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












Base de datos
Intervalo de año de publicación
1.
Cell Signal ; 76: 109783, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32956789

RESUMEN

Pathological neointimal growth can develop in patients as a result of vascular injury following percutaneous coronary intervention and coronary artery bypass grafting using autologous saphenous vein, leading to arterial or vein graft occlusion. Neointima formation driven by intimal hyperplasia occurs as a result of a complex interplay between molecular and cellular processes involving different cell types including endothelial cells, vascular smooth muscle cells and various inflammatory cells. Therefore, understanding the intercellular communication mechanisms underlying this process remains of fundamental importance in order to develop therapeutic strategies to preserve endothelial integrity and vascular health post coronary interventions. Extracellular vesicles (EVs), including microvesicles and exosomes, are membrane-bound particles secreted by cells which mediate intercellular signalling in physiological and pathophysiological states, however their role in neointima formation is not fully understood. The purification and characterization techniques currently used in the field are associated with many limitations which significantly hinder the ability to comprehensively study the role of specific EV types and make direct functional comparisons between EV subpopulations. In this review, the current knowledge focusing on EV signalling in neointima formation post vascular injury is discussed.


Asunto(s)
Células Endoteliales , Vesículas Extracelulares/metabolismo , Músculo Liso Vascular , Miocitos del Músculo Liso , Neointima/metabolismo , Lesiones del Sistema Vascular , Animales , Células Cultivadas , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Lesiones del Sistema Vascular/metabolismo , Lesiones del Sistema Vascular/patología
2.
Sci Rep ; 9(1): 3154, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30816157

RESUMEN

The counter-regulatory axis, Angiotensin Converting Enzyme 2, Angiotensin-(1-7), Mas receptor (ACE2/Ang-1-7/MasR), of the renin angiotensin system (RAS) is a potential therapeutic target in stroke, with Ang-(1-7) reported to have neuroprotective effects in pre-clinical stroke models. Here, an extensive investigation of the functional and mechanistic effects of Ang-(1-7) was performed in a rodent model of stroke. Using longitudinal magnetic resonance imaging (MRI) it was observed that central administration of Ang-(1-7) following transient middle cerebral artery occlusion (MCAO) increased the amount of tissue salvage compared to reperfusion alone. This protective effect was not due to early changes in blood brain barrier (BBB) permeability, microglia activation or inflammatory gene expression. However, increases in NADPH oxidase 1 (Nox1) mRNA expression were observed in the treatment group compared to control. In order to determine whether Ang-(1-7) has direct cerebrovascular effects, laser speckle contrast imaging (LSCI) was performed to measure dynamic changes in cortical perfusion following reperfusion. Delivery of Ang-(1-7) did not have any effect on cortical perfusion following reperfusion however; it showed an indication to prevent the 'steal phenomenon' within the contralateral hemisphere. The comprehensive series of studies have demonstrated a moderate protective effect of Ang-(1-7) when given alongside reperfusion to increase tissue salvage.


Asunto(s)
Angiotensina I/farmacología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , NADPH Oxidasa 1/genética , Fragmentos de Péptidos/farmacología , Peptidil-Dipeptidasa A/genética , Accidente Cerebrovascular/tratamiento farmacológico , Enzima Convertidora de Angiotensina 2 , Animales , Barrera Hematoencefálica/efectos de los fármacos , Medios de Contraste/farmacología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Infarto de la Arteria Cerebral Media/diagnóstico por imagen , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/fisiopatología , Inflamación/diagnóstico por imagen , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/patología , Imagen por Resonancia Magnética , Microglía/efectos de los fármacos , Microglía/patología , Arteria Cerebral Media/diagnóstico por imagen , Arteria Cerebral Media/efectos de los fármacos , Fármacos Neuroprotectores , Proto-Oncogenes Mas , ARN Mensajero/genética , Ratas , Sistema Renina-Angiotensina/genética , Reperfusión/métodos , Accidente Cerebrovascular/diagnóstico por imagen , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/patología
3.
Gene Ther ; 15(6): 443-51, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18004401

RESUMEN

Targeted delivery of biological agents to atherosclerotic plaques may provide a novel treatment and/or useful tool for imaging of atherosclerosis in vivo. However, there are no known viral vectors that possess the desired tropism. Two plaque-targeting peptides, CAPGPSKSC (CAP) and CNHRYMQMC (CNH) were inserted into the capsid of adeno-associated virus 2 (AAV2) to assess vector retargeting. AAV2-CNH produced significantly higher levels of transduction than unmodified AAV2 in human, murine and rat endothelial cells, whereas transduction of nontarget HeLa cells was unaltered. Transduction studies and surface plasmon resonance suggest that AAV2-CNH uses membrane type 1 matrix metalloproteinase as a surface receptor. AAV2-CAP only produced higher levels of transduction in rat endothelial cells, possibly because the virus was found to be affected by proteasomal degradation. In vivo substantially higher levels of both peptide-modified AAV2 vectors was detected in the brachiocephalic artery (site of advanced atherosclerotic plaques) and aorta, whereas reduced levels were detected in all other organs examined. These results suggest that in the AAV2 platform the peptides are exposed on the capsid surface in a way that enables efficient receptor binding and so creates effective atherosclerotic plaque targeted vectors.


Asunto(s)
Aterosclerosis/terapia , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Animales , Apolipoproteínas E/genética , Línea Celular , Dependovirus/metabolismo , Marcación de Gen , Ingeniería Genética , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Heparina/metabolismo , Humanos , Metaloproteinasa 1 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fragmentos de Péptidos/genética , Unión Proteica , Ratas , Resonancia por Plasmón de Superficie , Transducción Genética/métodos , Transgenes
4.
Gene Ther ; 11(16): 1296-300, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15292916

RESUMEN

Adenovirus (Ad)-mediated gene delivery is a promising approach for genetic manipulation of the vasculature and is being used in both preclinical models and clinical trials. However, safety concerns relating to infection of nontarget tissue and the poor infectivity of vascular cells compared to other cell types necessitates Ad vector refinement. Here, we combine a transductional targeting approach to improve vascular cell infectivity through RGD peptide insertion into adenovirus fibers, combined with transcriptional targeting to endothelial cells using a approximately 1 kb fragment of the fms-like tyrosine kinase receptor-1 (FLT-1) promoter. Single- and double-modified vectors were characterized in human cell lines that either support or have silenced FLT-1 expression. In rat hepatocytes and endothelial cells, the double modification substantially shifted transduction profiles toward vascular endothelial cells. Furthermore, in intact aortae derived from spontaneously hypertensive rats that display enhanced alphav integrin expression on dysfunctional endothelium, enhanced levels of transduction were observed using the double-modified vector but not in aortae derived from normotensive control rats. Our data indicate that Ad-mediated transduction can be beneficially modified in vitro and in vivo by combining fiber modification and a cell-selective promoter within a single-component vector system.


Asunto(s)
Infecciones por Adenoviridae/genética , Adenoviridae/genética , Marcación de Gen/métodos , Animales , Aorta/fisiopatología , Línea Celular , Células Cultivadas , Células Endoteliales/fisiología , Terapia Genética/métodos , Vectores Genéticos/genética , Hepatocitos/fisiología , Humanos , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Proteínas Tirosina Quinasas Receptoras , Receptores Virales/genética , Transcripción Genética/genética , Transducción Genética/métodos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
5.
Mol Ther ; 4(6): 534-42, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11735337

RESUMEN

Adenovirus type 5 (Ad) based vectors transduce vascular endothelial cells (EC) and have been widely used for vascular gene transfer. However, many cell types express the Ad receptor (cox-sackievirus adenovirus receptor; CAR), preventing selective EC infection and precluding clinical use. We previously isolated the human EC-binding peptides SIGYPLP and LSNFHSS by phage display and demonstrated by means of a bispecific antibody that SIGYPLP directs efficient, high-level, EC-selective Ad-mediated gene transfer. We now generate genetically modified Ad fiber proteins with selective EC tropism by engineering these peptides into the HI loop of the Ad fiber. SIGYPLP, but not LSNFHSS, enhanced EC selectivity, demonstrating maintenance of peptide-cell binding fidelity upon incorporation into virions. Combining fiber mutations that block CAR binding (detargeting) with SIGYPLP insertion (retargeting) generated a novel Ad vector, AdKO1SIG, in a single component system. AdKO1SIG demonstrated efficient and selective tropism for EC compared with control Ad vectors. This is the first demonstration of genetic incorporation of a novel, mammalian, cell-selective ligand that retains its targeting fidelity in the Ad fiber HI loop, in combination with point mutations that abolish fiber-CAR interaction. This study demonstrates the potential for improving the cell-selectivity and safety of adenoviral vectors.


Asunto(s)
Adenoviridae/fisiología , Endotelio Vascular/virología , Terapia Genética/métodos , Fragmentos de Péptidos/genética , Receptores Virales/metabolismo , Sitios de Unión , Unión Competitiva , Células Cultivadas , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Cartilla de ADN/química , Citometría de Flujo , Marcación de Gen/métodos , Vectores Genéticos , Proteínas Fluorescentes Verdes , Humanos , Immunoblotting , Proteínas Luminiscentes/metabolismo , Plásmidos/genética , Receptores Virales/genética , Linfocitos T/metabolismo , Transducción Genética , Transfección
6.
Circulation ; 104(13): 1538-43, 2001 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-11571249

RESUMEN

BACKGROUND: Mitogenic stimuli present at the site of coronary arterial balloon injury contribute to the progression and development of a restenotic lesion, many signaling through a common pathway involving the small G protein p21(ras). Our aim was to demonstrate in biochemical studies that farnesyl protein transferase inhibitor III (FPTIII) is an inhibitor of p21(ras) processing and that when it is given locally in vivo at the site of coronary balloon injury in a porcine model, it can inhibit neointima formation. METHODS AND RESULTS: FPTIII (1 to 25 micromol/L) concentration-dependently reduced p21(ras) levels in porcine coronary artery smooth muscle cell membranes. FPTIII also prevented p42/p44 MAPK activation and DNA synthesis in response to platelet-derived growth factor in these cells at a concentration of 25 micromol/L. Application of 25 micromol/L FPTIII locally for 15 minutes to balloon-injured porcine coronary arteries in vivo prevented neointima formation assessed at 4 weeks, reduced proteoglycan deposition, and inhibited adventitial hypertrophy. Coronary arteries from FPTIII-treated pigs had no deterioration in contraction or in endothelium-dependent relaxation. CONCLUSIONS: The study demonstrates in the pig that short-term local delivery of inhibitors of p21(ras)-dependent mitogenic signal transduction prevents restenosis after balloon angioplasty.


Asunto(s)
Transferasas Alquil y Aril/antagonistas & inhibidores , Angioplastia Coronaria con Balón/efectos adversos , Inhibidores Enzimáticos/uso terapéutico , Organofosfonatos/uso terapéutico , Complicaciones Posoperatorias/prevención & control , Animales , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/administración & dosificación , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Organofosfonatos/administración & dosificación , Transducción de Señal/efectos de los fármacos , Porcinos , Túnica Íntima/efectos de los fármacos , Túnica Íntima/patología , Enfermedades Vasculares/etiología , Enfermedades Vasculares/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...