Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Obesity (Silver Spring) ; 25(4): 697-703, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28240822

RESUMEN

OBJECTIVE: To investigate the role of secreted frizzled-related protein 3 (Sfrp3) in insulin sensitivity (ISi) and ß-cell function in humans across a spectrum of glucose homeostasis. METHODS: Subjects included those with normal glucose homeostasis (NGT; n = 18), prediabetes (PD; n = 11), or type 2 diabetes (T2D; n=12). Serum and skeletal muscle (SkM) Sfrp3 levels were measured by ELISA and qPCR, respectively, and insulin signaling pathway was assessed by Western blot. IS and ß-cell function were assessed by indices derived from frequently sampled intravenous glucose tolerance test. RESULTS: SkM Sfrp3 mRNA levels were significantly reduced in PD and T2D versus NGT. Similarly, serum Sfrp3 levels tended to be decreased in PD and T2D versus NGT. SkM Sfrp3 mRNA levels correlated negatively with circulating proinflammatory cytokines (IL-6, IFN-γ) and positively with IS. In vitro-differentiated myotubes from lean insulin-sensitive subjects treated with either lipopolysaccharide (LPS) or recombinant IL-6 demonstrated a dose-dependent reduction in Sfrp3 gene expression. Treatment of myotubes with recombinant Sfrp3 restored LPS- and IL-6-induced attenuation of insulin-stimulated Akt phosphorylation. CONCLUSIONS: Inflammation-induced reduction in SkM Sfrp3 expression may contribute to insulin resistance, and this effect may be prevented by addition of exogenous Sfrp3. Thus, Sfrp3 may be a novel target for insulin sensitization.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Resistencia a la Insulina/fisiología , Músculo Esquelético/metabolismo , Estado Prediabético/fisiopatología , Proteínas/metabolismo , Adulto , Glucemia/análisis , Western Blotting , Diferenciación Celular/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Ensayo de Inmunoadsorción Enzimática , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Inflamación/tratamiento farmacológico , Inflamación/fisiopatología , Insulina/sangre , Células Secretoras de Insulina/fisiología , Interferón gamma/sangre , Interleucina-6/sangre , Masculino , Persona de Mediana Edad , Fibras Musculares Esqueléticas/metabolismo , Fosforilación/efectos de los fármacos , Estado Prediabético/tratamiento farmacológico , Proteínas/uso terapéutico , ARN Mensajero/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/fisiología
2.
J Mol Cell Cardiol ; 48(6): 1255-65, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20096288

RESUMEN

Bone morphogenetic protein (BMP) signaling regulates embryonic development of many organ systems and defective BMP signaling has been implicated in adult disorders of many of these systems. However, its relevance in cardiac disease has not been reported. Here we demonstrate for the first time that Bmp4 activity promotes cellular apoptosis following ischemia-reperfusion (I/R) injury induced myocardial infarction (MI). Bmp4 heterozygous null mice (Bmp4(+/)(-)) demonstrated reduced infarct size, less myocardial apoptosis and down-regulation of pro-apoptotic proteins relative to wild-type mice following I/R injury. This was associated with reduction in I/R induced BMP4 levels in the left ventricular infarcted region. Furthermore, treatment of neonatal cardiomyocytes with BMP4 resulted in time and dose-dependent increase in cellular apoptosis and activation of the JNK MAP kinase pathway. In contrast, while JNK activation was significantly attenuated in Bmp4(+/)(-) mice and following Smad1 inhibition in myocytes, inhibition of JNK with a specific inhibitory peptide, TAT-JBD(20,) blocked BMP4 induced apoptosis. In vivo treatment of mice with Noggin, an endogenous extracellular BMP antagonist, or dorsomorphin, a small molecule inhibitor of BMP signaling, reduced infarct size, and inhibited pro-apoptotic signaling accompanied by an inhibition of Smad1 phosphorylation and JNK activation. These studies identify a novel role for Bmp4 in the pathogenesis of myocardial infarction and illustrate the use of a small molecule inhibitor of BMP signaling for treatment of acute I/R injury.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Daño por Reperfusión Miocárdica/patología , Transducción de Señal , Animales , Apoptosis , Proteínas Portadoras/metabolismo , Supervivencia Celular , Heterocigoto , Humanos , Masculino , Ratones , Ratones Transgénicos , Estrés Oxidativo , Oxígeno/química , Proteínas Recombinantes/química
3.
Int Immunol ; 21(7): 817-29, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19515797

RESUMEN

Myocardial ischemia with subsequent reperfusion (MI/R) can lead to significant myocardial damage. Ischemia initiates inflammation at the blood-microvascular endothelial cell interface and contributes significantly to both acute injury and repair of the damaged tissue. We have found that MI/R injury in mice is associated with a cellular immune response to troponin. Myocardial cells exclusively synthesize troponin and release the troponin into the bloodstream following injury. Mucosally administered proteins induce T cells that secrete anti-inflammatory cytokines such as IL-10 and transforming growth factor beta at the anatomical site where the protein localizes. We found that nasal administration of the three subunits of troponin (C, I and T isoforms), given prior to or 1 h following MI/R, decreased infarct size by 40% measured 24 h later. At 1.5 months following MI/R, there was a 50% reduction in infarct size and improvement in cardiac function as measured by echocardiography. Protection was associated with a reduction of cellular immunity to troponin. Immunohistochemistry demonstrated increased IL-10 and reduced IFN-gamma in the area surrounding the ischemic infarct following nasal troponin. Adoptive transfer of CD4+ T cells to mice from nasally troponin-treated mice 1 h after the MI/R decreased infarct size by 72%, whereas CD4+ T cells from IL-10-/- mice or nasally BSA-treated mice had no effect. Our results demonstrate that IL-10-secreting CD4+ T cells induced by nasal troponin reduce injury following MI/R. Modulation of cardiac inflammation by nasal troponin provides a novel treatment to decrease myocardial damage and enhance recovery after myocardial ischemia.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Interferón gamma/inmunología , Interleucina-10/inmunología , Daño por Reperfusión Miocárdica/prevención & control , Troponina/administración & dosificación , Administración Intranasal , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Interferón gamma/antagonistas & inhibidores , Interleucina-10/agonistas , Interleucina-10/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Daño por Reperfusión Miocárdica/inmunología , Daño por Reperfusión Miocárdica/patología , Miocardio/inmunología , Miocardio/patología , Isoformas de Proteínas/inmunología , Troponina/inmunología , Vacunación
4.
J Mol Cell Cardiol ; 43(5): 580-92, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17920074

RESUMEN

Heme-oxygenase-1 (HO-1), a stress-inducible protein, is an important cytoprotective agent against ischemia/reperfusion (I/R) injury. However, the role of downstream mediators involved in HO-1-induced cytoprotection is not clear. In the current study we investigated the role of biliverdin reductase, an enzyme involved in the conversion of HO-1-derived biliverdin into bilirubin and the PI3K/Akt pathway in mediating the cytoprotective effects of HO-1 against hypoxia and reoxygenation (H/R) injury in vitro and in vivo. H9c2 cardiomyocytes were transfected with a plasmid expressing HO-1 or LacZ and exposed to 24 h of hypoxia followed by 12 h of reoxygenation. At the end of reoxygenation, reactive oxygen species generation was determined using CM-H(2)DCFDA dye and apoptosis was assessed by TUNEL, caspase activity and Bad phosphorylation. p85 and Akt phosphorylation were determined using cell-based ELISA and phospho-specific antibodies, respectively. HO-1 overexpression increased phosphorylation of the regulatory subunit of the PI3K (p85alpha) and downstream effector Akt in H9c2 cells, leading to decreased ROS and apoptosis. Furthermore, cardiac expression of HO-1 increased basal phosphorylated Akt levels and decreased infarct size in response to LAD ligation and release induced I/R injury. Conversely, PI3K inhibition reversed the effects of HO-1 on Akt phosphorylation, cell death and infarct size. In addition, knockdown of biliverdin reductase (BVR) expression with siRNA attenuated HO-1-induced Akt phosphorylation and increased H/R-induced apoptosis of H9c2 cells. Co-immunoprecipitation revealed protein-protein interaction between BVR and the phosphorylated p85 subunit of the PI3 kinase. Taken together, these results suggest that the enzyme biliverdin reductase plays an important role in mediating cytoprotective effects of HO-1. This effect is mediated, at least in part, via interaction with and activation of the PI3K/Akt pathway.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Hipoxia/prevención & control , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Daño por Reperfusión/prevención & control , Animales , Hemo-Oxigenasa 1/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Consumo de Oxígeno , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo
5.
Am J Physiol Heart Circ Physiol ; 293(1): H48-59, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17322421

RESUMEN

We reported previously that predelivery of heme oxygenase-1 (HO-1) gene to the heart by adeno-associated virus-2 (AAV-2) markedly reduces ischemia and reperfusion (I/R)-induced myocardial injury. However, the effect of preemptive HO-1 gene delivery on long-term survival and prevention of postinfarction heart failure has not been determined. We assessed the effect of HO-1 gene delivery on long-term survival, myocardial function, and left ventricular (LV) remodeling 1 yr after myocardial infarction (MI) using echocardiographic imaging, pressure-volume (PV) analysis, and histomorphometric approaches. Two groups of Lewis rats were injected with 2 x 10(11) particles of AAV-LacZ (control) or AAV-human HO-1 (hHO-1) in the anterior-posterior apical region of the LV wall. Six weeks after gene transfer, animals were subjected to 30 min of ischemia by ligation of the left anterior descending artery followed by reperfusion. Echocardiographic measurements and PV analysis of LV function were obtained at 2 wk and 12 mo after I/R. One year after acute MI, mortality was markedly reduced in the HO-1-treated animals compared with the LacZ-treated animals. PV analysis demonstrated significantly enhanced LV developed pressure, elevated maximal dP/dt, and lower end-diastolic volume in the HO-1 animals compared with the LacZ animals. Echocardiography showed a larger apical anterior-to-posterior wall ratio in HO-1 animals compared with LacZ animals. Morphometric analysis revealed extensive myocardial scarring and fibrosis in the infarcted LV area of LacZ animals, which was reduced by 62% in HO-1 animals. These results suggest that preemptive HO-1 gene delivery may be useful as a therapeutic strategy to reduce post-MI LV remodeling and heart failure.


Asunto(s)
Modelos Animales de Enfermedad , Terapia Genética/métodos , Hemo Oxigenasa (Desciclizante)/uso terapéutico , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/terapia , Disfunción Ventricular Izquierda/prevención & control , Disfunción Ventricular Izquierda/fisiopatología , Animales , Hemo Oxigenasa (Desciclizante)/genética , Humanos , Masculino , Infarto del Miocardio/complicaciones , Ratas , Ratas Endogámicas Lew , Análisis de Supervivencia , Tasa de Supervivencia , Transfección/métodos , Resultado del Tratamiento , Disfunción Ventricular Izquierda/etiología
6.
J Am Coll Cardiol ; 47(3): 635-43, 2006 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-16458149

RESUMEN

OBJECTIVES: We assessed the hypothesis that overexpression of the antioxidant enzyme heme oxygenase (HO)-1 may protect against chronic recurrent ischemia/reperfusion injury. BACKGROUND: Multiple and recurring episodes of myocardial ischemia can result in significant myocardial damage, including myocyte death, fibrosis, and wall thinning, leading to impaired ventricular function and cardiac failure. METHODS: In this study we used a closed-chest rodent model of chronic recurring myocardial ischemia and reperfusion to investigate the efficacy of pre-emptive gene therapy in overexpressing the antioxidant enzyme HO-1, using adeno-associated virus (AAV)-2 as the delivery vector. RESULTS: We show that constitutive overexpression of HO-1 can prevent myocardial wall thinning, inflammation, fibrosis, and deterioration of cardiac function (as measured by echocardiography, histology, and immunohistochemistry) induced by repeated transient myocardial ischemia and reperfusion injury. With HO-1 therapy, there was a significant reduction in apoptosis as determined by levels of markers of survival proteins and terminal deoxynucleotidyltransferase dUTP nick end-labeling staining. This prevention of tissue damage was also associated with reduction in superoxide generation. CONCLUSIONS: Taken together we provide the first evidence of the therapeutic efficacy of pre-emptive AAV-HO-1 delivery for prevention against multiple ischemic injury. This approach protects myocytes by simultaneously activating protective response and inhibiting pathological left ventricular remodeling and, therefore, may be a useful cardio-protective strategy for patients with coronary artery disease at a high risk for recurrent myocardial ischemia.


Asunto(s)
Terapia Genética , Hemo-Oxigenasa 1/genética , Daño por Reperfusión Miocárdica/prevención & control , Animales , Apoptosis , Dependovirus/genética , Vectores Genéticos , Hemo-Oxigenasa 1/metabolismo , Hemo-Oxigenasa 1/uso terapéutico , Etiquetado Corte-Fin in Situ , Masculino , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/metabolismo , Miocardio/patología , Ratas , Ratas Sprague-Dawley , Recurrencia , Superóxidos/metabolismo
7.
FASEB J ; 20(2): 207-16, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16449792

RESUMEN

We reported previously that predelivery of the anti-oxidant gene heme oxygenase-1 (HO-1) to the heart by adeno associated virus (AAV) markedly reduces injury after acute myocardial infarction (MI). However, the effect of HO-1 gene delivery on postinfarction recovery has not been investigated. In the current study, we assessed the effect of HO-1 gene delivery on post-MI left ventricle (LV) remodeling and function using echocardiographic imaging and histomorphometric approaches. Two groups of Sprague-Dawley rats were injected with 4 x 10(11) particles of AAV-LacZ (control) or AAV-hHO-1 in the LV wall. Eight wk after gene transfer, the animals were subjected to 30 min of ischemia by ligation of left anterior descending artery (LAD) followed by reperfusion. Echocardiographic measurements were obtained in a blinded fashion prior and at 1.5 and 3 months after I/R. Ejection fraction (EF) was reduced by 13% and 40% in the HO-1 and LacZ groups, respectively at 1.5 months after MI. Three months after MI, EF recovered fully in the HO-1, but only partially in the LacZ-treated animals. Post-MI LV dimensions were markedly increased and the anterior wall was markedly thinned in the LacZ-treated animals compared with the HO-1-treated animals. Significant myocardial scarring and fibrosis were observed in the LacZ-group in association with elevated levels of interstitial collagen I and III and MMP-2 activity. Post-MI myofibroblast accumulation was reduced in the HO-1-treated animals, and retroviral overexpression of HO-1 reduced proliferation of isolated cardiac fibroblasts. Our data indicate that rAAV-HO-1 gene transfer markedly reduces fibrosis and ventricular remodeling and restores LV function and chamber dimensions after myocardial infarction.


Asunto(s)
Terapia Genética , Hemo-Oxigenasa 1/metabolismo , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/terapia , Remodelación Ventricular/fisiología , Animales , Proliferación Celular , Células Cultivadas , Colágeno/metabolismo , Fibroblastos , Fibrosis/genética , Fibrosis/patología , Fibrosis/terapia , Regulación Enzimológica de la Expresión Génica , Ventrículos Cardíacos/anatomía & histología , Hemo-Oxigenasa 1/genética , Humanos , Masculino , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/terapia , Miocardio/citología , Miocardio/metabolismo , Ratas , Ratas Sprague-Dawley , Función Ventricular
9.
Hypertension ; 46(1): 7-18, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15956118

RESUMEN

Endothelial dysfunction and cell loss are prominent features in cardiovascular disease. Endothelial progenitor cells (EPCs) originating from the bone marrow play a significant role in neovascularization of ischemic tissues and in re-endothelialization of injured blood vessels. Several studies have shown the therapeutic potential of EPC transplantation in rescue of tissue ischemia and in repair of blood vessels and bioengineering of prosthetic grafts. Recent small-scale trials have provided preliminary evidence of feasibility, safety, and efficacy in patients with myocardial and critical limb ischemia. However, several studies have shown that age and cardiovascular disease risk factors reduce the availability of circulating EPCs (CEPCs) and impair their function to varying degrees. In addition, the relative scarcity of CEPCs limits the ability to expand these cells in sufficient numbers for some therapeutic applications. Priority must be given to the development of strategies to enhance the number and improve the function of CEPCs. Furthermore, alternative sources of EPC such as chord blood need to be explored. Strategies for improvement of cell adhesion, survival, and prevention of cell senescence are also essential to ensure therapeutic viability. Genetic engineering of EPCs may be a useful approach to developing these cells into efficient therapeutic tools. In the clinical arena there is pressing need to standardize the protocols for isolation, culture, and therapeutic application of EPC. Large-scale multi-center randomized trials are required to evaluate the long-term safety and efficacy of EPC therapy. Despite these hurdles, the outlook for EPC-based therapy for cardiovascular disease is promising.


Asunto(s)
Enfermedades Cardiovasculares/cirugía , Células Endoteliales/trasplante , Trasplante de Células Madre , Envejecimiento , Animales , Separación Celular , Ingeniería Genética , Humanos , Terapia Recuperativa , Trasplante de Células Madre/efectos adversos
10.
Trends Mol Med ; 11(5): 240-50, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15882612

RESUMEN

Recent advances in understanding the molecular and cellular basis of cardiovascular diseases, together with the availability of tools for genetic manipulation of the cardiovascular system, offer possibilities for new treatments. Gene therapies have demonstrated potential usefulness for treating complex cardiovascular diseases, such as hypertension, atherosclerosis and myocardial ischemia, in various animal models. Some of these experimental therapies are now undergoing clinical evaluation in patients with cardiovascular disease. However, the successful transition of these therapies into mainstream clinical practice awaits further improvements to vector platforms and delivery tools and the documentation of clinical feasibility, safety and efficacy through multi-center randomized trials.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Animales , Enfermedades Cardiovasculares/metabolismo , Silenciador del Gen , Marcación de Gen/métodos , Técnicas de Transferencia de Gen/tendencias , Terapia Genética/tendencias , Humanos
11.
Proc Natl Acad Sci U S A ; 101(33): 12282-7, 2004 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-15302924

RESUMEN

Ischemia and reperfusion represent major mechanisms of tissue injury and organ failure. The timing of administration and the duration of action limit current treatment approaches using pharmacological agents. In this study, we have successfully developed a preemptive strategy for tissue protection using an adenoassociated vector system containing erythropoietin hypoxia response elements for ischemia-regulated expression of the therapeutic gene human heme-oxygenase-1 (hHO-1). We demonstrate that a single administration of this vector several weeks in advance of ischemia/reperfusion injury to multiple tissues such as heart, liver, and skeletal muscle yields rapid and timely induction of hHO-1 during ischemia that resulted in dramatic reduction in tissue damage. In addition, overexpression of therapeutic transgene prevented long-term pathological tissue remodeling and normalized tissue function. Application of this regulatable system using an endogenous physiological stimulus for expression of a therapeutic gene may be a feasible strategy for protecting tissues at risk of ischemia/reperfusion injury.


Asunto(s)
Terapia Genética/métodos , Hemo Oxigenasa (Desciclizante)/genética , Hipoxia/genética , Daño por Reperfusión/terapia , Animales , Línea Celular , Citocinas/genética , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes , Hemo-Oxigenasa 1 , Humanos , Hígado/enzimología , Hígado/inmunología , Luciferasas/biosíntesis , Luciferasas/genética , Proteínas Luminiscentes/biosíntesis , Proteínas Luminiscentes/genética , Masculino , Proteínas de la Membrana , Músculo Esquelético/enzimología , Músculo Esquelético/lesiones , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/terapia , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Daño por Reperfusión/enzimología , Daño por Reperfusión/genética
12.
Arterioscler Thromb Vasc Biol ; 24(10): 1761-74, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15308553

RESUMEN

Most common cardiovascular diseases are accompanied by endothelial dysfunction. Because of its predominant role in the pathogenesis of cardiovascular disease, the vascular endothelium is an attractive therapeutic target. The identification of promoter sequences capable of rendering endothelial-specific transgene expression together with the recent development of vectors with enhanced tropism for endothelium may offer opportunities for the design of new strategies for modulation of endothelial function. Such strategies may be useful in the treatment of chronic diseases such as hypertension, atherosclerosis, and ischemic artery disease, as well as in acute myocardial infarction and during open heart surgery for prevention of ischemia and reperfusion (I/R)-induced injury. The recent identification of putative endothelial progenitor cells in peripheral blood may allow the design of autologous cell-based strategies for neovascularization of ischemic tissues and for the repair of injured blood vessels and bioengineering of vascular prosthesis. "Proof-of-concept" for some of these strategies has been established in animal models of cardiovascular disease. However the successful translation of these novel strategies into clinical application will require further developments in vector and delivery technologies. Further characterization of the processes involved in mobilization, migration, homing, and incorporation of endothelial progenitor cells into the target tissues is necessary, and the optimal conditions for therapeutic application of these cells need to be defined and standardized.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Endotelio Vascular/química , Endotelio Vascular/metabolismo , Terapia Genética/métodos , Animales , Células Endoteliales/química , Células Endoteliales/metabolismo , Endotelio Vascular/patología , Técnicas de Transferencia de Gen , Humanos
14.
FASEB J ; 18(6): 648-63, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15054087

RESUMEN

Heart disease remains the prevalent cause of premature death and accounts for a significant proportion of all hospital admissions. Recent developments in understanding the molecular mechanisms of myocardial disease have led to the identification of new therapeutic targets, and the availability of vectors with enhanced myocardial tropism offers the opportunity for the design of gene therapies for both protection and rescue of the myocardium. Genetic therapies have been devised to treat complex diseases such as myocardial ischemia, heart failure, and inherited myopathies in various animal models. Some of these experimental therapies have made a successful transition to clinical trial and are being considered for use in human patients. The recent isolation of endothelial and cardiomyocyte precursor cells from adult bone marrow may permit the design of strategies for repair of the damaged heart. Cell-based therapies may have potential application in neovascularization and regeneration of ischemic and infarcted myocardium, in blood vessel reconstruction, and in bioengineering of artificial organs and prostheses. We expect that advances in the field will lead to the development of safer and more efficient vectors. The advent of genomic screening technology should allow the identification of novel therapeutic targets and facilitate the detection of disease-causing polymorphisms that may lead to the design of individualized gene and cell-based therapies.


Asunto(s)
Terapia Genética , Cardiopatías/terapia , Trasplante de Células Madre , Enfermedad de la Arteria Coronaria/terapia , Endotelio Vascular/citología , Cardiopatías Congénitas/terapia , Cardiopatías/metabolismo , Cardiopatías/fisiopatología , Humanos , Contracción Miocárdica , Isquemia Miocárdica/terapia , Miocardio/metabolismo , Ingeniería de Tejidos
15.
Biochem Biophys Res Commun ; 313(3): 528-33, 2004 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-14697221

RESUMEN

Intramyocardial injection of adeno-associated virus (AAV) has been shown to be an effective strategy for cardiac gene delivery. This approach leads to long-term gene expression in the heart, offering the possibility of chronic gene therapy. However, the long-term safety of this approach with regard to vector bio-distribution and extracardiac transgene expression has not been evaluated. To examine these issues, 8-week-old male Sprague-Dawley rats were injected intramyocardially with either 4x10(11) particles of AAV-2-lacZ or saline at five locations in the anterioposterior apical region of the left ventricle. Animals were sacrificed at 3 and 6 months after gene transfer, tissues were harvested and analyzed for lacZ expression by semi-quantitative RT-PCR and beta-galactosidase activity using X-gal staining. We observed high level of transgene expression in the myocardium at 3 months after gene transfer, which persisted up to 6 months of follow-up. Also, significantly we detected lacZ expression and beta-galactosidase activity in extracardiac tissues such as liver, kidney, and testes at 6 months. More significantly, late transgene expression was detected in cellular elements of the seminiferous tubule, including Sertoli cells and spermatogonia like cells. These data demonstrate the efficacy of AAV-2 delivery for long-term myocardial gene therapy, but raise concerns about the possibility of ectopic transgene expression and germ cell line infection.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Miocardio/metabolismo , Animales , Southern Blotting , ADN/metabolismo , ADN Complementario/metabolismo , Inmunohistoquímica , Operón Lac , Masculino , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transgenes , beta-Galactosidasa/metabolismo
16.
Hypertension ; 39(5): 969-75, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12019278

RESUMEN

Our studies have established that a single intracardiac administration of the retroviral vector containing angiotensin II type I receptor antisense gene causes prolonged antihypertensive actions in the spontaneously hypertensive rat. These results suggest that antisense gene therapy is a conceptually valid strategy for the control of hypertension at the genetic level. To evaluate whether attenuation of the pathophysiological aspects of hypertension are dependent on the blood pressure lowering actions of antisense gene therapy, we chose the renin transgenic rat as a hypertensive animal model and cardiac hypertrophy as the hypertension-associated pathophysiology. A single intracardiac administration of the retroviral vector containing angiotensin II type I receptor antisense in the neonatal rat resulted in long-term expression of the antisense transgene in various cardiovascular-relevant tissues, including the heart. This expression was associated with a significant attenuation of cardiac hypertrophy despite its failure to normalize high blood pressure. Developmental studies indicated that cardiac hypertrophy was evident as early as 16 days of age in viral vector-treated control transgenic rats, despite these animals exhibiting normal blood pressure. These observations demonstrate that, in the renin-transgenic rat, the onset of cardiac hypertrophy occurs during development and is prevented without normalization of high blood pressure. Collectively, these results provide further proof of the concept and indicate that antisense gene therapy could successfully target the local tissues' renin-angiotensin system to produce beneficial cardiovascular outcomes.


Asunto(s)
Presión Sanguínea/fisiología , Cardiomegalia/terapia , Terapia Genética , Receptores de Angiotensina/genética , Actinas/genética , Animales , Animales Modificados Genéticamente , Factor Natriurético Atrial/genética , Cardiomegalia/genética , Cardiomegalia/fisiopatología , ADN sin Sentido/genética , Femenino , Expresión Génica , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1 , Renina/genética , Retroviridae/genética , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...