r(2) = 0.72). Mean PAP by echocardiography was estimated as 58.7 - (1.21 x PAAT). Invasive and noninvasive CO measurement correlated well (r(2) >or= 0.75). Noninvasive measures of RVFW thickness/mass correlated well with postmortem measurements. We conclude that high-resolution echocardiography and MRI accurately determine CO, PAP, and RV thickness/mass, offering similar results as high-fidelity right heart catheterization and autopsy, and that PAAT accurately estimates PAP and permits serial monitoring of experimental PAH. These tools are useful for assessment of the rodent pulmonary circulation and RVH.


Asunto(s)
Cateterismo Cardíaco/métodos , Ecocardiografía/métodos , Hipertensión Pulmonar/diagnóstico , Imagen por Resonancia Magnética , Animales , Presión Sanguínea , Gasto Cardíaco , Cateterismo , Hemodinámica , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/diagnóstico , Masculino , Monocrotalina , Arteria Pulmonar/fisiopatología , Circulación Pulmonar , Ratas , Ratas Sprague-Dawley , Termodilución/instrumentación
13.
Artif Organs ; 34(1): E17-21, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20420595

RESUMEN

We evaluated the hemodynamics of tissue-engineered hybrid graft in vivo. The hybrid expanded polytetrafluoroethylene (ePTFE) scaffold was fabricated by coating the ePTFE graft with poly (lactide-co-glycolide) (PLGA) solution. This scaffold was turned into an engineered hybrid graft by culturing smooth muscle cells on its surface. Both the ePTFE (n = 6) and the engineered hybrid grafts (n = 8) were implanted in the carotid arteries of mongrel dogs. The length of intima in the engineered hybrid graft was greater than the ePTFE. The neoarterial thickness in the engineered hybrid group was greater, and the foreign body reaction was more severe. We compared the hemodynamics (diameter, flow rate, pulsatile index, mean velocity, shear stress, resistance index, and systolic/diastolic ratio) of the native arteries in the distal anastmosis. The shear rate in the engineered hybrid group was higher immediately after implantation, and the resistance index was lower, but there was no significant difference after 4 weeks. The engineered grafts demonstrated similar hemodynamics with the ePTFE grafts after 4 weeks implantation.


Asunto(s)
Prótesis Vascular , Hemodinámica , Ingeniería de Tejidos , Animales , Arterias Carótidas/patología , Arterias Carótidas/cirugía , Perros , Miocitos del Músculo Liso , Andamios del Tejido
14.
Artif Organs ; 33(8): 657-62, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19624584

RESUMEN

In this study, we developed a small pneumatic actuator that can be used as an extracorporeal biventricular assist device. It incorporated a bellows-transforming mechanism to generate blood-pumping pressure. The cylindrical unit is 88 +/- 0.1 mm high, has a diameter of 150 +/- 0.1 mm, and weighs 2.4 +/- 0.01 kg. In vitro, maximal outflow at the highest pumping rate (PR) exceeded 8 L/min when two 55 mL blood sacs were used under an afterload pressure of 100 mm Hg. At a pumping rate of 100 beats per minute (bpm), maximal hydraulic efficiency was 9.34% when the unit supported a single ventricle and 13.8% when it supported both ventricles. Moreover, pneumatic efficiencies of the actuator were 17.3% and 33.1% for LVAD and BVAD applications, respectively. The energy equivalent pressure was 62.78 approximately 208.10 mm Hg at a PR of 60 approximately 100 bpm, and the maximal value of dP/dt during systole was 1269 mm Hg/s at a PR of 60 bpm and 979 mm Hg/s at a PR of 100 bpm. When the unit was applied to 15 calves, it stably pumped 3 approximately 4 L/min of blood at 60 bpm, and no mechanical malfunction was experienced over 125 days of operation. We conclude that the presently developed pneumatic actuator can be utilized as an extracorporeal biventricular assist device.


Asunto(s)
Corazón Auxiliar , Animales , Bovinos , Diseño de Equipo
15.
PLoS One ; 12(9): e0185046, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28934276

RESUMEN

RATIONALE: Post-ischemic changes in cellular metabolism alter myocardial and neurological function. Pyruvate dehydrogenase (PDH), the limiting step in mitochondrial glucose oxidation, is inhibited by increased expression of PDH kinase (PDK) during ischemia/reperfusion injury. This results in decreased utilization of glucose to generate cellular ATP. Post-cardiac arrest (CA) hypothermia improves outcomes and alters metabolism, but its influence on PDH and PDK activity following CA are unknown. We hypothesized that therapeutic hypothermia (TH) following CA is associated with the inhibition of PDK activity and increased PDH activity. We further hypothesized that an inhibitor of PDK activity, dichloroacetate (DCA), would improve PDH activity and post-CA outcomes. METHODS AND RESULTS: Anesthetized and ventilated adult female C57BL/6 wild-type mice underwent a 12-minute KCl-induced CA followed by cardiopulmonary resuscitation. Compared to normothermic (37°C) CA controls, administering TH (30°C) improved overall survival (72-hour survival rate: 62.5% vs. 28.6%, P<0.001), post-resuscitation myocardial function (ejection fraction: 50.9±3.1% vs. 27.2±2.0%, P<0.001; aorta systolic pressure: 132.7±7.3 vs. 72.3±3.0 mmHg, P<0.001), and neurological scores at 72-hour post CA (9.5±1.3 vs. 5.4±1.3, P<0.05). In both heart and brain, CA increased lactate concentrations (1.9-fold and 3.1-fold increase, respectively, P<0.01), decreased PDH enzyme activity (24% and 50% reduction, respectively, P<0.01), and increased PDK protein expressions (1.2-fold and 1.9-fold, respectively, P<0.01). In contrast, post-CA treatment with TH normalized lactate concentrations (P<0.01 and P<0.05) and PDK expressions (P<0.001 and P<0.05), while increasing PDH activity (P<0.01 and P<0.01) in both the heart and brain. Additionally, treatment with DCA (0.2 mg/g body weight) 30 min prior to CA improved both myocardial hemodynamics 2 hours post-CA (aortic systolic pressure: 123±3 vs. 96±4 mmHg, P<0.001) and 72-hour survival rates (50% vs. 19%, P<0.05) in normothermic animals. CONCLUSIONS: Enhanced PDH activity in the setting of TH or DCA administration is associated with improved post-CA resuscitation outcomes. PDH is a promising therapeutic target for improving post-CA outcomes.


Asunto(s)
Ácido Dicloroacético/uso terapéutico , Paro Cardíaco/terapia , Hipotermia Inducida , Complejo Piruvato Deshidrogenasa/metabolismo , Animales , Reanimación Cardiopulmonar , Terapia Combinada , Femenino , Paro Cardíaco/enzimología , Paro Cardíaco/mortalidad , Hemodinámica , Ratones , Ratones Endogámicos C57BL , Tasa de Supervivencia
16.
Oncotarget ; 8(21): 34082-34098, 2017 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-28423723

RESUMEN

Doxorubicin is the chemotherapeutic drug of choice for a wide variety of cancers, and cardiotoxicity is one of the major side effects of doxorubicin treatment. One of the main cellular targets of doxorubicin in the heart is mitochondria. Mitochondrial sirtuin, SIRT3 has been shown to protect against doxorubicin-induced cardiotoxicity. We have recently identified honokiol (HKL) as an activator of SIRT3, which protects the heart from developing pressure overload hypertrophy. Here, we show that HKL-mediated activation of SIRT3 also protects the heart from doxorubicin-induced cardiac damage without compromising the tumor killing potential of doxorubicin. Doxorubicin-induced cardiotoxicity is associated with increased ROS production and consequent fragmentation of mitochondria and cell death. HKL-mediated activation of SIRT3 prevented Doxorubicin induced ROS production, mitochondrial damage and cell death in rat neonatal cardiomyocytes. HKL also promoted mitochondrial fusion. We also show that treatment with HKL blocked doxorubicin-induced cardiac toxicity in mice. This was associated with reduced mitochondrial DNA damage and improved mitochondrial function. Furthermore, treatments of mice, bearing prostrate tumor-xenografts, with HKL and doxorubicin showed inhibition of tumor growth with significantly reduced cardiac toxicity. Our results suggest that HKL-mediated activation of SIRT3 protects the heart from doxorubicin-induced cardiotoxicity and represents a potentially novel adjunct for chemotherapy treatments.


Asunto(s)
Compuestos de Bifenilo/administración & dosificación , Cardiomiopatías/prevención & control , Doxorrubicina/efectos adversos , Lignanos/administración & dosificación , Mitocondrias Cardíacas/efectos de los fármacos , Animales , Compuestos de Bifenilo/farmacología , Cardiomiopatías/inducido químicamente , Línea Celular Tumoral , Células Cultivadas , Modelos Animales de Enfermedad , Lignanos/farmacología , Ratones , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Sirtuina 3 , Regulación hacia Arriba
17.
ASAIO J ; 52(4): 386-90, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16883117

RESUMEN

Most patients needing implantation of a ventricular assist device (VAD) require repeated sternotomy; some after cardiac surgery, and others later for heart transplantation. The purpose of this study was to establish the right thoracotomy technique as an alternative for VAD implantation to reduce repeated sternotomy-related morbidity and mortality. We performed a right thoracotomy in animals, preclinical cadaver fitting tests, and a clinical case. A total of 20 various animals underwent right thoracotomy for implantation of bi-VAD (BVAD, n = 17) and left VAD (LVAD, n = 3). The right chest cavity was entered through the fourth intercostal space with partial resection of the fifth rib. There was no procedure-related morbidity or mortality, except for one calf with right anterior leg paralysis. Preclinical fitting tests were performed on 7 human cadavers to observe the anatomical feasibility of BVAD cannulation from the right side of the heart. In humans, the ascending aorta, interatrial groove, right atrium, and main pulmonary artery were identified as optimal cannula insertion sites for BVAD implantation. A patient with cardiogenic shock underwent a right thoracotomy for implantation of an external LVAD. Cardiac function recovered after 3 weeks, and the device was successfully explanted through a repeat right thoracotomy. In conclusion, a right thoracotomy can be an alternative method to the standard median sternotomy for patients who need repeated sternotomy because of previous cardiac surgery, transplantation at a later date, or those with mediastinal infections.


Asunto(s)
Corazón Auxiliar , Implantación de Prótesis/métodos , Toracotomía/métodos , Anciano , Animales , Aorta , Bovinos , Perros , Femenino , Atrios Cardíacos , Humanos , Masculino , Persona de Mediana Edad , Diseño de Prótesis , Ajuste de Prótesis , Arteria Pulmonar , Ovinos , Toracotomía/estadística & datos numéricos
18.
ASAIO J ; 51(5): 609-13, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16322726

RESUMEN

An extracorporeal life support system (ECLS) with a conventional membrane oxygenator requires a driving force for the blood to pass through hollow fiber membranes. We hypothesized that if a gravity-flow hollow fiber membrane oxygenator is installed in the circuit, the twin blood sacs of a pulsatile ECLS (the Twin-Pulse Life Support, T-PLS) can be placed downstream of the membrane oxygenator. This would increase pump output by doubling pulse rate at a given pumpsetting rate while maintaining effective pulsatility. The purpose of this study was to determine the optimal circuit configuration for T-PLS with respect to energy and pump output. Animals were randomly assigned to 2 groups in a total cardiopulmonary bypass model. In the serial group, a conventional membrane oxygenator was located between the twin blood sacs of the T-PLS. In the parallel group, the twin blood sacs were placed downstream of the gravity-flow membrane oxygenator. Energy equivalent pressure (EEP), surplus hemodynamic energy (SHE) and pump output were collected at the different pump-setting rates of 30, 40, and 50 beats per minute (BPM). At a given pump-setting rate the pulse rate doubled in the parallel group. Percent changes of mean arterial pressure to EEP were 13.0 +/- 1.7, 12.0 +/- 1.9, and 7.6 +/- 0.9% in the parallel group, while 22.5 +/- 2.4, 23.2 +/- 1.9, and 21.8 +/- 1.4 in the serial group at 30, 40, and 50 BPM of pump-setting rates. SHE at each pump setting rate was 20,131 +/- 1408, 21,739 +/- 2470, and 15,048 +/- 2108 erg/ cm3 in the parallel group, while 33,968 +/- 3001, 38,232 +/- 3281, 37,964 +/- 2693 erg/cm3 in the serial group. Pump output was higher in the parallel circuit at 40, and 50 BPM pump-setting rates (3.1 +/- 0.2, 3.7 +/- 0.2 L/min vs. 2.2 +/- 0.1 and 2.5 +/- 0.1 L/min, respectively, p =0.01). Either parallel or serial circuit configuration of T-PLS generates effective pulsatility. As for the pump out, the parallel circuit configuration provides higher flow than the serial circuit configuration by doubling the pulse rate at a given pump-setting rate.


Asunto(s)
Circulación Extracorporea/instrumentación , Flujo Pulsátil , Sus scrofa/fisiología , Animales , Animales Recién Nacidos , Presión Sanguínea , Electrocardiografía , Metabolismo Energético , Estudios de Evaluación como Asunto , Circulación Extracorporea/métodos , Gravitación , Hemodinámica , Oxigenadores de Membrana , Distribución Aleatoria
19.
ASAIO J ; 51(1): 30-6, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15745131

RESUMEN

This study was conducted to directly compare the effects of pulsatile and nonpulsatile blood flow in the extracorporeal circulation upon renal tissue perfusion by using a tissue perfusion measurement system. A total cardiopulmonary bypass circuit was constructed to accommodate twelve Yorkshire swine, weighing 20 approximately 30 kg. Animals were randomly assigned to group 1 (n = 6, nonpulsatile centrifugal pump) or group 2 (n = 6, pulsatile T-PLS pump). A tissue perfusion measurement probe (Q-Flow 500) was inserted into the renal parenchymal tissue, and the extracorporeal circulation was maintained for an hour at a pump flow rate of 2 L/min after aortic cross-clamping. Tissue perfusion flow in the kidney was measured before bypass and every 10 minutes after bypass. Renal tissue perfusion flow was substantially higher in the pulsatile group throughout bypass (ranging 48.5-64.1 ml/min/100 g in group 1 vs. 51.0-88.1 ml/min/100 g in group 2). The intergroup difference was significant at 30 minutes (47.5 +/- 18.3 ml/min/100 g in group 1 vs. 83.4 +/- 28.5 ml/min/100 g in group 2; p = 0.026). Pulsatile flow achieves higher levels of tissue perfusion of the kidney during short-term extracorporeal circulation. A further study is required to observe the effects of pulsatile flow upon other vital organs and its long-term significance.


Asunto(s)
Puente Cardiopulmonar , Contrapulsación/métodos , Circulación Extracorporea , Riñón/irrigación sanguínea , Flujo Pulsátil/fisiología , Animales , Contrapulsación/instrumentación , Porcinos , Factores de Tiempo
20.
J Biosci Bioeng ; 118(4): 461-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24856052

RESUMEN

Hydrogel has been used for regenerating myocardial infraction (MI) as a delivery vehicle for cells and growth factors. This study showed that injectable hyaluronic acid (HA)-based hydrogels alone would effectively regenerate the damaged infarcted heart tissue. We found that there are two major factors of regeneration in MI. One is molecular weight of HA and another is the progression of MI; sub-acute and chronic. Rat MI model was prepared by ligating the left anterior descending coronary artery (LAD). Four weeks after injection of hydrogel, functional analysis of the heart and histological analysis was assessed. When different molecular weight HA-based hydrogels with 50 kDa, 130 kDa, and 170 kDa were applied to the infarcted area in the sub-acute model, 50 kDa HA-based hydrogel showed the most significant regeneration of myocardium as well as functional recovery among samples. For the disease progression, 50 kDa HA-based hydrogels were injected to sub-acute and chronic MI models. The regeneration activity was significantly decreased in the chronic models reflecting that injection timing of the therapeutic agents is also major determinants in the regeneration process. These results suggest that injection time and composition of hydrogel are two major points treating MI.


Asunto(s)
Materiales Biomiméticos/farmacología , Cardiotónicos/farmacología , Corazón/efectos de los fármacos , Ácido Hialurónico/farmacología , Infarto del Miocardio/tratamiento farmacológico , Regeneración/efectos de los fármacos , Enfermedad Aguda , Animales , Materiales Biomiméticos/química , Cardiotónicos/química , Enfermedad Crónica , Modelos Animales de Enfermedad , Corazón/fisiopatología , Ácido Hialurónico/química , Hidrogeles , Masculino , Peso Molecular , Infarto del Miocardio/fisiopatología , Ratas , Ratas Sprague-Dawley , Recuperación de la Función , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA