Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Intervalo de año de publicación
1.
Transpl Immunol ; 48: 1-9, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29425775

RESUMEN

OBJECTIVE: The aim of the present study was to investigate the inhibitory effect of wild-type P53 gene transfer on graft coronary artery disease (GCAD) after heart transplantation and the underlying mechanisms. METHODS: A rat model of heterotopic heart transplantation was established using Wistar rats as donors and Sprague-Dawley (SD) rats as recipients. The donor hearts were collected and perfused, via the coronary artery, with 800 µl of recombinant adenovirus carrying the P53 gene (Ad-P53). Thirty minutes later, heart transplant was performed. At 5 d after the transplant surgery, the expression of the exogenous P53 gene and protein in the coronary artery tissues of the donor hearts was examined. At 28 d after the transplant surgery, tissues were collected from the transplanted hearts. The degree of coronary artery stenosis was examined, and apoptosis of the coronary artery smooth muscle cells in the donor hearts was analysed. In addition, histological changes in the vital organs of the recipient rats and the levels of serum biochemical indicators in the rats were also examined. RESULTS: The exogenous gene was successfully transferred into donor heart tissues and the coronary artery and was highly expressed. At 28 d after the transplant surgery, the ratio of tunica intima thickness to tunica media thickness (I/M) and the ratio of wall thickness to the lumen diameter of the coronary artery were decreased in the Ad-P53 group compared to those in the Ad-LacZ group and the control group (P < 0.05). A terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) assay revealed that the percentage of apoptotic coronary artery smooth muscle cells in the donor hearts was significantly increased in the Ad-P53 group compared to that in the Ad-LacZ group and the control group (P < 0.01). The wild-type P53 gene had no effect on the morphology and functions of the vital organs of the recipient rats. CONCLUSIONS: P53 gene transfer inhibits coronary artery intimal hyperplasia and reduces the degree of luminal stenosis in transplanted hearts. The inhibitory effect may be related to the wild-type P53 gene-induced apoptosis of vascular smooth muscle cells and inhibition of vascular smooth muscle cell proliferation. This approach is effective and safe and may have good prospects for clinical application.


Asunto(s)
Vasos Coronarios/patología , Oclusión de Injerto Vascular/prevención & control , Trasplante de Corazón , Corazón/fisiología , Miocitos del Músculo Liso/patología , Proteína p53 Supresora de Tumor/genética , Túnica Íntima/patología , Adenoviridae/genética , Animales , Apoptosis , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Terapia Genética , Oclusión de Injerto Vascular/etiología , Humanos , Hiperplasia , Masculino , Miocardio/patología , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Proteína p53 Supresora de Tumor/metabolismo
2.
Int J Mol Med ; 41(1): 184-194, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29115374

RESUMEN

Adipose-tissue derived mesenchymal stem cell (ADSC)-based therapy is a promising option for patients with atherosclerotic conditions, including coronary artery disease. However, the potential differences in the metabolic characteristics between bone marrow-derived mesenchymal stem cells (BMSCs) and ADSCs have remained to be fully elucidated. The present study aimed to compare the metabolic profiles of BMSCs and ADSCs via liquid chromatography quadrupole time-of-flight mass spectrometry. BMSCs and ADSCs obtained from elderly coronary heart disease patients were cultured, and after three passages, supernatants of each cell type were collected and systematically analysed. Substantial differences were detected between the metabolite signatures of ADSCs and BMSCs. In addition, further analysis using partial least-squares discriminant analysis score plots indicated significant differences between the supernatants of the two cell types. The following metabolites were deemed to be responsible for the potential differences in the metabolic characteristics of BMSCs and ADSCs: D-lactic acid, hydroxyindoleacetaldehyde, α-D-glucose, bovinic acid, 9,10-epoxyoctadecenoic acid, glyceraldehyde, phenylpyruvic acid, L-octanoylcarnitine, retinyl ester, α-ketoisovaleric acid, guanidoacetic acid, N-acetylneuraminic acid, imidazoleacetic acid riboside, sphingosine and pseudouridine 5'-phosphate. Based on these findings, there may be significant differences in the following metabolic pathways: The linoleic acid metabolic pathway, galactose metabolism, argentines and proline metabolism, retinol metabolism, glycine and serine metabolism, galactose metabolism, and amino sugar and nucleotide sugar metabolism. In conclusion, substantial differences in metabolic characteristics were detected between BMSCs and ADSCs, which may be associated with the different efficacies of atherosclerosis therapies employing these cell types.


Asunto(s)
Enfermedad Coronaria/metabolismo , Ácido Linoleico/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Anciano , Anciano de 80 o más Años , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Diferenciación Celular/genética , Proliferación Celular/genética , Cromatografía Liquida , Enfermedad Coronaria/genética , Enfermedad Coronaria/patología , Enfermedad Coronaria/terapia , Femenino , Humanos , Ácidos Linoleicos Conjugados/metabolismo , Masculino , Redes y Vías Metabólicas , Persona de Mediana Edad , Osteogénesis/genética
3.
J Cell Mol Med ; 18(11): 2298-310, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25210848

RESUMEN

Sirtuin3 (SIRT3) is an important member of the sirtuin family of protein deacetylases that is localized to mitochondria and linked to lifespan extension in organisms ranging from yeast to humans. As aged cells have less regenerative capacity and are more susceptible to oxidative stress, we investigated the effect of ageing on SIRT3 levels and its correlation with antioxidant enzyme activities. Here, we show that severe oxidative stress reduces SIRT3 levels in young human mesenchymal stromal/stem cells (hMSCs). Overexpression of SIRT3 improved hMSCs resistance to the detrimental effects of oxidative stress. By activating manganese superoxide dismutase (MnSOD) and catalase (CAT), SIRT3 protects hMSCs from apoptosis under stress. SIRT3 expression, levels of MnSOD and CAT, as well as cell survival showed little difference in old versus young hMSCs under normal growth conditions, whereas older cells had a significantly reduced capacity to withstand oxidative stress compared to their younger counterparts. Expression of the short 28 kD SIRT3 isoform was higher, while the long 44 kD isoform expression was lower in young myocardial tissues compared with older ones. These results suggest that the active short isoform of SIRT3 protects hMSCs from oxidative injury by increasing the expression and activity of antioxidant enzymes. The expression of this short isoform decreases in cardiac tissue during ageing, leading to a reduced capacity for the heart to withstand oxidative stress.


Asunto(s)
Apoptosis/genética , Células Madre Mesenquimatosas/metabolismo , Estrés Oxidativo/genética , Sirtuina 3/genética , Envejecimiento , Antioxidantes/metabolismo , Catalasa/genética , Línea Celular , Regulación de la Expresión Génica , Humanos , Células Madre Mesenquimatosas/patología , Especies Reactivas de Oxígeno/metabolismo , Sirtuina 3/biosíntesis , Superóxido Dismutasa/genética
4.
Transpl Immunol ; 30(4): 149-58, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24727088

RESUMEN

OBJECTIVES: Extracellular matrix (ECM) remodelling is a critical aspect of cardiac remodelling following myocardial infarction. Tissue inhibitors of metalloproteinases (TIMPs) are physiological inhibitors of matrix metalloproteinases (MMPs) that degrade the ECM proteins. TIMP-3 is highly expressed in the heart and is markedly downregulated in patients with ischaemic cardiomyopathy. Cell-based gene therapy can enhance the effects of cell transplantation by temporally and spatially regulating the release of the gene product. The purpose of this study was to investigate the role of TIMP-3 gene-transfected vascular smooth muscle cells (VSMCs) in modifying heart structure and function in rats when transplanted 3days after myocardial infarction (MI). METHODS: Anesthetised rats were subjected to coronary artery ligation followed 3days later by thoracotomy and transplantation of TIMP-3 gene-transfected VSMCs, untransfected VSMCs or medium injected directly into the ischaemic myocardium. We assessed left ventricular structure and function by echocardiography and morphometry, and measured the levels of myocardial matrix metalloproteinase-2 and -9 (MMP-2, MMP-9), TIMP-3 and tumour necrosis factor-α (TNF-α) at 4weeks post-myocardial infarction. RESULTS: Transplantation of TIMP-3 gene-transfected VSMCs and untransfected VSMCs significantly decreased scar expansion and ventricular dilatation 25days post-transplantation (4weeks after MI). MMPs and TNF-α levels were reduced in the transplantation groups when compared to the group that was given an injection of medium only. Transplantation of TIMP-3 gene-transfected VSMCs was more effective in preventing progressive cardiac dysfunction, ventricular dilatation and in reducing MMP-2, MMP-9 and TNF-α levels when compared to the transplantation of untransfected VSMCs. CONCLUSIONS: TIMP-3 gene transfection was associated with attenuated left ventricular dilation and recovery of systolic function after MI compared with the control. TIMP-3 transfection enhanced the effects of transplanted VSMCs in rats by inhibiting matrix degradation and inflammatory cytokine expression, leading to improved myocardial remodelling.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Músculo Liso Vascular/trasplante , Infarto del Miocardio/terapia , Inhibidor Tisular de Metaloproteinasa-3/genética , Remodelación Ventricular/fisiología , Animales , Ecocardiografía , Matriz Extracelular/fisiología , Femenino , Corazón/fisiología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocardio/patología , Ratas , Ratas Wistar , Inhibidor Tisular de Metaloproteinasa-3/biosíntesis , Transfección , Factor de Necrosis Tumoral alfa/metabolismo , Función Ventricular Izquierda/fisiología
5.
Rejuvenation Res ; 15(5): 495-506, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22950427

RESUMEN

Mesenchymal stem cell (MSC) transplantation has been proposed as a potential therapeutic approach for ischemic heart disease, but the regenerative capacity of these cells decreases with age. In this study, we genetically engineered old human MSCs (O-hMSCs) with tissue inhibitor of matrix metalloproteinase-3 (TIMP3) and vascular endothelial growth factor (VEGF) and evaluated the effects on the efficacy of cell-based gene therapy in a rat myocardial infarction (MI) model. Cultured O-hMSCs were transfected with TIMP3 (O-TIMP3) or VEGF (O-VEGF) and compared with young hMSCs (Y-hMSCs) and non-transfected O-hMSCs for growth, clonogenic capacity, and differentiation potential. In vivo, rats were subjected to left coronary artery ligation with subsequent injection of Y-hMSCs, O-hMSCs, O-TIMP3, O-VEGF, or medium. Echocardiography was performed prior to and at 1, 2, and 4 weeks after MI. Myocardial levels of matrix metalloproteinase-2 (MMP2), MMP9, TIMP3, and VEGF were assessed at 1 week. Hemodynamics, morphology, and histology were measured at 4 weeks. In vitro, genetically modified O-hMSCs showed no changes in growth, colony formation, or multi-differentiation capacity. In vivo, transplantation with O-TIMP3, O-VEGF, or Y-hMSCs increased capillary density, preserved cardiac function, and reduced infarct size compared to O-hMSCs and medium control. O-TIMP3 and O-VEGF transplantation enhanced TIMP3 and VEGF expression, respectively, in the treated animals. O-hMSCs genetically modified with TIMP3 or VEGF can increase angiogenesis, prevent adverse matrix remodeling, and restore cardiac function to a degree similar to Y-hMSCs. This gene-modified cell therapy strategy may be a promising clinical treatment to rejuvenate stem cells in elderly patients.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Infarto del Miocardio/terapia , Inhibidor Tisular de Metaloproteinasa-3/genética , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Diferenciación Celular , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/metabolismo , Neovascularización Fisiológica/genética , Ratas , Ratas Wistar , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Transfección , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Cell Transplant ; 21(5): 1039-53, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21944319

RESUMEN

After a myocardial infarction (MI), an increase in the cardiac ratio of matrix metalloproteinases (MMPs) relative to their inhibitors (TIMPs) causes extracellular matrix modulation that leads to ventricular dilatation and congestive heart failure. Cell therapy can mitigate these effects. In this study, we tested whether increasing MMP inhibition via cell-based gene transfer of Timp-3 further preserved ventricular morphometry and cardiac function in a rat model of MI. We also measured the effect of treatment timing. We generated MI (coronary artery ligation) in adult rats. Three or 14 days later, we implanted medium (control) or vascular smooth muscle cells transfected with empty vector (VSMCs) or Timp-3 (C-TIMP-3) into the peri-infarct region (n = 15-24/group). We assessed MMP-2 and -9 expression and activity, TIMP-3, and TNF-α expression, cell apoptosis, infarct size and thickness, ventricular morphometry, and cardiac function (by echocardiography). Relative to medium, VSMCs delivered at either time point significantly reduced cardiac expression and activity of MMP-2 and -9, reduced expression of TNF-α, and increased expression of TIMP-3. Cell therapy also reduced apoptosis and scar area, increased infarct thickness, preserved ventricular structure, and reduced functional loss. All these effects were augmented by C-TIMP-3 treatment. Survival and cardiac function were significantly greater when VSMCs or C-TIMP-3 were delivered at 3 (vs. 14) days after MI. Upregulating post-MI cardiac TIMP-3 expression via cell-based gene therapy contributed additional regulation of MMP, TIMP, and TNF-α levels, thereby boosting the structural and functional effects of VSMCs transplanted at 3 or 14 days after an MI in rats. Early treatment may be superior to late, though both are effective.


Asunto(s)
Inhibidores de la Metaloproteinasa de la Matriz , Infarto del Miocardio/terapia , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Enfermedad Aguda , Animales , Apoptosis , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Enfermedad Crónica , Modelos Animales de Enfermedad , Ecocardiografía , Femenino , Ventrículos Cardíacos/fisiopatología , Ventrículos Cardíacos/ultraestructura , Inflamación/metabolismo , Inflamación/patología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Músculo Liso Vascular/citología , Infarto del Miocardio/mortalidad , Infarto del Miocardio/fisiopatología , Ratas , Ratas Wistar , Tasa de Supervivencia , Factores de Tiempo , Inhibidor Tisular de Metaloproteinasa-3/genética , Transfección , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA