Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Zhonghua Xin Xue Guan Bing Za Zhi ; 42(4): 321-6, 2014 Apr.
Artículo en Zh | MEDLINE | ID: mdl-24924460

RESUMEN

OBJECTIVE: To observe the impact of PDE5shRNA on cardiac remodeling and heart function following myocardial infarction in mice. METHODS: Myocardial infarction (MI) was induced in mice by left coronary artery ligation. Mice were randomly assigned to sham group (n = 6), PDE5shRNA group (n = 12), common adenovirus group (n = 15) and DMEM group (n = 8). Four weeks post-MI, the survival rate was evaluated. Cardiac function was examined by echocardiography. HE staining and Masson staining were used to evaluate the myocardial infarction size and fibrosis. The number of blood vessels was evaluated by immunohistochemistry, PDE5 protein expression in the left ventricular was detected using Western blot, level of cGMP or PKG activity in the left ventricle was evaluated with ELISA. RESULTS: Four weeks post-MI, all mice survived in the sham group, 3(37%) mice died in the DMEM group, 1 (8%) died in the PDE5shRNA group and 5 died in the common adenovirus group (33%). Infarct size was significantly reduced in PDE5shRNA group compared with the common adenovirus group and DMEM group [(25.4 ± 2.9)% vs. (42.0 ± 3.2)% and (43.4 ± 2.6) %, P < 0.05]. Cardiac function was significantly improved in PDE5shRNA group compared to common adenovirus group and DMEM group[LVFS: (21.1 ± 3.7)% vs. (14.2 ± 2.9)% and (14.22 ± 2.91)%, all P < 0.05; LVEF: (48.2 ± 7.1)% vs. (34.6 ± 6.2)% and (38.1 ± 2.8)%, all P < 0.05; LVESD: (3.87 ± 0.45) mm vs.(4.91 ± 0.62) mm and (4.63 ± 0.37) mm, all P < 0.05]. The blood vessel density was also higher in PDE5shRNA group compared with common adenovirus group (infarct area:14.3 ± 2.0 vs. 6.6 ± 1.2, P < 0.05; periinfarct area: 23.6 ± 2.1 vs. 13.7 ± 2.4, P < 0.05). Compared with common adenovirus group, level of PDE5 was significantly downregulated and level of cGMP or PKG was significantly upregulated in PDE5shRNA group (all P < 0.05). CONCLUSIONS: Present study suggests PDE5shRNA improves cardiac function and attenuates cardiac remodeling through reducing infarction size and cardiac fibrosis and these beneficial effects are possibly mediated by activating cGMP/PKG signaling pathway.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/genética , Insuficiencia Cardíaca/terapia , ARN Interferente Pequeño/genética , Remodelación Ventricular , Adenoviridae/genética , Animales , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/complicaciones
2.
Circ Res ; 109(1): 60-70, 2011 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-21566212

RESUMEN

RATIONALE: Skeletal myoblasts (SMs) with inherent myogenic properties are better candidates for reprogramming to pluripotency. OBJECTIVE: To reprogram SMs to pluripotency and show that reprogrammed SMs (SiPS) express embryonic gene and microRNA profiles and that transplantation of predifferentiated cardiac progenitors reduce tumor formation. METHODS AND RESULTS: The pMXs vector containing mouse cDNAs for Yamanaka's quartet of stemness factors were used for transduction of SMs purified from male Oct4-GFP(+) transgenic mouse. Three weeks later, GFP(+) colonies of SiPS were isolated and propagated in vitro. SiPS were positive for alkaline phosphatase, expressed SSEA1, and displayed a panel of embryonic stem (ES) cell-specific pluripotency markers. Embryoid body formation yielded beating cardiomyocyte-like cells, which expressed early and late cardiac-specific markers. SiPS also had an microRNA profile that was altered during their cardiomyogenic differentiation. Noticeable abrogation of let-7 family and significant up-regulation of miR-200a-c was observed in SiPS and SiPS-derived cardiomyocytes, respectively. In vivo studies in an experimental model of acute myocardial infarction showed extensive survival of SiPS and SiPS-derived cardiomyocytes in mouse heart after transplantation. Our results from 4-week studies in DMEM without cells (group 1), SMs (group-2), SiPS (group-3), and SiPS-derived cardiomyocytes (group 4) showed extensive myogenic integration of the transplanted cells in group 4 with attenuated infarct size and improved cardiac function without tumorgenesis. CONCLUSIONS: Successful reprogramming was achieved in SMs with ES cell-like microRNA profile. Given the tumorgenic nature of SiPS, their predifferentiation into cardiomyocytes would be important for tumor-free cardiogenesis in the heart.


Asunto(s)
Neoplasias Cardíacas/prevención & control , Células Madre Pluripotentes Inducidas/trasplante , Mioblastos Esqueléticos/citología , Infarto del Miocardio/terapia , Miocitos Cardíacos/trasplante , Animales , Diferenciación Celular , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/análisis , Desarrollo de Músculos , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Regeneración , Sístole , Función Ventricular Izquierda
3.
Am J Physiol Heart Circ Physiol ; 302(10): H2112-21, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22447941

RESUMEN

We previously showed that treatment with tadalafil, a long-acting phosphodiesterase-5a (PDE5a) inhibitor, effectively prevented adverse left ventricular (LV) remodeling of the infarcted heart. We hypothesized that short-hairpin RNA (shRNA) therapy targeting PDE5a would simulate the effects of pharmacological intervention for treatment of postinfarction LV remodeling and dysfunction. Experimental model of myocardial infarction was developed in female mice by permanent ligation of left coronary artery. Immediately after that, an adenoviral vector encoding for shRNA sequence targeting PDE5a (Ad-shPDE5a) was injected intramyocardially, which specifically inhibited PDE5a in the heart. Four weeks later, Ad-shPDE5a treated mice showed significant mitigation of the left ventricle (LV) dilatation and dysfunction as indicated by smaller LV cavity and more preserved ejection fraction and fractional shortening. Infarction size and fibrosis were significantly reduced in Ad-shPDE5a-treated mice. Additionally, more salvaged cardiomyocytes, significantly reduced collagen contents, and higher blood vessel density were observed in Ad-shPDE5a-treated mice. The cytoprotective effects of Ad-shPDE5a were demonstrated in vitro in Ad-shPDE5a transfected cardiomyocytes cultured under oxygen glucose deprivation. Among downstream mediators of PDE5a signaling, cyclic GMP (cGMP) and cGMP-dependent protein kinase G (PKG) were activated with concomitant reduction in caspase-3 activity. However, no significant change in PKA and cAMP activities were observed in Ad-shPDE5a-treated hearts. Inhibition with shRNA improved cardiac remodeling and dysfunction by reducing infarction size and cardiac fibrosis and increased cGMP and PKG activity. These findings suggest that PDE5 inhibition with Ad-shPDE5a is a novel approach for treatment of myocardial infarction.


Asunto(s)
Adenoviridae/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Infarto del Miocardio/complicaciones , ARN Interferente Pequeño/uso terapéutico , ARN Viral/uso terapéutico , Disfunción Ventricular Izquierda/prevención & control , Remodelación Ventricular , Animales , Células Cultivadas , Vasos Coronarios/fisiopatología , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/efectos de los fármacos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/genética , Femenino , Ligadura/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Infarto del Miocardio/etiología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , ARN Interferente Pequeño/farmacología , ARN Viral/farmacología , Transfección , Disfunción Ventricular Izquierda/fisiopatología , Remodelación Ventricular/efectos de los fármacos , Remodelación Ventricular/fisiología
4.
Am J Pathol ; 176(2): 687-98, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20035047

RESUMEN

Activation of Fas signaling is a key mediator of doxorubicin cardiotoxicity, which involves both cardiomyocyte apoptosis and myocardial inflammation. In this study, acute cardiotoxicity was induced in mice by doxorubicin, and some mice simultaneously received an intramuscular injection of adenoviral vector encoding mouse soluble Fas (sFas) gene (Ad.CAG-sFas), an inhibitor of Fas/Fas ligand interaction. Two weeks later, left ventricular dilatation and dysfunction were apparent in the LacZ-treated control group, but both were significantly mitigated in the sFas-treated group. The in situ nick-end labeling-positive rate were similar in the two groups, and although electron microscopy revealed cardiomyocyte degeneration, no apoptotic structural features and no activation of caspases were detected, suggesting an insignificant role of apoptosis in this model. Instead, sFas treatment reversed doxorubicin-induced down-regulation of GATA-4 and attenuated ubiquitination of myosin heavy chain and troponin I to preserve these sarcomeric proteins. In addition, doxorubicin-induced significant leukocyte infiltration, fibrosis, and oxidative damage to the myocardium, all of which were largely reversed by sFas treatment. sFas treatment also suppressed doxorubicin-induced p53 overexpression, phosphorylation of c-Jun N-terminal kinase, c-Jun, and inhibitor of nuclear factor-kappaB, as well as production of cyclooxygenase-2 and monocyte chemoattractant protein-1, and it restored extracellular signal-regulated kinase activation. Therefore, sFas gene therapy prevents the progression of doxorubicin-induced acute cardiotoxicity, with accompanying attenuation of the cardiomyocyte degeneration, inflammation, fibrosis, and oxidative damage caused by Fas signaling.


Asunto(s)
Apoptosis/fisiología , Doxorrubicina , Terapia Genética/métodos , Cardiopatías/inducido químicamente , Cardiopatías/terapia , Receptor fas/genética , Animales , Apoptosis/genética , Daño del ADN/genética , Ecocardiografía , Fibrosis Endomiocárdica/genética , Fibrosis Endomiocárdica/patología , Fibrosis Endomiocárdica/prevención & control , Cardiopatías/genética , Cardiopatías/patología , Inflamación/genética , Inflamación/patología , Operón Lac , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocardio/patología , Miocardio/ultraestructura , Estrés Oxidativo/genética , Estrés Oxidativo/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Solubilidad , Receptor fas/antagonistas & inhibidores
5.
Mol Med Rep ; 24(2)2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34080028

RESUMEN

Dysregulation of long non­coding RNA (IncRNA) antisense non­coding RNA in the INK4 locus (ANRIL) is associated with the risk of myocardial infarction (MI). Therefore, the present study aimed to determine the mechanisms underlying this association, which is currently poorly understood, to the best of our knowledge. The current study used an in vitro myocardial ischemia and reperfusion (MI/R) model, in which H9c2 cardiomyocytes were exposed to hypoxia/reoxygenation (H/R), which demonstrated that ANRIL expression was downregulated and that ANRIL positively regulated sirtuin 1 (SIRT1) expression following H/R injury. Subsequently, it was demonstrated that ANRIL upregulated SIRT1 expression by sponging microRNA­181a (miR­181a). In addition, ANRIL overexpression reduced lactate dehydrogenase release and apoptosis of H9c2 cardiomyocytes exposed to H/R, indicating that ANRIL prevented H/R­induced cardiomyocyte injury. Moreover, both miR­181a overexpression and SIRT1 knockdown significantly decreased the protective effects of ANRIL on H/R­induced cardiomyocyte injury, thus demonstrating that SIRT1 upregulation via sponging miR­181a is a critical mechanism that mediates the function of ANRIL. These results provided a novel mechanistic insight into the role of ANRIL in H/R­injured cardiomyocytes and suggested that the ANRIL/miR­181a/SIRT1 axis may be a therapeutic target for reducing MI/R injury.


Asunto(s)
Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo , Animales , Apoptosis/genética , Hipoxia de la Célula/genética , Línea Celular , Regulación hacia Abajo/genética , MicroARNs/metabolismo , Modelos Biológicos , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Oxidación-Reducción , Ratas , Regulación hacia Arriba/genética
6.
Am J Pathol ; 174(5): 1705-14, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19342365

RESUMEN

To examine the functional significance and morphological characteristics of starvation-induced autophagy in the adult heart, we made green fluorescent protein-microtubule-associated protein 1-light chain 3 (LC3) transgenic mice starve for up to 3 days. Electron microscopy revealed round, homogenous, electron-dense lipid droplet-like vacuoles that initially appeared in cardiomyocytes as early as 12 hours after starvation; these vacuoles were identified as lysosomes based on cathepsin D-immunopositive reactivity and acid phosphatase activity. The increase in the number of lysosomes depended on the starvation interval; typical autophagolysosomes with intracellular organelles also appeared, and their numbers increased at the later phases of starvation. Myocardial expression of autophagy-related proteins, LC3-II, cathepsin D, and ubiquitin, increased, whereas both myocardial ATP content and starvation integral decreased. Treatment with bafilomycin A1, an autophagy inhibitor, did not affect cardiac function in normally fed mice but significantly depressed cardiac function and caused significant left ventricular dilatation in mice starved for 3 days. The cardiomyocytes were occupied with markedly accumulated lysosomes in starved mice treated with bafilomycin A1, and both the myocardial amino acid content, which was increased during starvation, and the myocardial ATP content were severely decreased, potentially contributing to cardiac dysfunction. The present findings suggest a critical role of autophagy in the maintenance of cardiac function during starvation in the adult.


Asunto(s)
Autofagia , Proteínas Asociadas a Microtúbulos/fisiología , Miocitos Cardíacos/citología , Miocitos Cardíacos/ultraestructura , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Peso Corporal/efectos de los fármacos , Catepsina D/metabolismo , Inhibidores Enzimáticos/farmacología , Técnica del Anticuerpo Fluorescente , Proteínas Fluorescentes Verdes/genética , Pruebas de Función Cardíaca , Técnicas para Inmunoenzimas , Lisosomas/metabolismo , Lisosomas/ultraestructura , Macrólidos/farmacología , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , ATPasas de Translocación de Protón/antagonistas & inhibidores , Ratas , Ubiquitina/metabolismo , Vacuolas/metabolismo , Vacuolas/ultraestructura , Disfunción Ventricular Izquierda/metabolismo
7.
Circ Res ; 103(1): 98-106, 2008 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-18519944

RESUMEN

Although recanalization of the infarct-related artery late after myocardial infarction (MI) is known to reduce both cardiac remodeling and mortality, the mechanisms responsible are not yet fully understood. We compared infarcted rat hearts in which the infarct-related coronary artery was opened 24 hours after infarction (late reperfusion [LR] group) with those having a permanently occluded artery. Left ventricular dilatation and dysfunction were significantly mitigated in the LR group 1, 2, and 4 weeks post-MI. Attributable, in large part, to the greater number of cells present, the infarcted wall was significantly thicker in the LR group, which likely reduced wall stress and mitigated cardiac dysfunction. Granulation tissue cell proliferation was increased to a greater degree in the LR group 4 days post-MI, whereas the incidence of apoptosis was significantly lower throughout the subacute stage (4 days, 1 week, and 2 weeks post-MI), further suggesting preservation of granulation tissue cells contributes to the thick, cell-rich scar. Functionally, myocardial debris was more rapidly removed from the infarcted areas in the LR group during subacute stages, and stouter collagen was more rapidly synthesized in those areas. Direct acceleration of Fas-mediated apoptosis by hypoxia was confirmed in vitro using infarct tissue-derived myofibroblasts. In salvaged cardiomyocytes, degenerative changes, but not apoptosis, were mitigated in the LR group, accompanied by restoration of GATA-4 and sarcomeric protein expression. Along with various mechanisms proposed earlier, the present findings appear to provide an additional pathophysiological basis for the benefits of late reperfusion.


Asunto(s)
Infarto del Miocardio/fisiopatología , Reperfusión Miocárdica , Miocitos Cardíacos , Remodelación Ventricular , Animales , Apoptosis , Proteínas Portadoras/metabolismo , Colágeno/biosíntesis , Proteínas del Citoesqueleto , Fibroblastos/metabolismo , Fibroblastos/patología , Factor de Transcripción GATA4/metabolismo , Humanos , Masculino , Infarto del Miocardio/metabolismo , Infarto del Miocardio/mortalidad , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Reperfusión Miocárdica/métodos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas , Ratas Wistar , Factores de Tiempo
8.
Am J Physiol Heart Circ Physiol ; 297(4): H1504-13, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19684189

RESUMEN

The small leucine-rich proteoglycan decorin is a natural inhibitor of transforming growth factor-beta (TGF-beta) and exerts antifibrotic effects in heart and to stimulate skeletal muscle regeneration. We investigated decorin's chronic effects on postinfarction cardiac remodeling and dysfunction. Myocardial infarction (MI) was induced in mice by left coronary artery ligation. An adenoviral vector encoding human decorin (Ad. CAG-decorin) was then injected into the hindlimbs on day 3 post-MI (control, Ad.CAG-LacZ). Four weeks post-MI, the decorin-treated mice showed significant mitigation of the left ventricular dilatation and dysfunction seen in control mice. Although infarct size did not differ between the two groups, the infarcted wall thickness was greater and the segmental length of the infarct was smaller in decorin-treated mice. In addition, cellular components, including myofibroblasts and blood vessels, were more abundant within the infarcted area in decorin-treated mice, and fibrosis was significantly reduced in both the infarcted and noninfarcted areas of the left ventricular wall. Ten days post-MI, there was greater cell proliferation and less apoptosis among granulation tissue cells in the infarcted areas of decorin-treated mice. The treatment, however, did not affect proliferation and apoptosis of salvaged cardiomyocytes. Although decorin gene therapy did not affect TGF-beta1 expression in the infarcted heart, it inhibited Smad2/3 activation (downstream mediators of TGF-beta signaling). In summary, postinfarction decorin gene therapy mitigated cardiac remodeling and dysfunction by altering infarct tissue noncardiomyocyte dynamics and preventing cardiac fibrosis, accompanying inhibition of Smad2/3 activation.


Asunto(s)
Adenoviridae/genética , Proteínas de la Matriz Extracelular/biosíntesis , Terapia Genética , Vectores Genéticos , Hipertrofia Ventricular Izquierda/prevención & control , Infarto del Miocardio/terapia , Miocitos Cardíacos/metabolismo , Proteoglicanos/biosíntesis , Disfunción Ventricular Izquierda/prevención & control , Remodelación Ventricular , Animales , Apoptosis , Proliferación Celular , Enfermedad Crónica , Decorina , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/genética , Fibrosis , Células HeLa , Humanos , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/patología , Proteoglicanos/genética , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factores de Tiempo , Transducción Genética , Transfección , Factor de Crecimiento Transformador beta1/metabolismo , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/fisiopatología
9.
Circulation ; 113(4): 535-43, 2006 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-16449733

RESUMEN

BACKGROUND: Doxorubicin is a highly effective antineoplastic drug, but its clinical use is limited by its adverse side effects on the heart. We investigated possible protective effects of erythropoietin against doxorubicin-induced cardiomyopathy. METHODS AND RESULTS: Cardiomyopathy was induced in mice by a single intraperitoneal injection of doxorubicin (15 mg/kg). In some cases, human recombinant erythropoietin (5000 U/kg) was started simultaneously. Two weeks later, left ventricular dilatation and dysfunction were apparent in mice given doxorubicin but were significantly attenuated by erythropoietin treatment. Erythropoietin also protected hearts against doxorubicin-induced cardiomyocyte atrophy and degeneration, myocardial fibrosis, inflammatory cell infiltration, and downregulation of expression of GATA-4 and 3 sarcomeric proteins, myosin heavy chain, troponin I, and desmin. Cyclooxygenase-2 expression was upregulated in doxorubicin-treated hearts, and that, too, was attenuated by erythropoietin. No doxorubicin-induced apoptotic effects were seen, nor were any changes seen in the expression of tumor necrosis factor-alpha or transforming growth factor-beta1. Antiatrophic and GATA-4 restoring effects of erythropoietin were demonstrated in the in vitro experiments with cultured cardiomyocytes exposed to doxorubicin, which indicated the direct cardioprotective effects of erythropoietin beyond erythropoiesis. Cardiac erythropoietin receptor expression was downregulated in doxorubicin-induced cardiomyopathy but was restored by erythropoietin. Among the downstream mediators of erythropoietin receptor signaling, activation of extracellular signal-regulated kinase was reduced by doxorubicin but restored by erythropoietin. By contrast, erythropoietin was ineffective when administered after cardiac dysfunction was established in the chronic stage. CONCLUSIONS: The present study indicates a protective effect of erythropoietin against doxorubicin-induced cardiomyopathy.


Asunto(s)
Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/prevención & control , Eritropoyetina/farmacología , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/prevención & control , Animales , Antibióticos Antineoplásicos , Cardiomiopatías/inducido químicamente , Caspasa 3 , Caspasas/metabolismo , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Doxorrubicina , Recuento de Eritrocitos , Eritropoyetina/sangre , Factor de Transcripción GATA4/metabolismo , Insuficiencia Cardíaca/inducido químicamente , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Receptores de Eritropoyetina/metabolismo , Sarcómeros/metabolismo , Tasa de Supervivencia , Disfunción Ventricular Izquierda/inducido químicamente , Disfunción Ventricular Izquierda/tratamiento farmacológico , Disfunción Ventricular Izquierda/prevención & control
10.
J Card Fail ; 13(2): 155-62, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17395057

RESUMEN

BACKGROUND: In patients with end-stage renal disease, angiotensin II type 1A receptor (AT1) blockade attenuates the associated cardiac dysfunction. We investigated the molecular signaling mediating that effect. METHODS AND RESULTS: We used 5/6 nephrectomy to induce significant renal dysfunction in AT1 knockout (AT1KO) and wild-type mice (WT). Twelve weeks after nephrectomy, WT showed significant left ventricular dilation and dysfunction that were accompanied by cardiomyocyte hypertrophy, fibrosis, and reduced capillary density. All of these effects were significantly mitigated in AT1KO. Nephrectomy led to upregulation of myocardial expression of AT1, transforming growth factor-beta1 (TGF-beta1), matrix metalloproteinase (MMP)-2, MMP-9, tissue inhibitor of metalloproteinase-1 (TIMP-1), and phosphorylated Akt (p-Akt), and also led to increased oxidative damage in cardiomyocytes. In AT1KO, TGF-beta1, TIMP-1, oxidative damage levels were lower, whereas MMPs and p-Akt levels were higher. Treating nephrectomized WT mice with valsartan (an AT1 blocker), but not hydralazine, improved cardiac function and altered molecular signaling in a manner similar to that seen in AT1KO mice. Notably, AT1 expression was downregulated in valsartan-treated but not hydralazine-treated hearts. CONCLUSIONS: These findings provide novel insight into the mechanism underlying the beneficial effects of AT1 blockade on cardiac function in a model of renal dysfunction-associated heart failure.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Antihipertensivos/uso terapéutico , Insuficiencia Cardíaca/etiología , Fallo Renal Crónico/prevención & control , Receptor de Angiotensina Tipo 1/sangre , Transducción de Señal/fisiología , Tetrazoles/uso terapéutico , Valina/análogos & derivados , Animales , Nitrógeno de la Urea Sanguínea , Western Blotting , Creatinina/sangre , ADN/genética , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/fisiopatología , Inmunohistoquímica , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/metabolismo , Masculino , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Nefrectomía/efectos adversos , Estrés Oxidativo/genética , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Receptor de Angiotensina Tipo 1/genética , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/genética , Resultado del Tratamiento , Regulación hacia Arriba/efectos de los fármacos , Valina/uso terapéutico , Valsartán , Función Ventricular Izquierda/efectos de los fármacos
11.
Cardiovasc Res ; 71(4): 684-94, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16828072

RESUMEN

OBJECTIVES: Late treatment with erythropoietin (EPO), as well as the administration before the onset of or during the acute stage of myocardial infarction (MI), has recently been shown to mitigate post-MI heart failure. We investigated the mechanisms, including the downstream signaling pathways, for the beneficial effect of late treatment with EPO on chronic post-MI heart failure. METHODS AND RESULTS: EPO (1500 U/kg, twice a week) was administered to mice beginning 6 weeks after induction of large MI. The EPO treatment for 4 weeks diminished left ventricular dilatation and improved function. It significantly reduced inflammatory cell infiltration and fibrosis, and increased vascular density in noninfarcted areas. The elevated levels of the inflammatory cytokines interleukin (IL)-1beta, IL-6, tumor necrosis factor-alpha and transforming growth factor-beta1 seen in the failing hearts were returned nearly to control levels by EPO treatment. Oxidative damage in surviving cardiomyocytes was also significantly attenuated by EPO. Expression of EPO receptor was upregulated in failing hearts, and EPO treatment led to myocardial activation of signal transducer and activator of transcription-3 (Stat3), Stat5, and Akt. These in vivo effects of EPO were confirmed in vitro in experiments that showed the anti-inflammatory and anti-oxidant effects of EPO to be mediated via Stat and Akt activation. Finally, the beneficial effects of EPO were found to persist for 4 weeks after discontinuing treatment. CONCLUSIONS: It thus appears that Stat-mediated reduction of inflammation and cytokine production and Akt-mediated attenuation of oxidative stress accompany the beneficial effects of late treatment with EPO on chronic post-MI heart failure.


Asunto(s)
Citocinas/metabolismo , Eritropoyetina/uso terapéutico , Insuficiencia Cardíaca/inmunología , Miocardio/inmunología , Animales , Western Blotting , Células Cultivadas , Ecocardiografía , Fibroblastos/efectos de los fármacos , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/patología , Hematócrito , Peróxido de Hidrógeno/farmacología , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Modelos Animales , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Oxidación-Reducción , Distribución Aleatoria , Receptores de Eritropoyetina/análisis , Receptores de Eritropoyetina/metabolismo , Proteínas Recombinantes , Transducción de Señal/efectos de los fármacos , Tiempo , Remodelación Ventricular/efectos de los fármacos
12.
PLoS One ; 10(12): e0145766, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26709517

RESUMEN

INTRODUCTION: Treatment with short hairpin RNA (shRNA) interference therapy targeting phosphodiesterase 5a after myocardial infarction (MI) has been shown to mitigate post-MI heart failure. We investigated the mechanisms that underpin the beneficial effects of PDE5a inhibition through shRNA on post-MI heart failure. METHODS: An adenoviral vector with an shRNA sequence inserted was adopted for the inhibition of phosphodiesterase 5a (Ad-shPDE5a) in vivo and in vitro. Myocardial infarction (MI) was induced in male C57BL/6J mice by left coronary artery ligation, and immediately after that, the Ad-shPDE5a was injected intramyocardially around the MI region and border areas. RESULTS: Four weeks post-MI, the Ad-shPDE5a-treated mice showed significant mitigation of the left ventricular (LV) dilatation and dysfunction compared to control mice. Infarction size and fibrosis were also significantly reduced in Ad-shPDE5a-treated mice. Additionally, Ad-shPDE5a treatment decreased the MI-induced inflammatory cytokines interleukin (IL)-1ß, IL-6, tumor necrosis factor-α, and transforming growth factor-ß1, which was confirmed in vitro in Ad-shPDE5a transfected myofibroblasts cultured under oxygen glucose deprivation. Finally, Ad-shPDE5a treatment was found to activate the myocardial Akt signaling pathway in both in vivo and in vitro experiments. CONCLUSION: These findings indicate that PDE5a inhibition by Ad-shPDE5a via the Akt signal pathway could be of significant value in the design of future therapeutics for post-MI heart failure.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Citocinas/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/terapia , Inhibidores de Fosfodiesterasa 5/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Células Cultivadas , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/genética , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/terapia , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/patología , Miocardio/patología , Miofibroblastos/metabolismo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico , Transducción de Señal
13.
J Cardiol ; 62(2): 110-6, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23731918

RESUMEN

BACKGROUND: It is not clear yet how tadalafil affects nonischemic cardiomyopathy, although its beneficial effects on acute myocardial infarction are well-known. We investigated tadalafil's beneficial effects on nonischemic cardiomyopathy and the specific mechanisms of its effects. METHODS: Cardiomyopathy was induced in mice by a single intraperitoneal injection of doxorubicin (15 mg/kg). In some cases, tadalafil (4 mg/kg/day, p.o., 14 days) was started simultaneously. After two weeks, cardiac function was evaluated by echocardiography and cardiac catheterization, then all of the mice were killed and cardiac specimens were subjected for hemotoxylin and eosin staining, Masson's trichrome staining, terminal deoxynucleotidyltransferase dUTP nick-end labeling assay, enzyme-linked immunosorbent assay, and Western blot. RESULTS: Two weeks later, left ventricular dilatation and dysfunction were apparent in mice given doxorubicin but were significantly attenuated by tadalafil treatment. Tadalafil also protected hearts against doxorubicin-induced cardiomyocyte atrophy/degeneration and myocardial fibrosis. No doxorubicin-induced apoptotic effects were seen between groups. Cardiac cGMP level was lower in the doxorubicin-treated group, however it was significantly increased with tadalafil treatment. Compared to the control group, the myocardial expression of 3 sarcomeric proteins, myosin heavy chain, troponin I, and desmin were significantly decreased in the doxorubicin-treated group, which were restored by the tadalafil treatment. CONCLUSIONS: The present study indicates a protective effect of tadalafil mainly through cGMP signaling pathway against doxorubicin-induced nonischemic cardiomyopathy.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Carbolinas/farmacología , Carbolinas/uso terapéutico , Cardiomiopatías/inducido químicamente , Cardiomiopatías/tratamiento farmacológico , GMP Cíclico/metabolismo , Doxorrubicina/efectos adversos , Inhibidores de Fosfodiesterasa 5/farmacología , Inhibidores de Fosfodiesterasa 5/uso terapéutico , Disfunción Ventricular Izquierda/inducido químicamente , Disfunción Ventricular Izquierda/tratamiento farmacológico , Animales , Atrofia , Cardiomiopatías/metabolismo , Cardiotónicos , GMP Cíclico/fisiología , Modelos Animales de Enfermedad , Fibrosis , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/patología , Transducción de Señal/efectos de los fármacos , Tadalafilo , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/patología
14.
J Invasive Cardiol ; 25(11): 600-4, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24184895

RESUMEN

BACKGROUND: Adverse cardiac event rates are higher for percutaneous coronary intervention (PCI) of bifurcation lesions. Currently, provisional stenting or a simple stenting strategy is the standard treatment for bifurcated lesions, but its performance remains limited because of a risk of side-branch (SB) closure and a higher rate of target lesion revascularization (TLR). We report a new provisional side-branch stenting strategy to treat coronary bifurcation lesions using a "balloon-stent kissing" technique (BSKT). METHODS: From January 2011 to December 2012, a total of 60 patients with 60 bifurcation lesions underwent PCI using BSKT. Baseline and postprocedural quantitative coronary angiography (QCA) analyses were performed. Procedural and immediate clinical outcomes were reviewed. RESULTS: The majority of patients presented with acute coronary syndrome (98%) and had true bifurcation lesions (98%). TIMI-3 flow was established in 100% of the main branch and SB lesions. QCA revealed preservation of the bifurcation angle after PCI (pre PCI, 57.0 ± 16.3; post PCI, 60.5 ± 16.1; P=.24). Five patients (8%) had lesions that required rewiring and 2 patients (3%) required provisional stenting of the SB. No SB loss occurred during PCI. No patient had a periprocedural myocardial infarction, nor was the balloon or wire entrapped during any PCI. CONCLUSIONS: As a new coronary bifurcation provisional stenting method, the BSKT is associated with a high procedural success rate, improved SB patency, and a low rate of immediate cardiac events.


Asunto(s)
Angioplastia Coronaria con Balón/instrumentación , Estenosis Coronaria/cirugía , Stents Liberadores de Fármacos , Angiografía Coronaria , Estenosis Coronaria/diagnóstico por imagen , Diseño de Equipo , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Factores de Tiempo , Resultado del Tratamiento , Ultrasonografía Intervencional
15.
J Am Coll Cardiol ; 56(23): 1949-58, 2010 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-21109120

RESUMEN

OBJECTIVES: We examined the effect of asialoerythropoietin (asialoEPO), a nonerythrogenic derivative of erythropoietin (EPO), on renal dysfunction-associated heart failure. BACKGROUND: Although EPO is known to exert beneficial effects on cardiac function, the clinical benefits in patients with chronic kidney disease are controversial. It remains to be addressed whether previously reported outcomes were the result of relief of the anemia, adverse effects of EPO, or direct cardiovascular effects. METHODS: Mice underwent 5/6 nephrectomy to cause renal dysfunction. Eight weeks later, when renal dysfunction was established, anemia and cardiac dysfunction and remodeling were apparent. Mice were then assigned to receive saline (control), recombinant human erythropoietin (rhEPO) at 5,000 IU (714 pmol)/kg, or asialoEPO at 714 pmol/kg, twice/week for 4 weeks. RESULTS: Although only rhEPO relieved the nephrectomy-induced anemia, both rhEPO and asialoEPO significantly and similarly mitigated left ventricular dilation and dysfunction. The hearts of rhEPO- or asialoEPO-treated mice showed less hypertrophy, reflecting decreases in cardiomyocyte hypertrophy and degenerative subcellular changes, as well as significant attenuation of fibrosis, leukocyte infiltration, and oxidative deoxyribonucleic acid damage. These phenotypes were accompanied by restored expression of GATA-4, sarcomeric proteins, and vascular endothelial growth factor and decreased inflammatory cytokines and lipid peroxidation. Finally, myocardial activation was observed of extracellular signal-regulated protein kinase and signal transducer and activator of transcription pathways in the treated mice. CONCLUSIONS: EPO receptor signaling exerts direct cardioprotection in an animal model of renal dysfunction-associated heart failure, probably by mitigating degenerative, pro-fibrosis, inflammatory, and oxidative processes but not through relief of anemia.


Asunto(s)
Anemia/complicaciones , Asialoglicoproteínas/uso terapéutico , Eritropoyetina/análogos & derivados , Eritropoyetina/uso terapéutico , Insuficiencia Cardíaca/etiología , Receptores de Eritropoyetina/metabolismo , Insuficiencia Renal/complicaciones , Transducción de Señal , Anemia/tratamiento farmacológico , Anemia/metabolismo , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Proteínas Recombinantes , Insuficiencia Renal/tratamiento farmacológico , Insuficiencia Renal/metabolismo
16.
Med Mol Morphol ; 42(2): 92-101, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19536616

RESUMEN

To address whether adult rat ventricular cardiomyocytes (ARVCs) exposed to oxidant stress die via apoptosis (secondarily by necrosis) or primarily by necrosis, we exposed ARVCs to hydrogen peroxide (H2O2; 0.1-100 microM) for up to 24 h and then compared them with isoproterenol-induced apoptotic and Triton X-induced necrotic controls. Cellular shrinkage preceded plasma membrane disruption, reflected by trypan blue uptake in ARVCs exposed to lower concentrations of H2O2 (<1 microM; an apoptotic pattern), but the order was reversed in cells exposed to higher concentrations of H2O2 (>1 microM; a necrotic pattern). DNA fragmentation, caspase-3 activation, mitochondrial membrane potential preservation, and ATP preservation were all apparent in ARVCs treated with low H2O2 (0.5 microM), but not in those treated with high H2O2 (10 microM). In addition, electron microscopy revealed unique morphology in H2O2-treated ARVCs; i.e., the nuclei had a homogeneous ground glass-like appearance that was never accompanied by chromatin condensation. Apparently, high concentrations of H2O2 caused primary necrosis in ARVCs, whereas low concentrations induced biochemically comparable apoptosis, although the latter did not satisfy the morphological criteria of apoptosis. These findings caution against the use of oxidant stress, H2O2 in particular, as an inducer of apoptosis in ARVCs.


Asunto(s)
Apoptosis , Peróxido de Hidrógeno/farmacología , Miocitos Cardíacos/citología , Oxidantes/farmacología , Estrés Oxidativo , Adenosina Trifosfato/metabolismo , Animales , Caspasa 3/metabolismo , Células Cultivadas , Fragmentación del ADN , Ventrículos Cardíacos/citología , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/ultraestructura , Modelos Animales , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/ultraestructura , Necrosis , Ratas , Ratas Sprague-Dawley
17.
J Am Coll Cardiol ; 53(25): 2378-88, 2009 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-19539150

RESUMEN

OBJECTIVES: We hypothesized that erythropoietin (EPO)-immersed gelatin hydrogel microspheres (GHM) injected into ischemic legs might continuously release a small amount of EPO to locally stimulate angiogenesis without unfavorable systemic effects. BACKGROUND: EPO is a potent angiogenic factor, but its use for relieving ischemic organs is limited because of the untoward systemic erythrogenic effect and its short half-life in plasma. METHODS: The right femoral arteries of BALB/c mice were ligated. Recombinant human EPO (5,000 IU/kg)-immersed GHM was injected into the right hind limb muscles (n = 12); the control groups included a saline-injected group (n = 12), an EPO-injected group (n = 8), and an empty GHM-injected group (n = 8). RESULTS: Eight weeks later, improvement of blood perfusion to the ischemic limb was significantly augmented in the EPO-GHM group compared with any of the control groups. There was no increase in the hemoglobin level, nor was there any increase in endothelial progenitor cells. However, capillary and arteriolar densities were significantly increased in this group. Although the treatment did not affect the levels of vascular endothelial growth factor or interleukin-1 beta, it up-regulated the EPO receptor and matrix metalloproteinase-2 and activated the downstream signaling of Akt and also endothelial nitric oxide synthase in ischemic limbs, which might have been associated with the evident angiogenic and arteriogenic effects in the present system. CONCLUSIONS: The present drug delivery system is suggested to have potential as a novel noninvasive therapy for ischemic peripheral artery disease.


Asunto(s)
Eritropoyetina/administración & dosificación , Isquemia/tratamiento farmacológico , Extremidad Inferior/irrigación sanguínea , Enfermedades Vasculares Periféricas/tratamiento farmacológico , Células Madre Adultas/efectos de los fármacos , Inductores de la Angiogénesis/metabolismo , Animales , Células Cultivadas , Preparaciones de Acción Retardada , Células Endoteliales/efectos de los fármacos , Hidrogeles/uso terapéutico , Isquemia/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Microesferas , Miocitos del Músculo Liso/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Enfermedades Vasculares Periféricas/metabolismo , Receptores de Eritropoyetina/metabolismo , Proteínas Recombinantes , Flujo Sanguíneo Regional/efectos de los fármacos
18.
Am J Physiol Heart Circ Physiol ; 296(3): H616-26, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19151252

RESUMEN

We hypothesized that therapy, composed of antiapoptotic soluble Fas (sFas) gene transfer, combined with administration of the cardioprotective cytokine granulocyte colony-stimulating factor (G-CSF), would markedly mitigate cardiac remodeling and dysfunction following myocardial infarction (MI). On the 3rd day after MI induced by ligating the left coronary artery in mice, four different treatments were initiated: saline injection (Group C, n = 26); G-CSF administration (Group G, n = 27); adenoviral transfer of sFas gene (Group F, n = 26); and the latter two together (Group G+F, n = 26). Four weeks post-MI, Group G+F showed better survival than Group C (96 vs. 65%, P < 0.05) and the best cardiac function among the four groups. In Group G, the infarct scar was smaller and less fibrotic, whereas in Group F the scar was thicker, without a reduction in area, and contained abundant myofibroblasts and vascular cells; Group G+F showed both phenotypes. G-CSF exerted a beneficial effect on infarct tissue dynamics through antifibrotic and proliferative effects on granulation tissue; however, it also exerts an adverse proapoptotic effect that leads to thinning of the infarct scar. sFas appeared to offset the latter drawback. In vitro study using cultured myofibroblasts derived from the infarct tissue revealed that G-CSF increased proliferating activity of those cells accompanying activation of Akt and signal transducer and activator of transcription 3, while accelerating Fas-mediated apoptosis with increasing Bax-to-Bcl-2 ratio. The results suggest that combined use of G-CSF administration and sFas gene therapy is a potentially powerful tool against post-MI heart failure.


Asunto(s)
Apoptosis , Cardiotónicos/farmacología , Técnicas de Transferencia de Gen , Terapia Genética , Factor Estimulante de Colonias de Granulocitos/farmacología , Insuficiencia Cardíaca/terapia , Infarto del Miocardio/terapia , Miocardio/patología , Receptor fas/genética , Adenoviridae/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Terapia Combinada , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Fibrosis , Vectores Genéticos , Tejido de Granulación/efectos de los fármacos , Tejido de Granulación/patología , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Lenograstim , Masculino , Ratones , Ratones Endogámicos C57BL , Contracción Miocárdica/efectos de los fármacos , Infarto del Miocardio/complicaciones , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Recombinantes/farmacología , Regeneración/efectos de los fármacos , Regeneración/genética , Factores de Tiempo , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/terapia , Remodelación Ventricular/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo , Receptor fas/metabolismo
19.
Am J Physiol Heart Circ Physiol ; 294(2): H1048-57, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18083897

RESUMEN

Hepatocyte growth factor (HGF) reportedly exerts beneficial effects on the heart following myocardial infarction and during nonischemic cardiomyopathy, but the precise mechanisms underlying the latter have not been well elucidated. We generated nonischemic cardiomyopathy in mice by injecting them with doxorubicin (15 mg/kg ip). Two weeks later, when cardiac dysfunction was apparent, an adenoviral vector encoding human HGF gene (Ad.CAG-HGF, 1x10(11) particles/mouse) was injected into the hindlimb muscles; LacZ gene served as the control. Left ventricular dilatation and dysfunction normally seen 4 wk after doxorubicin administration were significantly mitigated in HGF-treated mice, as were the associated cardiomyocyte atrophy/degeneration and myocardial fibrosis. Myocardial expression of GATA-4 and a sarcomeric protein, myosin heavy chain, was downregulated by doxorubicin, but the expression of both was restored by HGF treatment. The protective effect of HGF against doxorubicin-induced cardiomyocyte atrophy was confirmed in an in vitro experiment, which also showed that neither cardiomyocyte apoptosis nor proliferation plays significant roles in the present model. Upregulation of c-Met/HGF receptor was noted in HGF-treated hearts. Among the mediators downstream of c-Met, the activation of extracellular signal-regulated kinase (ERK) was reduced by doxorubicin, but the activity was restored by HGF. Levels of transforming growth factor-beta1 and cyclooxygenase-2 did not differ between the groups. Our findings suggest the HGF gene delivery exerts therapeutic antiatrophic/degenerative and antifibrotic effects on myocardium in cases of established cardiac dysfunction caused by doxorubicin. These beneficial effects appear to be related to HGF-induced ERK activation and upregulation of c-Met, GATA-4, and sarcomeric proteins.


Asunto(s)
Adenoviridae/genética , Antibióticos Antineoplásicos , Cardiomiopatías/inducido químicamente , Cardiomiopatías/genética , Doxorrubicina , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/fisiología , Animales , Animales Recién Nacidos , Western Blotting , Cardiomiopatías/fisiopatología , Ciclooxigenasa 2/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/fisiología , Vectores Genéticos , Corazón/fisiología , Humanos , Inmunohistoquímica , Inmunoprecipitación , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Plásmidos/genética , Proteínas Proto-Oncogénicas c-met/metabolismo
20.
J Cell Mol Med ; 12(4): 1272-83, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18298650

RESUMEN

Although beneficial effects of granulocyte colony-stimulating factor (G-CSF) have been demonstrated on post-myocardia infarction (MI) process, the mechanisms and feasibility are not fully agreed yet. We investigated effects of a long-term treatment with a low-dose G-CSF started 1 day after the onset of MI, on post-infarction process. One day after being made MI by left coronary ligation, mice were given G-CSF (10 microg/kg/day) for 4 weeks. The G-CSF treatment resulted in a significant mitigation of cardiac remodelling and dysfunction. In the G-CSF-treated hearts, the infarcted scar was smaller with less fibrosis and abundant vessels while in the non-infarcted area, hypertrophic cardiomyocytes with attenuated degenerative changes and reduced fibrosis were apparent. These effects were accompanied by activation of signal transducer and activator of transcription 3 (STAT3) and Akt and also by up-regulation of GATA-4, myosin heavy chain and matrix metalloproteinases-2 and -9. Apoptosis of cardiomyocytes appeared insignificant at any stages. Parthenolide, a STAT3 inhibitor, completely abolished the beneficial effects of G-CSF on cardiac function and remodelling with loss of effect on both anti-cardiomyocyte degeneration and anti-fibrosis. In contrast, wortmannin, an Akt inhibitor, did not affect G-CSF-induced benefits despite cancelling vessel increase. In conclusion, treatment with G-CSF at a small dose but for a long duration beneficially affects the post-infarction process possibly through STAT3-mediated anti-cardiomyocyte degeneration and anti-fibrosis, but not through anti-cardiomyocyte apoptosis or Akt-mediated angio-genesis. The findings may also imply a more feasible way of G-CSF administration in the clinical settings.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Miocardio/patología , Animales , Apoptosis/efectos de los fármacos , Recuento de Células Sanguíneas , Supervivencia Celular/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/farmacología , Granulocitos/citología , Granulocitos/efectos de los fármacos , Pruebas de Función Cardíaca , Humanos , Quinasas Janus/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infarto del Miocardio/enzimología , Infarto del Miocardio/fisiopatología , Miocardio/enzimología , Miocardio/ultraestructura , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Miocitos Cardíacos/ultraestructura , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocito/metabolismo , Proteínas Recombinantes , Factores de Transcripción STAT/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA