Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Proc Natl Acad Sci U S A ; 105(5): 1668-73, 2008 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-18216245

RESUMEN

Coronary artery disease is the most common cause of cardiac failure in the Western world, and to date there is no alternative to bypass surgery for severe coronary atherosclerosis. We report that c-kit-positive cardiac progenitor cells (CPCs) activated with insulin-like growth factor 1 and hepatocyte growth factor before their injection in proximity of the site of occlusion of the left coronary artery in rats, engrafted within the host myocardium forming temporary niches. Subsequently, CPCs divided and differentiated into endothelial cells and smooth muscle cells and, to a lesser extent, into cardiomyocytes. The acquisition of vascular lineages appeared to be mediated by the up-regulation of hypoxia-inducible factor 1alpha, which promoted the synthesis and secretion of stromal-derived factor 1 from hypoxic coronary vessels. Stromal-derived factor 1 was critical in the conversion of CPCs to the vascular fate. CPCs formed conductive and intermediate-sized coronary arteries together with resistance arterioles and capillaries. The new vessels were connected with the primary coronary circulation, and this increase in vascularization more than doubled myocardial blood flow in the infarcted myocardium. This beneficial effect, together with myocardial regeneration attenuated postinfarction dilated myopathy, reduced infarct size and improved function. In conclusion, locally delivered activated CPCs generate de novo coronary vasculature and may be implemented clinically for restoration of blood supply to the ischemic myocardium.


Asunto(s)
Vasos Coronarios/fisiología , Mioblastos Cardíacos/fisiología , Neovascularización Fisiológica , Regeneración , Células Madre/fisiología , Animales , Diferenciación Celular , Quimiocina CXCL12/metabolismo , Vasos Coronarios/citología , Células Endoteliales/citología , Femenino , Factor de Crecimiento de Hepatocito/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Mioblastos Cardíacos/efectos de los fármacos , Mioblastos Cardíacos/trasplante , Isquemia Miocárdica/metabolismo , Miocitos del Músculo Liso/citología , Proteínas Proto-Oncogénicas c-kit/análisis , Ratas , Ratas Endogámicas F344 , Trasplante de Células Madre , Células Madre/química , Células Madre/efectos de los fármacos
2.
Circulation ; 120(10): 876-87, 2009 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-19704095

RESUMEN

BACKGROUND: Cardiac progenitor cells (CPCs) possess the insulin-like growth factor-1 (IGF-1)-IGF-1 receptor system, and IGF-1 can be tethered to self-assembling peptide nanofibers (NF-IGF-1), leading to prolonged release of this growth factor to the myocardium. Therefore, we tested whether local injection of clonogenic CPCs and NF-IGF-1 potentiates the activation and differentiation of delivered and resident CPCs enhancing cardiac repair after infarction. METHODS AND RESULTS: Myocardial infarction was induced in rats, and untreated infarcts and infarcts treated with CPCs or NF-IGF-1 only and CPCs and NF-IGF-1 together were analyzed. With respect to infarcts exposed to CPCs or NF-IGF-1 alone, combination therapy resulted in a greater increase in the ratio of left ventricular mass to chamber volume and a better preservation of +dP/dt, -dP/dt, ejection fraction, and diastolic wall stress. Myocardial regeneration was detected in all treated infarcts, but the number of newly formed myocytes with combination therapy was 32% and 230% higher than with CPCs and NF-IGF-1, respectively. Corresponding differences in the volume of regenerated myocytes were 48% and 115%. Similarly, the length density of newly formed coronary arterioles with both CPCs and NF-IGF-1 was 73% and 83% greater than with CPCs and NF-IGF-1 alone, respectively. Importantly, activation of resident CPCs by paracrine effects contributed to cardiomyogenesis and vasculogenesis. Collectively, CPCs and NF-IGF-1 therapy reduced infarct size more than CPCs and NF-IGF-1 alone. CONCLUSIONS: The addition of nanofiber-mediated IGF-1 delivery to CPC therapy improved in part the recovery of myocardial structure and function after infarction.


Asunto(s)
Biotina , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/cirugía , Miocitos Cardíacos , Nanoestructuras , Regeneración , Trasplante de Células Madre , Células Madre , Adaptación Fisiológica , Animales , Apoptosis , Fusión Celular , Proliferación Celular , Células Cultivadas , Vasos Coronarios/fisiopatología , Femenino , Infarto del Miocardio/patología , Miocardio/patología , Miocitos Cardíacos/patología , Ratas , Ratas Endogámicas F344 , Regeneración/efectos de los fármacos , Supervivencia Tisular , Función Ventricular
3.
Circ Res ; 103(1): 107-16, 2008 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-18556576

RESUMEN

Ischemic heart disease is characterized chronically by a healed infarct, foci of myocardial scarring, cavitary dilation, and impaired ventricular performance. These alterations can only be reversed by replacement of scarred tissue with functionally competent myocardium. We tested whether cardiac progenitor cells (CPCs) implanted in proximity of healed infarcts or resident CPCs stimulated locally by hepatocyte growth factor and insulin-like growth factor-1 invade the scarred myocardium and generate myocytes and coronary vessels improving the hemodynamics of the infarcted heart. Hepatocyte growth factor is a powerful chemoattractant of CPCs, and insulin-like growth factor-1 promotes their proliferation and survival. Injection of CPCs or growth factors led to the replacement of approximately 42% of the scar with newly formed myocardium, attenuated ventricular dilation and prevented the chronic decline in function of the infarcted heart. Cardiac repair was mediated by the ability of CPCs to synthesize matrix metalloproteinases that degraded collagen proteins, forming tunnels within the fibrotic tissue during their migration across the scarred myocardium. New myocytes had a 2n karyotype and possessed 2 sex chromosomes, excluding cell fusion. Clinically, CPCs represent an ideal candidate cell for cardiac repair in patients with chronic heart failure. CPCs may be isolated from myocardial biopsies and, following their expansion in vitro, administered back to the same patients avoiding the adverse effects associated with the use of nonautologous cells. Alternatively, growth factors may be delivered locally to stimulate resident CPCs and promote myocardial regeneration. These forms of treatments could be repeated over time to reduce progressively tissue scarring and expand the working myocardium.


Asunto(s)
Cicatriz/terapia , Insuficiencia Cardíaca/terapia , Infarto del Miocardio/terapia , Miocardio , Trasplante de Células Madre , Células Madre , Animales , Movimiento Celular/efectos de los fármacos , Enfermedad Crónica , Cicatriz/etiología , Cicatriz/metabolismo , Cicatriz/patología , Colágeno/metabolismo , Colagenasas/biosíntesis , Diploidia , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Hemodinámica , Factor de Crecimiento de Hepatocito/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Infarto del Miocardio/complicaciones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas , Regeneración/efectos de los fármacos , Trasplante de Células Madre/métodos , Células Madre/metabolismo , Células Madre/patología , Trasplante Homólogo , Disfunción Ventricular/etiología , Disfunción Ventricular/metabolismo , Disfunción Ventricular/patología , Disfunción Ventricular/terapia
4.
Circ Res ; 102(5): 597-606, 2008 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-18202313

RESUMEN

Heart failure is the leading cause of death in the elderly, but whether this is the result of a primary aging myopathy dictated by depletion of the cardiac progenitor cell (CPC) pool is unknown. Similarly, whether current lifespan reflects the ineluctable genetic clock or heart failure interferes with the genetically determined fate of the organ and organism is an important question. We have identified that chronological age leads to telomeric shortening in CPCs, which by necessity generate a differentiated progeny that rapidly acquires the senescent phenotype conditioning organ aging. CPC aging is mediated by attenuation of the insulin-like growth factor-1/insulin-like growth factor-1 receptor and hepatocyte growth factor/c-Met systems, which do not counteract any longer the CPC renin-angiotensin system, resulting in cellular senescence, growth arrest, and apoptosis. However, pulse-chase 5-bromodeoxyuridine-labeling assay revealed that the senescent heart contains functionally competent CPCs that have the properties of stem cells. This subset of telomerase-competent CPCs have long telomeres and, following activation, migrate to the regions of damage, where they generate a population of young cardiomyocytes, reversing partly the aging myopathy. The senescent heart phenotype and heart failure are corrected to some extent, leading to prolongation of maximum lifespan.


Asunto(s)
Células Madre Adultas/efectos de los fármacos , Envejecimiento/efectos de los fármacos , Insuficiencia Cardíaca/terapia , Factor de Crecimiento de Hepatocito/uso terapéutico , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Miocitos Cardíacos/efectos de los fármacos , Células Madre Adultas/metabolismo , Células Madre Adultas/patología , Envejecimiento/patología , Animales , Antígenos de Diferenciación/biosíntesis , Apoptosis/efectos de los fármacos , Recuento de Células , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Modelos Animales de Enfermedad , Vías de Administración de Medicamentos , Corazón/efectos de los fármacos , Corazón/crecimiento & desarrollo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Masculino , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fenotipo , Ratas , Ratas Endogámicas F344 , Receptores de Factores de Crecimiento/metabolismo , Regeneración/efectos de los fármacos , Tasa de Supervivencia , Telómero/metabolismo
5.
Cardiovasc Res ; 79(4): 611-20, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18541523

RESUMEN

AIMS: We investigated the effect of an erythropoietin (EPO)-gelatin hydrogel drug delivery system (DDS) applied to the heart on myocardial infarct (MI) size, left ventricular (LV) remodelling and function. METHODS AND RESULTS: Experiments were performed in a rabbit model of MI. The infarct size was reduced, and LV remodelling and function were improved 14 days and 2 months after MI but not at 2 days after MI in the EPO-DDS group. The number of cluster of differentiation 31(CD31)-positive microvessels and the expression of erythropoietin receptor (EPO-R), phosphorylated-Akt (p-Akt), phosphorylated glycogen synthase kinase 3beta (p-GSK-3beta), phosphorylated extracellular signal-regulated protein kinase (p-ERK), phosphorylated signal transducer and activator of transcription 3 (p-Stat3), vascular endothelial growth factor (VEGF), and matrix metalloproteinase-1 (MMP-1) were significantly increased in the myocardium of the EPO-DDS group. CONCLUSION: Post-MI treatment with an EPO-DDS improves LV remodelling and function by activating prosurvival signalling, antifibrosis, and angiogenesis without causing any side effect.


Asunto(s)
Fármacos Cardiovasculares/farmacología , Eritropoyetina/farmacología , Gelatina/química , Hidrogeles , Infarto del Miocardio/tratamiento farmacológico , Miocardio/patología , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Animales , Western Blotting , Fármacos Cardiovasculares/administración & dosificación , Fármacos Cardiovasculares/química , Supervivencia Celular/efectos de los fármacos , Química Farmacéutica , Modelos Animales de Enfermedad , Formas de Dosificación , Portadores de Fármacos , Eritropoyetina/administración & dosificación , Eritropoyetina/química , Fibrosis , Humanos , Inmunohistoquímica , Inyecciones Subcutáneas , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/enzimología , Neovascularización Fisiológica/efectos de los fármacos , Conejos , Proteínas Recombinantes , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
6.
J Card Fail ; 13(10): 874-83, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18068622

RESUMEN

BACKGROUND: Hepatocyte growth factor (HGF) is reported to protect the heart against ischemia-reperfusion injury. However, whether in vivo adenovirus-mediated HGF gene transfer before ischemia is protective against ischemia-reperfusion and its precise mechanisms are still unknown. METHODS AND RESULTS: By using a rabbit model of ischemia-reperfusion injury, we demonstrate that HGF gene transfer is cardioprotective through its multiple beneficial actions, such as angiogenesis, Bcl-2 overexpression, and decreasing hydroxyl radicals, deoxyuride-5'-triphosphate biotin nick end labeling (TUNEL)-positive myocytes, and fibrotic area. After HGF gene transfer, the rabbits underwent 30 minutes of coronary occlusion and 30 minutes, 4 hours, 48 hours, and 14 days of reperfusion. The infarct size at 48 hours of reperfusion was significantly reduced in the HGF group (13.4% +/- 2.3%) compared with that in the LacZ group (36.5% +/- 2.0%) and saline group (40.3% +/- 3.2%). At 14 days of reperfusion, HGF gene transfer improved left ventricular ejection fraction and fractional shortening, reduced the fibrotic area, and increased the capillary density in the risk area. At 4 hours of reperfusion, Bcl-2 protein was overexpressed and the incidence of TUNEL-positive myocytes was significantly decreased in the risk area in the HGF group compared with the LacZ and saline groups. The myocardial interstitial 2,5-dihydroxybenzoic acid level, an indicator of hydroxyl radical, increased during 30 minutes of ischemia and 30 minutes of reperfusion in the LacZ and saline groups, and was significantly inhibited in the HGF group. CONCLUSION: HGF gene therapy may be a novel therapeutic strategy against unstable angina pectoris or severe angina pectoris, which may progress to acute myocardial infarction.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética/métodos , Factor de Crecimiento de Hepatocito/genética , Daño por Reperfusión Miocárdica/terapia , Animales , Apoptosis , Modelos Animales de Enfermedad , Factor de Crecimiento de Hepatocito/biosíntesis , Immunoblotting , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Masculino , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Miocardio/patología , Conejos , Resultado del Tratamiento
7.
Cardiovasc Res ; 71(3): 455-65, 2006 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16769043

RESUMEN

OBJECTIVE: Granulocyte-colony stimulating factor (G-CSF) accelerates repair following myocardial infarction (MI). Recently, the beneficial effects of post-MI administration of G-CSF were reported to be mediated by direct activation of the Jak-Stat pathway in cardiomyocytes. Our aim was to test the hypothesis that bone marrow-derived cells recruited into the infarcted myocardium are the primary mediators of the beneficial effects by G-CSF. METHODS AND RESULTS: MI was induced using a 30-min ischemia-reperfusion protocol (day 0) in 40 rabbits treated with G-CSF (10 microg/kg/day from days 3 to 7) or saline. Another 40 rabbits received the same G-CSF or saline protocol but also received AMD3100 (200 microg/kg/day), a specific inhibitor of CXCR4. On day 28 post-MI, left ventricular ejection fractions and end-diastolic dimensions were significantly better in the G-CSF group than in the control saline group, and the scar area/left ventricular wall area ratio was significantly smaller in the G-CSF group. G-CSF administration also led to increased mobilization of CXCR4+ bone marrow cells, including RAM11+ macrophages, into infarcted areas. And within those areas there was significant upregulation of expression of stromal cell-derived factor (SDF)-1, a chemoattractant of circulating CXCR4+ cells, as well as of the collagenase matrix metalloproteinase-1. AMD3100 significantly inhibited all of these beneficial effects of G-CSF, but did not affect the upregulation of SDF-1 or phospho-Stat3. CONCLUSION: Recruitment of CXCR4+ cells into infarcted myocardial tissues via stimulation of the CXCR4/SDF-1 axis plays a critical role in the beneficial effects of G-CSF.


Asunto(s)
Células de la Médula Ósea/patología , Factor Estimulante de Colonias de Granulocitos/farmacología , Infarto del Miocardio/patología , Receptores CXCR4/análisis , Animales , Bencilaminas , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Ciclamas , Factor Estimulante de Colonias de Granulocitos/antagonistas & inhibidores , Hemodinámica/efectos de los fármacos , Compuestos Heterocíclicos/farmacología , Recuento de Leucocitos , Masculino , Metaloproteinasa 1 de la Matriz/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Conejos , Receptores CXCR4/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Cardiovasc Res ; 69(2): 476-90, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16368087

RESUMEN

OBJECTIVE: Whether bone marrow cells injected following acute myocardial infarction (MI) transdifferentiate into cardiomyocytes remains controversial, and how these cells affect repair-related cytokines is not known. METHODS: Autologous bone marrow-derived mononuclear cells (BM-MNCs) labeled with DiI, 1,1'-dioctadecyl-1 to 3,3,3',3'-tetramethylindocarbocyanine perchlorate, or saline were intravenously injected into rabbits 5 h following a 30-min ischemia and reperfusion protocol, and cardiac function and the general pathology of the infarcted heart were followed up 1 and 3 months post-MI. To search for regenerated myocardium, electron microscopy as well as confocal microscopy were performed in the infarcted myocardium 7 days post-MI. Expression levels of repair-related cytokines were evaluated by immunohistochemistry and Western blotting. RESULTS: Improvements in cardiac function and reductions in infarct size were observed in the BM-MNC group 1 month and 3 months post-MI. Using electron microscopy 7 days after infarction, clusters of very immature (fetal) and relatively mature cardiomyocytes undergoing differentiation were identified in the infarcted anterior LV wall in the BM-MNC group, though their numbers were small. These cells contained many small and dense DiI particles (a BM-MNC marker), indicating that cardiomyocytes had regenerated from the injected BM-MNCs. The expression of both transforming growth factor-beta, which stimulates collagen synthesis and matrix metalloproteinase-1, a collagenase, were both down-regulated 7 days and 1 month post-MI in the BM-MNC group. Stromal cell-derived factor-1, which is known to recruit BM-MNCs into target tissues, was overexpressed in the infarcted areas of BM-MNC hearts 7 days post-MI. CONCLUSIONS: Intravenous transplantation of BM-MNCs leads to the development of BM-MNC-derived myocyte-like cells and regulates the expression of repair-related cytokines that facilitate repair following myocardial infarction.


Asunto(s)
Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Citocinas/análisis , Isquemia Miocárdica/cirugía , Miocitos Cardíacos/patología , Regeneración , Animales , Western Blotting/métodos , Diferenciación Celular , Quimiocina CXCL12 , Quimiocinas CXC/análisis , Inmunohistoquímica/métodos , Metaloproteinasa 1 de la Matriz/análisis , Microscopía Confocal , Microscopía Electrónica , Isquemia Miocárdica/inmunología , Isquemia Miocárdica/patología , Miocitos Cardíacos/ultraestructura , Conejos , Factores de Tiempo , Factor de Crecimiento Transformador beta/análisis
9.
Circulation ; 109(21): 2572-80, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15123535

RESUMEN

BACKGROUND: We investigated whether the improvement of cardiac function and remodeling after myocardial infarction (MI) by granulocyte colony-stimulating factor (G-CSF) relates to acceleration of the healing process, in addition to myocardial regeneration. METHODS AND RESULTS: In a 30-minute coronary occlusion and reperfusion rabbit model, saline (S) or 10 microg x kg(-1) x d(-1) of human recombinant G-CSF (G) was injected subcutaneously from 1 to 5 days after MI. Smaller left ventricular (LV) dimension, increased LV ejection fraction, and thicker infarct-LV wall were seen in G at 3 months after MI. At 2, 7, and 14 days and 3 months after MI, necrotic tissue areas were 14.2+/-1.5/13.4+/-1.1, 0.4+/-0.1/1.8+/-0.5*, 0/0, and 0/0 mm2 x slice(-1) x kg(-1), granulation areas 0/0, 4.0+/-0.7/8.5+/-1.0*, 3.9+/-0.8/5.7+/-0.7,* and 0/0 mm2 x slice(-1) x kg(-1), and scar areas 0/0, 0/0, 0/0, and 4.2+/-0.5/7.9+/-0.9* mm2 x slice(-1) x kg(-1) in G and S, respectively (*P<0.05, G versus S). Clear increases of macrophages and of matrix metalloproteinases (MMP) 1 and 9 were seen in G at 7 days after MI. This suggests that G accelerates absorption of necrotic tissues via increase of macrophages and reduces granulation and scar tissues via expression of MMPs. Meanwhile, surviving myocardial tissue areas within the risk areas were significantly increased in G despite there being no difference in LV weight, LV wall area, or cardiomyocyte size between G and S. Confocal microscopy revealed significant increases of cardiomyocytes with positive 3,3,3',3'-tetramethylindocarbocyanine perchlorate and positive troponin I in G, suggesting enhanced myocardial regeneration by G. CONCLUSIONS: The acceleration of the healing process and myocardial regeneration may play an important role for the beneficial effect of post-MI G-CSF treatment.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Corazón/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Miocardio/patología , Remodelación Ventricular/efectos de los fármacos , Animales , Tamaño de la Célula/efectos de los fármacos , Cicatriz/etiología , Cicatriz/patología , Cicatriz/prevención & control , Evaluación Preclínica de Medicamentos , Ecocardiografía , Tejido de Granulación/patología , Factor Estimulante de Colonias de Granulocitos/farmacología , Macrófagos/fisiología , Metaloproteinasa 1 de la Matriz/análisis , Metaloproteinasa 9 de la Matriz/análisis , Microscopía Confocal , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/enzimología , Miocitos Cardíacos/efectos de los fármacos , Conejos , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Regeneración/efectos de los fármacos
10.
Hypertens Res ; 28(4): 361-8, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16138567

RESUMEN

Dihydropyridine Ca channel blockers are widely prescribed for the treatment of hypertension and coronary artery diseases, but it remains unknown whether these agents protect against arrhythmias. We investigated whether cilnidipine, an N+L-type Ca channel blocker, reduces the incidences of ventricular premature beats (VPBs) and, if so, via what mechanisms. Japanese white rabbits underwent 30 min of ischemia and 48 h of reperfusion. Cilnidipine (0.5 or 1.0 microg/kg/min, i.v.) or saline (i.v.) was administered from 30 min before ischemia to 30 min after reperfusion. Electrocardiogram and blood pressure were monitored and the incidences of VPBs were measured. At 48 h after reperfusion, myocardial infarct was measured. Myocardial interstitial noradrenaline levels were determined before, during and after 30 min of ischemia with cilnidipine (0.5 and 1.0 microg/kg/min) or saline. The incidences of VPBs during ischemia and reperfusion were significantly attenuated in the cilnidipine 0.5 group (15.6 +/- 3.1 and 6.8 +/- 1.9 beats/30 min) and in the cilnidipine 1.0 group (10.4 +/- 4.9 and 3.5 +/- 1.0 beats/30 min) compared to the control group (27.2 +/- 4.5 and 24.2 +/- 3.1 beats/30 min), respectively. Myocardial interstitial noradrenaline levels were significantly reduced in the cilnidipine 0.5 and 1.0 groups compared to the control group during ischemia and reperfusion. The antiarrhythmic effect of cilnidipine may be related to the attenuation of cardiac sympathetic nerve activity. This finding may provide new insight into therapeutic strategies for hypertensive patients with ventricular arrhythmias.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Dihidropiridinas/farmacología , Infarto del Miocardio/tratamiento farmacológico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Fibrilación Ventricular/prevención & control , Animales , Presión Sanguínea , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo N/metabolismo , Modelos Animales de Enfermedad , Frecuencia Cardíaca , Incidencia , Masculino , Infarto del Miocardio/mortalidad , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/mortalidad , Daño por Reperfusión Miocárdica/patología , Norepinefrina/metabolismo , Conejos , Tasa de Supervivencia , Sistema Nervioso Simpático/metabolismo , Taquicardia Ventricular/epidemiología , Taquicardia Ventricular/prevención & control , Fibrilación Ventricular/epidemiología , Complejos Prematuros Ventriculares/epidemiología , Complejos Prematuros Ventriculares/prevención & control
11.
Circ J ; 71(4): 574-9, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17384462

RESUMEN

BACKGROUND: It is still unclear whether celiprolol, a beta(1)-selective blocker, reduces myocardial infarct size. This study will examine whether celiprolol reduces myocardial infarct size, as well as investigate the mechanisms for its infarct size-reducing effect in rabbits. METHODS AND RESULTS: Japanese white rabbits underwent 30 min of ischemia and 48 h of reperfusion. Celiprolol (1 or 10 mg x kg (-1) x h(-1) for 60 min, iv) was administered 20 min before ischemia with or without pretreatment with N(omega)-nitro-L-arginine methylester (L-NAME, 10 mg/kg, iv, a nitric oxide synthase inhibitor) or 5-hydroxydecanoic acid sodium salt (5-HD, 5 mg/kg, iv, a mitochondrial K(ATP) channel blocker). The area at risk as a percentage of the left ventricle was determined by using Evans blue dye, and the infarct size was determined as a percentage of the area at risk by triphenyl tetrazolium chloride staining. Celiprolol 1 and 10 mg x kg(-1) x h(-1) significantly reduced the infarct size in a dose-dependent manner (36.4+/-1.7%, n=7 and 25.4+/-2.9%, n=7, respectively) compared with the control (46.2+/-3.1%, n=8). The infarct size-reducing effect of celiprolol was completely blocked by L-NAME (40.4 +/-2.8%, n=8) but not by 5-HD (27.3+/-1.0%, n=8). Celiprolol 1 mg x kg(-1) x h (-1) increased the myocardial interstitial levels of NOx, an indicator of nitric oxide, and reduced the intensity of dihydro-ethidium staining of myocardium, an indicator of superoxide, during reperfusion after 30 min of ischemia. CONCLUSION: Celiprolol reduces myocardial infarct size and also increases nitric oxide production and reduces superoxide levels but not mitochondrial K(ATP) channels in rabbits.


Asunto(s)
Antagonistas Adrenérgicos beta/uso terapéutico , Celiprolol/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Óxido Nítrico/metabolismo , Animales , Presión Sanguínea/fisiología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Frecuencia Cardíaca/fisiología , Masculino , Mitocondrias Cardíacas/fisiología , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Canales de Potasio/fisiología , Conejos , Superóxidos/metabolismo
12.
Circ J ; 71(4): 580-90, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17384463

RESUMEN

BACKGROUND: The purpose of the present study was to investigate the effect of granulocyte colony-stimulating factor (G-CSF) in combination with myelo-suppressives on post-myocardial infarction (MI) myocardial repair. METHODS AND RESULTS: Twenty-four hours after 30-min ischemia and reperfusion (day 0), rabbits were assigned to 4 treatment groups: myelo-suppressives (M group), G-CSF (G group), the 2 in combination (MG group) or saline (S group). Significantly greater numbers of circulating stem cells were seen in the MG group than in the G group, with attenuated leukocytosis. In addition, MG caused the greatest upregulation of stromal cell-derived factor (SDF)-1 within the infarcted myocardium and thus recruitment of stem cells from the circulation into the infarcted tissue. This led to enhanced myocardial repair, as indicated by the numbers of bone marrow cell-derived cardiomyocytes and endothelial cells, reduction in scar tissue, improvement in cardiac function and reduction in left ventricular remodeling during the chronic phase of MI. These beneficial effects were entirely abolished by the administration of a CXCR4 antagonist AMD3100, which indicates the importance of CXCR4/SDF-1-axis as a mechanism underlying myocardial repair. CONCLUSION: The combination of G-CSF and myelo-suppressives may be a useful new therapy that overcomes the insufficiency seen with G-CSF alone.


Asunto(s)
Ciclofosfamida/uso terapéutico , Fluorouracilo/uso terapéutico , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Inmunosupresores/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Bencilaminas , Recuento de Células Sanguíneas , Quimiocina CXCL12 , Quimiocinas CXC/fisiología , Ciclamas , Quimioterapia Combinada , Ecocardiografía , Células Endoteliales/patología , Compuestos Heterocíclicos/farmacología , Masculino , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , Conejos , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/fisiología , Remodelación Ventricular/efectos de los fármacos
13.
Circ J ; 71(10): 1622-8, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17895562

RESUMEN

BACKGROUND: Statins reportedly protect against myocardial infarction, but the precise mechanism is unclear. METHODS AND RESULTS: Rabbits underwent 30 min of coronary occlusion followed by 48 h of reperfusion. Pravastatin (1 or 5 mg/kg) or saline was intravenously administered 10 min before ischemia. Pravastatin (5 mg/kg) was also administered 10 min before reperfusion. N(omega)-nitro-L-arginine methylester (L-NAME, 10 mg/kg), chelerythrine (5 mg/kg) or 5-hydroxydecanoic acid sodium salt (5-HD, 5 mg/kg) was intravenously administered 10 min before pravastatin injection. The infarct size was determined. The myocardial interstitial levels of 2,5-dihydroxybenzoic acid (DHBA) and nitrogen oxide (NOx), and the intensity of myocardial dihydroethidium staining were measured. Pre-ischemic treatment with pravastatin reduced the infarct size (34+/-5% and 24+/-4%, 1 and 5 mg/kg, respectively), but not pre-reperfusion treatment (42.1+/-3.7%), compared with the control (45+/-3%). This effect was blocked by L-NAME (42.6+/-4%), chelerythrine (50.9+/-3%) and 5-HD (52.7+/-2%). Pre-ischemic treatment with pravastatin increased myocardial NOx levels, and attenuated both the 2,5-DHBA level and the intensity of dihydroethidium staining during reperfusion. Chelerythrine abolished the increase in NOx levels by pravastatin. CONCLUSION: Pre-ischemic treatment with pravastatin reduces the myocardial infarct size via protein kinase C-dependent nitric oxide production, decreasing hydroxyl radicals and superoxide, and opening the mitochondrial adenosine triphosphate-sensitive potassium channels.


Asunto(s)
Canales KATP/metabolismo , Mitocondrias Cardíacas/metabolismo , Infarto del Miocardio/prevención & control , Óxido Nítrico/metabolismo , Pravastatina/farmacología , Proteína Quinasa C/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Frecuencia Cardíaca/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Precondicionamiento Isquémico Miocárdico/métodos , Masculino , Infarto del Miocardio/patología , Conejos , Superóxidos/metabolismo
14.
Nat Med ; 13(12): 1467-75, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18037896

RESUMEN

The serine-threonine kinases Pim-1 and Akt regulate cellular proliferation and survival. Although Akt is known to be a crucial signaling protein in the myocardium, the role of Pim-1 has been overlooked. Pim-1 expression in the myocardium of mice decreased during postnatal development, re-emerged after acute pathological injury in mice and was increased in failing hearts of both mice and humans. Cardioprotective stimuli associated with Akt activation induced Pim-1 expression, but compensatory increases in Akt abundance and phosphorylation after pathological injury by infarction or pressure overload did not protect the myocardium in Pim-1-deficient mice. Transgenic expression of Pim-1 in the myocardium protected mice from infarction injury, and Pim-1 expression inhibited cardiomyocyte apoptosis with concomitant increases in Bcl-2 and Bcl-X(L) protein levels, as well as in Bad phosphorylation levels. Relative to nontransgenic controls, calcium dynamics were significantly enhanced in Pim-1-overexpressing transgenic hearts, associated with increased expression of SERCA2a, and were depressed in Pim-1-deficient hearts. Collectively, these data suggest that Pim-1 is a crucial facet of cardioprotection downstream of Akt.


Asunto(s)
Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/fisiología , Animales , Apoptosis , Núcleo Celular/metabolismo , Humanos , Ratones , Ratones Noqueados , Miocardio/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/biosíntesis , Ratas , Proteína bcl-X/metabolismo
15.
Circ J ; 70(9): 1093-8, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16936417

RESUMEN

BACKGROUND: The purpose of this study was to determine whether treatment with granulocyte colony-stimulating factor (G-CSF), which mobilizes endothelial progenitor cells from bone marrow, can safely improve the clinical outcomes of patients with atherosclerotic peripheral artery disease (PAD). METHODS AND RESULTS: Thirty-nine patients with intractable PAD were randomly assigned to 3 groups: a negative control group (n=12) treated with conventional drug therapy; a positive control group (n=13) treated with conventional drug therapy plus bone marrow transplantation (BMT); and a G-CSF group (n=14) treated with conventional therapy plus subcutaneous injection of 2-5 microg/kg of recombinant human G-CSF once daily for 10 days. One month after treatment, subjective symptoms improved significantly in the G-CSF and BMT groups. Ankle-brachial pressure index and transcutaneous oxygen pressure increased significantly in the BMT and G-CSF groups, but no such improvements were seen in the group receiving conventional therapy alone. CONCLUSIONS: G-CSF improves the clinical signs and symptoms of patients with intractable PAD to the same degree as BMT does. This noninvasive treatment may thus represent a useful new approach to managing the disease.


Asunto(s)
Aterosclerosis/terapia , Trasplante de Médula Ósea , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Movilización de Célula Madre Hematopoyética , Anciano , Femenino , Movilización de Célula Madre Hematopoyética/métodos , Humanos , Masculino , Persona de Mediana Edad , Proteínas Recombinantes , Regeneración/efectos de los fármacos , Inducción de Remisión , Trasplante Autólogo
16.
Clin Exp Pharmacol Physiol ; 33(11): 1035-41, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17042911

RESUMEN

1. In the present study, we investigated the effect of 3-methyl-1-phenyl-2-pyrazolin-5-one (edaravone), a free radical scavenger, on myocardial infarct (MI) size and cardiac function in an in vivo model of MI in rabbits. We further investigated the contribution of hydroxyl radicals, superoxide and nitric oxide (NO) to its effects. 2. Anaesthetized open-chest Japanese white male rabbits were subjected to 30 min coronary occlusion and 48 h reperfusion. The control group (n = 10) was injected with saline 10 min before reperfusion. The edaravone group (n = 10) was injected with a bolus of 3 mg/kg edaravone 10 min before reperfusion. The edaravone + N(G)-nitro-L-arginine methyl ester (L-NAME) group (n = 5) was given 10 mg/kg, i.v., L-NAME 10 min before the administration of 3 mg/kg edaravone. The L-NAME group (n = 5) was given 10 mg/kg, i.v., L-NAME 20 min before reperfusion. Infarct size was measured using the triphenyl tetrazolium chloride method and is expressed as a percentage of area at risk. Cardiac function was assessed by echocardiography 14 days after infarction. 3. In another series of experiments, rabbits were subjected to 30 min coronary occlusion and 30 min reperfusion and myocardial interstitial 2,3-dihydroxybenzoic acid (DHBA) and 2,5-DHBA levels, indicators of hydroxyl radical, were measured using a microdialysis technique. 4. Infarct size in the edaravone group was significantly reduced compared with that in the control group (27.4 +/- 6.8 vs 43.4 +/- 6.8%, respectively; P < 0.05). The edaravone-induced reduction of infarct size was abolished by pretreatment with L-NAME. Myocardial interstitial levels of 2,3-DHBA and 2,5-DHBA increased 20 and 30 min after ischaemia and peaked at 10 min reperfusion in the control group. Edaravone significantly inhibited the increase in 2,3-DHBA and 2,5-DHBA levels seen during reperfusion. Dihydroethidium staining showing in situ detection of superoxide was less intense in ischaemic myocardium in the edaravone-treated group compared with the control group. Edaravone improved cardiac function and left ventricular remodelling 14 days after infarction. 5. In conclusion, edaravone significantly reduces MI size and improves cardiac function and LV remodelling by decreasing hydroxyl radicals and superoxide in the myocardium and increasing the production of NO during reperfusion in rabbits.


Asunto(s)
Antipirina/análogos & derivados , Presión Sanguínea/efectos de los fármacos , Depuradores de Radicales Libres/uso terapéutico , Frecuencia Cardíaca/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Remodelación Ventricular/efectos de los fármacos , Animales , Antipirina/uso terapéutico , Edaravona , Inhibidores Enzimáticos/uso terapéutico , Gentisatos/metabolismo , Hidroxibenzoatos/metabolismo , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , NG-Nitroarginina Metil Éster/uso terapéutico , Conejos , Superóxidos/metabolismo
17.
Circ J ; 70(4): 430-7, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16565560

RESUMEN

BACKGROUND: In animal models, granulocyte colony-stimulating factor (G-CSF) improves post-infarct cardiac function. However, in pilot studies involving patients with angina and acute myocardial infarction (AMI), G-CSF at a high dose frequently induced coronary occlusion or restenosis, but those at a low dose showed no significant beneficial effect. We hypothesized that a low dose but long duration of G-CSF will have a beneficial effect without serious complications to patients with coronary heart disease. METHODS AND RESULTS: Forty-six patients with angina or AMI were randomly assigned into G-CSF and non-G-CSF control groups, respectively. Recombinant G-CSF was subcutaneously injected once a day for 10 days. The leukocyte counts in the peripheral blood were controlled at approximately 30,000/microl. One month later, a Thallium-201 single photon emission computed tomography revealed the increased percentage uptake and the reduced extent and severity scores in the G-CSF angina group. In the G-CSF AMI group, the curve between the ejection fraction and peak creatine kinase shifted significantly upward, compared with that of the non-G-CSF AMI group. Serious complications were not observed during the 6 months of observation. CONCLUSIONS: A low dose but long duration of G-CSF treatment may have a beneficial effect without any serious complications in patients with coronary heart disease.


Asunto(s)
Angina de Pecho/tratamiento farmacológico , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Anciano , Angina de Pecho/fisiopatología , Antígenos CD34/sangre , Angiografía Coronaria , Vasos Coronarios/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Lenograstim , Recuento de Leucocitos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/fisiopatología , Proyectos Piloto , Tomografía de Emisión de Positrones , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Flujo Sanguíneo Regional/efectos de los fármacos , Factores de Tiempo , Función Ventricular Izquierda/fisiología
18.
Heart Vessels ; 21(3): 180-7, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16715193

RESUMEN

Recent studies suggest transplanted bone marrow cells (BMCs) can be used to reconstitute coronary vessels and myocardium following acute myocardial infarction, thereby improving cardiac function. We sought to investigate the therapeutic potential of BMC transplantation in the treatment of nonischemic cardiomyopathy. Experimental cardiomyopathy was produced by treating rabbits for 8 weeks with doxorubicin (2 mg/kg per week). Two weeks after the treatment was finished, freshly aspirated BMCs or an equivalent volume of phosphate-buffered saline was injected directly into the left ventricular free wall. Four weeks later, heart function was examined during perfusion on a Langendorff apparatus. Left ventricular developed pressure and +/-dp/dt were significantly better in the transplantation group, among which echocardiography also showed significantly better ejection fractions. In addition, left ventricular weights as a fraction of body weight and left ventricular wall thicknesses were both lower in rabbits transplanted with BMCs than in controls. Immunohistochemical analyses carried out 2 weeks after transplantation showed no new myocardium and a very small number of endothelial cells originating from BMCs. On the other hand, immunoblotting revealed upregulated expression of transforming growth factor-beta1 and downregulated expression of matrix metalloproteinase-1 and tumor necrosis factor-alpha following BMC transplantation. In conclusion, autologous BMC transplantation into cardiomyopathic rabbit hearts ameliorates the decline in ventricular function without regenerating cardiomyocytes, most likely by altering expression of various cytokines.


Asunto(s)
Trasplante de Médula Ósea , Cardiomiopatías/fisiopatología , Cardiomiopatías/cirugía , Función Ventricular Izquierda , Animales , Vasos Sanguíneos/fisiología , Cardiomiopatías/inducido químicamente , Doxorrubicina , Inmunohistoquímica , Microscopía Confocal , Conejos , Recuperación de la Función , Regeneración , Volumen Sistólico , Factor de Necrosis Tumoral alfa/metabolismo
19.
Lab Invest ; 86(1): 32-44, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16304579

RESUMEN

Chronic heart failure remains a leading cause of mortality. Although granulocyte colony-stimulating factor (G-CSF) is reported to have a beneficial affect on postinfarction cardiac remodeling and dysfunction when administered before the onset of or at the acute stage of myocardial infarction (MI), its effect on established heart failure is unknown. We show here that subcutaneous administration of G-CSF greatly improves the function of murine hearts failing due to a large, healed MI. G-CSF changed the geometry of the infarct scar from elongated and thin to short and thick, induced hypertrophy among surviving cardiomyocytes, and reduced myocardial fibrosis. Expression of G-CSF receptor was confirmed in failing hearts and was upregulated by G-CSF treatment. G-CSF treatment also led to activation of signal transducer and activator of transcription-3 and induction of GATA-4 and various sarcomeric proteins such as myosin heavy chain, troponin I and desmin. Expression of metalloproteinase-2 and -9 was also increased in G-CSF-treated hearts, while that of tumor necrosis factor-alpha, angiotensin II type 1 receptor (AT1) and transforming growth factor-beta1 was reduced. Although activation of Akt was noted in G-CSF-treated hearts, vessel density was unchanged, and apoptosis was too rare to exert a meaningful effect. No bone marrow-derived cardiomyocytes or vascular cells were detected in the failing hearts of green fluorescent protein chimeric mice. Finally, beneficial effects of G-CSF on cardiac function were found persisting long after discontinuing the treatment (2 weeks). Collectively, these findings suggest G-CSF administration could be an effective approach to treating chronic heart failure following a large MI.


Asunto(s)
Gasto Cardíaco Bajo/tratamiento farmacológico , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Animales , Apoptosis , Western Blotting , Enfermedad Crónica , Ensayo de Inmunoadsorción Enzimática , Corazón/fisiopatología , Ratones , Ratones Endogámicos C57BL , Regeneración
20.
Am J Pathol ; 168(2): 386-97, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16436654

RESUMEN

In UM-X7.1 hamster model of human dilated cardiomyopathy, heart failure progressively develops and causes 50% mortality by 30 weeks of age. Through ultrastructural analysis, we found that many cardiomyocytes of this model contain typical autophagic vacuoles including degraded mitochondria, glycogen granules, and myelin-like figures. In addition, ubiquitin, cathepsin D, and Rab7 were overexpressed as determined by immunoassays. Importantly, most cardiomyocytes with leaky plasma membranes were positive for cathepsin D, suggesting a direct link between autophagic degeneration and cell death. Meanwhile, cardiomyocyte apoptosis appeared insignificant. Granulocyte colony-stimulating factor (10 microg/kg/day), injected 5 days/week from 15 to 30 weeks of age, improved survival among 30-week-old hamsters (100% versus 53% in the untreated hamsters, P < 0.0001); ventricular function and remodeling, increased cardiomyocyte size, and reduced myocardial fibrosis followed by a dramatic reduction in the autophagic findings were also seen. Granulocyte colony-stimulating factor also down-regulated tumor necrosis factor-alpha and increased activities of Akt signal transducer and activator of transcription-3, and matrix metalloproteinases. However, there was no clear evidence of transdifferentiation from bone marrow cells into cardiomyocytes. In conclusion, autophagic death is important for cardiomyocyte loss in the cardiomyopathic hamster, and the beneficial effect of granulocyte colony-stimulating factor acts mainly via an anti-autophagic mechanism rather than anti-apo-ptosis or regeneration.


Asunto(s)
Apoptosis , Autofagia , Cardiomiopatía Dilatada/prevención & control , Fibrosis/prevención & control , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Miocitos Cardíacos/patología , Animales , Cardiomiopatía Dilatada/metabolismo , Catepsina D/metabolismo , Cricetinae , Fibrosis/metabolismo , Masculino , Mesocricetus , Mitocondrias/patología , Vaina de Mielina , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Recombinantes , Factor de Transcripción STAT3/metabolismo , Tasa de Supervivencia , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina/metabolismo , Vacuolas , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA