Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Circulation ; 133(21): 2050-65, 2016 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-27052414

RESUMEN

BACKGROUND: Phenotypic switching of vascular smooth muscle cells from a contractile to a synthetic state is implicated in diverse vascular pathologies, including atherogenesis, plaque stabilization, and neointimal hyperplasia. However, very little is known about the role of long noncoding RNA (lncRNA) during this process. Here, we investigated a role for lncRNAs in vascular smooth muscle cell biology and pathology. METHODS AND RESULTS: Using RNA sequencing, we identified >300 lncRNAs whose expression was altered in human saphenous vein vascular smooth muscle cells following stimulation with interleukin-1α and platelet-derived growth factor. We focused on a novel lncRNA (Ensembl: RP11-94A24.1), which we termed smooth muscle-induced lncRNA enhances replication (SMILR). Following stimulation, SMILR expression was increased in both the nucleus and cytoplasm, and was detected in conditioned media. Furthermore, knockdown of SMILR markedly reduced cell proliferation. Mechanistically, we noted that expression of genes proximal to SMILR was also altered by interleukin-1α/platelet-derived growth factor treatment, and HAS2 expression was reduced by SMILR knockdown. In human samples, we observed increased expression of SMILR in unstable atherosclerotic plaques and detected increased levels in plasma from patients with high plasma C-reactive protein. CONCLUSIONS: These results identify SMILR as a driver of vascular smooth muscle cell proliferation and suggest that modulation of SMILR may be a novel therapeutic strategy to reduce vascular pathologies.


Asunto(s)
Proliferación Celular/fisiología , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/fisiología , ARN Largo no Codificante/fisiología , Proteínas de Caenorhabditis elegans , Células Cultivadas , Técnicas de Silenciamiento del Gen , Humanos , Músculo Liso Vascular/citología , Vena Safena/citología , Vena Safena/fisiología
2.
Circ Res ; 111(3): 290-300, 2012 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-22715469

RESUMEN

RATIONALE: Despite improved understanding of the underlying genetics, pulmonary arterial hypertension (PAH) remains a severe disease. Extensive remodeling of small pulmonary arteries, including proliferation of pulmonary artery smooth muscle cells (PASMCs), characterizes PAH. MicroRNAs (miRNAs) are noncoding RNAs that have been shown to play a role in vascular remodeling. OBJECTIVE: We assessed the role of miR-145 in PAH. METHODS AND RESULTS: We localized miR-145 in mouse lung to smooth muscle. Using quantitative PCR, we demonstrated increased expression of miR-145 in wild-type mice exposed to hypoxia. PAH was evaluated in miR-145 knockout and mice treated with anti-miRs via measurement of systolic right ventricular pressure, right ventricular hypertrophy, and percentage of remodeled pulmonary arteries. miR-145 deficiency and anti-miR-mediated reduction resulted in significant protection from the development of PAH. In contrast, miR-143 anti-miR had no effect. Furthermore, we observed upregulation of miR-145 in lung tissue of patients with idiopathic and heritable PAH compared with unaffected control subjects and demonstrated expression of miR-145 in SMC of remodeled vessels from such patients. Finally, we show elevated levels of miR-145 expression in primary PASMCs cultured from patients with BMPR2 mutations and also in the lungs of BMPR2-deficient mice. CONCLUSIONS: miR-145 is dysregulated in mouse models of PAH. Downregulation of miR-145 protects against the development of PAH. In patient samples of heritable PAH and idiopathic PAH, miR-145 is expressed in remodeled vessels and mutations in BMPR2 lead to upregulation of miR-145 in mice and PAH patients. Manipulation of miR-145 may represent a novel strategy in PAH treatment.


Asunto(s)
Modelos Animales de Enfermedad , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , MicroARNs/fisiología , Animales , Regulación hacia Abajo/genética , Femenino , Técnicas de Sustitución del Gen , Humanos , Hipertensión Pulmonar/prevención & control , Pulmón/patología , Pulmón/fisiología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/antagonistas & inhibidores , MicroARNs/genética
3.
Eur Heart J ; 34(22): 1636-43, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23530023

RESUMEN

AIMS: The long-term failure of autologous saphenous vein bypass grafts due to neointimal thickening is a major clinical burden. Identifying novel strategies to prevent neointimal thickening is important. Thus, this study aimed to identify microRNAs (miRNAs) that are dysregulated during neointimal formation and determine their pathophysiological relevance following miRNA manipulation. METHODS AND RESULTS: We undertook a microarray approach to identify dysregulated miRNAs following engraftment in an interpositional porcine graft model. These profiling experiments identified a number of miRNAs which were dysregulated following engraftment. miR-21 levels were substantially elevated following engraftment and these results were confirmed by quantitative real-time PCR in mouse, pig, and human models of vein graft neointimal formation. Genetic ablation of miR-21 in mice or grafted veins dramatically reduced neointimal formation in a mouse model of vein grafting. Furthermore, pharmacological knockdown of miR-21 in human veins resulted in target gene de-repression and a significant reduction in neointimal formation. CONCLUSION: This is the first report demonstrating that miR-21 plays a pathological role in vein graft failure. Furthermore, we also provided evidence that knockdown of miR-21 has therapeutic potential for the prevention of pathological vein graft remodelling.


Asunto(s)
MicroARNs/genética , Neointima/genética , Vena Safena/metabolismo , Injerto Vascular , Animales , Arteria Carótida Común/metabolismo , Células Cultivadas , Técnicas de Silenciamiento del Gen , Rechazo de Injerto/genética , Rechazo de Injerto/metabolismo , Humanos , Ratones , Ratones Noqueados , MicroARNs/metabolismo , Análisis por Micromatrices , Vena Safena/trasplante , Porcinos , Venas Cavas/metabolismo
4.
Blood ; 116(15): 2656-64, 2010 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-20610817

RESUMEN

A major limitation for adenoviral transduction in vivo is the profound liver tropism of adenovirus type 5 (Ad5). Recently, we demonstrated that coagulation factor X (FX) binds to Ad5-hexon protein at high affinity to mediate hepatocyte transduction after intravascular delivery. We developed novel genetically FX-binding ablated Ad5 vectors with lower liver transduction. Here, we demonstrate that FX-binding ablated Ad5 predominantly localize to the liver and spleen 1 hour after injection; however, they had highly reduced liver transduction in both control and macrophage-depleted mice compared with Ad5. At high doses in macrophage-depleted mice, FX-binding ablated vectors transduced the spleen more efficiently than Ad5. Immunohistochemical studies demonstrated transgene colocalization with CD11c(+), ER-TR7(+), and MAdCAM-1(+) cells in the splenic marginal zone. Systemic inflammatory profiles were broadly similar between FX-binding ablated Ad5 and Ad5 at low and intermediate doses, although higher levels of several inflammatory proteins were observed at the highest dose of FX-binding ablated Ad5. Subsequently, we generated a FX-binding ablated virus containing a high affinity Ad35 fiber that mediated a significant improvement in lung/liver ratio in macrophage-depleted CD46(+) mice compared with controls. Therefore, this study documents the biodistribution and reports the retargeting capacity of FX binding-ablated Ad5 vectors in vitro and in vivo.


Asunto(s)
Adenovirus Humanos/genética , Adenovirus Humanos/metabolismo , Proteínas de la Cápside/metabolismo , Factor X/metabolismo , Vectores Genéticos , Adenovirus Humanos/clasificación , Animales , Proteínas de la Cápside/genética , Quimiocinas/metabolismo , Citocinas/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Hígado/metabolismo , Hígado/virología , Pulmón/metabolismo , Pulmón/virología , Masculino , Ratones , Ratones Transgénicos , Unión Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serotipificación , Bazo/metabolismo , Bazo/virología , Factores de Tiempo , Distribución Tisular , Transducción Genética , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
5.
Dermatol Ther (Heidelb) ; 12(7): 1501-1533, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35596901

RESUMEN

Type 2 immunity evolved to combat helminth infections by orchestrating a combined protective response of innate and adaptive immune cells and promotion of parasitic worm destruction or expulsion, wound repair, and barrier function. Aberrant type 2 immune responses are associated with allergic conditions characterized by chronic tissue inflammation, including atopic dermatitis (AD) and asthma. Signature cytokines of type 2 immunity include interleukin (IL)-4, IL-5, IL-9, IL-13, and IL-31, mainly secreted from immune cells, as well as IL-25, IL-33, and thymic stromal lymphopoietin, mainly secreted from tissue cells, particularly epithelial cells. IL-4 and IL-13 are key players mediating the prototypical type 2 response; IL-4 initiates and promotes differentiation and proliferation of naïve T-helper (Th) cells toward a Th2 cell phenotype, whereas IL-13 has a pleiotropic effect on type 2 inflammation, including, together with IL-4, decreased barrier function. Both cytokines are implicated in B-cell isotype class switching to generate immunoglobulin E, tissue fibrosis, and pruritus. IL-5, a key regulator of eosinophils, is responsible for eosinophil growth, differentiation, survival, and mobilization. In AD, IL-4, IL-13, and IL-31 are associated with sensory nerve sensitization and itch, leading to scratching that further exacerbates inflammation and barrier dysfunction. Various strategies have emerged to suppress type 2 inflammation, including biologics targeting cytokines or their receptors, and Janus kinase inhibitors that block intracellular cytokine signaling pathways. Here we review type 2 inflammation, its role in inflammatory diseases, and current and future therapies targeting type 2 pathways, with a focus on AD. INFOGRAPHIC.

6.
Cells ; 8(2)2019 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-30678104

RESUMEN

Vascular smooth muscle cell (VSMC) dedifferentiation is a common feature of vascular disorders leading to pro-migratory and proliferative phenotypes, a process induced through growth factor and cytokine signaling cascades. Recently, many studies have demonstrated that small non-coding RNAs (miRNAs) can induce phenotypic effects on VSMCs in response to vessel injury. However, most studies have focused on the contribution of individual miRNAs. Our study aimed to conduct a detailed and unbiased analysis of both guide and passenger miRNA expression in vascular cells in vitro and disease models in vivo. We analyzed 100 miRNA stem loops by TaqMan Low Density Array (TLDA) from primary VSMCs in vitro. Intriguingly, we found that a larger proportion of the passenger strands was significantly dysregulated compared to the guide strands after exposure to pathological stimuli, such as platelet-derived growth factor (PDGF) and IL-1α. Similar findings were observed in response to injury in porcine vein grafts and stent models in vivo. In these studies, we reveal that the miRNA passenger strands are predominantly dysregulated in response to vascular injury.


Asunto(s)
Vasos Sanguíneos/lesiones , Regulación de la Expresión Génica , MicroARNs/metabolismo , Animales , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/patología , Citocinas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Masculino , MicroARNs/genética , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Porcinos
7.
Clin Exp Optom ; 90(5): 371-5, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17697184

RESUMEN

A 60-year-old male with a history of migraine presented with evidence of branch retinal arterial occlusion that developed at the time of an attack of retinal migraine. The diagnosis of branch arterial occlusion secondary to migraine was made after exclusion of numerous possible medical conditions. The possible role of vasospasm in this condition is discussed.


Asunto(s)
Trastornos Migrañosos/complicaciones , Oclusión de la Arteria Retiniana/etiología , Diagnóstico Diferencial , Humanos , Masculino , Persona de Mediana Edad , Oclusión de la Arteria Retiniana/diagnóstico , Oclusión de la Arteria Retiniana/fisiopatología , Pruebas del Campo Visual , Campos Visuales
8.
J Am Coll Cardiol ; 65(21): 2314-27, 2015 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-26022821

RESUMEN

BACKGROUND: Drug-eluting stents reduce the incidence of in-stent restenosis, but they result in delayed arterial healing and are associated with a chronic inflammatory response and hypersensitivity reactions. Identifying novel interventions to enhance wound healing and reduce the inflammatory response may improve long-term clinical outcomes. Micro-ribonucleic acids (miRNAs) are noncoding small ribonucleic acids that play a prominent role in the initiation and resolution of inflammation after vascular injury. OBJECTIVES: This study sought to identify miRNA regulation and function after implantation of bare-metal and drug-eluting stents. METHODS: Pig, mouse, and in vitro models were used to investigate the role of miRNA in in-stent restenosis. RESULTS: We documented a subset of inflammatory miRNAs activated after stenting in pigs, including the miR-21 stem loop miRNAs. Genetic ablation of the miR-21 stem loop attenuated neointimal formation in mice post-stenting. This occurred via enhanced levels of anti-inflammatory M2 macrophages coupled with an impaired sensitivity of smooth muscle cells to respond to vascular activation. CONCLUSIONS: MiR-21 plays a prominent role in promoting vascular inflammation and remodeling after stent injury. MiRNA-mediated modulation of the inflammatory response post-stenting may have therapeutic potential to accelerate wound healing and enhance the clinical efficacy of stenting.


Asunto(s)
Reestenosis Coronaria/metabolismo , Stents Liberadores de Fármacos , MicroARNs/metabolismo , Remodelación Vascular , Lesiones del Sistema Vascular/metabolismo , Animales , Reestenosis Coronaria/prevención & control , Inflamación/metabolismo , Masculino , Ratones Noqueados , Porcinos
9.
Clin Exp Optom ; 87(6): 394-9, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15575814

RESUMEN

A 43-year-old patient presenting with a highly congruous homonymous hemianopia was shown by neuro-imaging to have a very large arteriovenous malformation of the brain. The significance of finding this visual field defect, its unusual cause and the absence of symptoms other than longstanding migraine with aura are discussed.


Asunto(s)
Hemianopsia/etiología , Hemianopsia/fisiopatología , Malformaciones Arteriovenosas Intracraneales/complicaciones , Lóbulo Occipital/irrigación sanguínea , Campos Visuales , Adulto , Angiografía Cerebral , Femenino , Hemianopsia/diagnóstico , Humanos , Malformaciones Arteriovenosas Intracraneales/diagnóstico , Imagen por Resonancia Magnética , Migraña con Aura/etiología , Tomografía Computarizada por Rayos X , Pruebas del Campo Visual
10.
J Appl Psychol ; 88(3): 561-9, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12814304

RESUMEN

The authors compared a feedback workshop with both a no-feedback control group and a comparison group of managers who received a feedback report but no feedback workshop. The multisource feedback was based on ratings of a manager's influence behavior by subordinates, peers, and bosses. Managers in the feedback workshop increased their use of some core influence tactics with subordinates, whereas there was no change in behavior for the control group or for the comparison group. The feedback was perceived to be more useful by managers who received it in a workshop with a facilitator than by managers who received only a printed feedback report.


Asunto(s)
Retroalimentación , Relaciones Interpersonales , Cultura Organizacional , Facilitación Social , Femenino , Humanos , Masculino , Encuestas y Cuestionarios
11.
Hypertension ; 64(1): 185-94, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24732886

RESUMEN

Pulmonary endothelial cell apoptosis is a transient, yet defining pathogenic event integral to the onset of many pulmonary vascular diseases such as pulmonary hypertension (PH). However, there is a paucity of information concerning the molecular pathway(s) that control pulmonary arterial endothelial cell apoptosis. Here, we introduce a molecular axis that when functionally active seems to induce pulmonary arterial endothelial cell apoptosis in vitro and PH in vivo. In response to apoptotic stimuli, human pulmonary arterial endothelial cells exhibited robust induction of a programmed cell death 4 (PDCD4)/caspase-3/apoptotic pathway that was reversible by direct PDCD4 silencing. Indirectly, this pathway was also repressed by delivery of a microRNA-21 mimic. In vivo, genetic deletion of microRNA-21 in mice (miR-21(-/-) mice) resulted in functional activation of the PDCD4/caspase-3 axis in the pulmonary tissues, leading to the onset of progressive PH. Conversely, microRNA-21-overexpressing mice (CAG-microRNA-21 mice) exhibited reduced PDCD4 expression in pulmonary tissues and were partially resistant to PH in response to chronic hypoxia plus SU 5416 injury. Furthermore, direct PDCD4 knockout in mice (PDCD4(-/-) mice) potently blocked pulmonary caspase-3 activation and the development of chronic hypoxia plus SU 5416 PH, confirming its importance in disease onset. Broadly, these findings support the existence of a microRNA-21-responsive PDCD4/caspase-3 pathway in the pulmonary tissues that when active serves to promote endothelial apoptosis in vitro and PH in vivo.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/fisiología , Caspasa 3/metabolismo , Células Endoteliales/metabolismo , Hipertensión Pulmonar/metabolismo , MicroARNs/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Humanos , Ratones , MicroARNs/genética
12.
J Cardiothorac Surg ; 8: 183, 2013 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-23937994

RESUMEN

BACKGROUND: Cardiovascular disorders, including coronary artery bypass graft failure and in-stent restenosis remain significant opportunities for the advancement of novel therapeutics that target neointimal hyperplasia, a characteristic of both pathologies. Gene therapy may provide a successful approach to improve the clinical outcome of these conditions, but would benefit from the development of more efficient vectors for vascular gene delivery. The aim of this study was to assess whether a novel genetically engineered Adenovirus could be utilised to produce enhanced levels of vascular gene expression. METHODS: Vascular transduction capacity was assessed in primary human saphenous vein smooth muscle and endothelial cells using vectors expressing the LacZ reporter gene. The therapeutic capacity of the vectors was compared by measuring smooth muscle cell metabolic activity and migration following infection with vectors that over-express the candidate therapeutic gene tissue inhibitor of matrix metalloproteinase-3 (TIMP-3). RESULTS: Compared to Adenovirus serotype 5 (Ad5), the novel vector Ad5T*F35++ demonstrated improved binding and transduction of human vascular cells. Ad5T*F35++ mediated expression of TIMP-3 reduced smooth muscle cell metabolic activity and migration in vitro. We also demonstrated that in human serum samples pre-existing neutralising antibodies to Ad5T*F35++ were less prevalent than Ad5 neutralising antibodies. CONCLUSIONS: We have developed a novel vector with improved vascular transduction and improved resistance to human serum neutralisation. This may provide a novel vector platform for human vascular gene transfer.


Asunto(s)
Adenoviridae/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Cápside , Células Cultivadas , Humanos , Vena Safena , Inhibidor Tisular de Metaloproteinasa-3/análisis , Inhibidor Tisular de Metaloproteinasa-3/farmacología , Transducción Genética , beta-Galactosidasa/análisis
13.
Cardiovasc Res ; 93(4): 594-604, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22065733

RESUMEN

Vascular remodelling is an integral pathological process central to a number of cardiovascular diseases. The complex interplay between distinct cell populations in the vessel wall following vascular injury leads to inflammation, cellular dysfunction, pro-growth signals in the smooth muscle cell (SMC) compartment, and the acquisition of a synthetic phenotype. Although the signals for vascular remodelling are diverse in different pathological contexts, SMC proliferation and migration are consistently observed. It is therefore critical to elucidate key mechanisms central to these processes. MicroRNAs (miRNAs) are small non-coding sequences of RNA that have the capacity to regulate many genes, pathways, and complex biological networks within cells, acting either alone or in concert with one another. In diseases such as cancer and cardiac disease, the role of miRNA in disease pathogenesis has been documented in detail. In contrast, despite a great deal of interest in miRNA, relatively few studies have directly assessed the role of miRNA in vascular remodelling. The potential for modulation of miRNA to achieve therapeutic benefits in this setting is attractive. Here, we focus on the role of miRNA in vascular inflammation and remodelling associated with acute vascular injury (vein graft disease, angioplasty restenosis, and in-stent restenosis) as well as in vascular remodelling associated with the development of pulmonary arterial hypertension.


Asunto(s)
Hipertensión Pulmonar/fisiopatología , MicroARNs/fisiología , Músculo Liso Vascular/fisiopatología , Lesiones del Sistema Vascular/fisiopatología , Animales , Modelos Animales de Enfermedad , Endotelio Vascular/fisiopatología , Humanos , Transducción de Señal/fisiología , Vasculitis/fisiopatología
14.
Pharmacol Ther ; 136(1): 23-34, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22796519

RESUMEN

Coronary artery bypass grafting (CABG) and percutaneous coronary intervention (PCI), including stent insertion, are established therapies in both acute coronary syndromes (ACS) and symptomatic chronic coronary artery disease refractory to pharmacological therapy. These continually advancing treatments remain limited by failure of conduit grafts in CABG and by restenosis or thrombosis of stented vessel segments in PCI caused by neointimal hyperplasia, impaired endothelialisation and accelerated atherosclerosis. While pharmacological and technological advancements have improved patient outcomes following both procedures, when grafts or stents fail these result in significant health burdens. In this review we discuss the pathophysiology of vein graft disease and in-stent restenosis, gene therapy vector development and design, and translation from pre-clinical animal models through human clinical trials. We identify the key issues that are currently preventing vascular gene therapy from interfacing with clinical use and introduce the areas of research attempting to overcome these.


Asunto(s)
Angioplastia Coronaria con Balón/efectos adversos , Reestenosis Coronaria/prevención & control , Terapia Genética , Stents/efectos adversos , Venas/trasplante , Adenoviridae/genética , Animales , Ensayos Clínicos como Asunto , Dependovirus/genética , Endotelio Vascular/fisiología , Humanos , Lentivirus/genética , Neointima/patología , Retroviridae/genética
15.
Hum Gene Ther ; 23(12): 1247-57, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22931362

RESUMEN

We have previously shown that injury-induced neointima formation was rescued by adenoviral-Nogo-B gene delivery. Integrase-competent lentiviral vectors (ICLV) are efficient at gene delivery to vascular cells but present a risk of insertional mutagenesis. Conversely, integrase-deficient lentiviral vectors (IDLV) offer additional benefits through reduced mutagenesis risk, but this has not been evaluated in the context of vascular gene transfer. Here, we have investigated the performance and genetic safety of both counterparts in primary human vascular smooth muscle cells (VSMC) and compared gene transfer efficiency and assessed the genotoxic potential of ICLVs and IDLVs based on their integration frequency and insertional profile in the human genome. Expression of enhanced green fluorescent protein (eGFP) mediated by IDLVs (IDLV-eGFP) demonstrated efficient transgene expression in VSMCs. IDLV gene transfer of Nogo-B mediated efficient overexpression of Nogo-B in VSMCs, leading to phenotypic effects on VSMC migration and proliferation, similar to its ICLV version and unlike its eGFP control and uninfected VSMCs. Large-scale integration site analyses in VSMCs indicated that IDLV-mediated gene transfer gave rise to a very low frequency of genomic integration compared to ICLVs, revealing a close-to-random genomic distribution in VSMCs. This study demonstrates for the first time the potential of IDLVs for safe and efficient vascular gene transfer.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Integrasas/genética , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Movimiento Celular , Proliferación Celular , Células Cultivadas , Proteínas Fluorescentes Verdes/genética , Humanos , Lentivirus/genética , Mutagénesis Insercional , Proteínas de la Mielina/genética , Proteínas Nogo
17.
Br J Pharmacol ; 159(3): 543-53, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20015089

RESUMEN

BACKGROUND AND PURPOSE: Sphingosine-1-phosphate and its receptors may be involved in vascular smooth muscle cell (VSMC) proliferation following vascular injury. Here, we evaluate the effect of d-erythro-N,N-dimethylsphingosine (DMS), a sphingosine kinase (SK) inhibitor, on VSMC proliferation, apoptosis and neointimal formation. EXPERIMENTAL APPROACH: Growth responses in vitro to fetal calf serum (FCS) were measured by [(3)H]-thymidine incorporation and extracellular signal-regulated kinase-1/2 (ERK-1/2) activation in quiescent primary cultures of porcine VSMC in the presence and absence of various concentrations of the SK inhibitor DMS. In vivo treatment with DMS was delivered with a local endoluminal catheter, following balloon injury of coronary arteries. The artery intimal formation was investigated by angiography, myography and histomorphometry. KEY RESULTS: In vitro experiments indicated that DMS induced a dose-dependent reduction in [(3)H]-thymidine incorporation and ERK-1/2 activation via a protein kinase C (PKC) independent mechanism with an IC(50) value of 12 +/- 6 and 15 +/- 10 microM respectively. DMS also reduced Akt signalling. Four weeks following in vivo delivery of DMS, complete functional endothelial regeneration was observed in all treatment groups, with significant reduction of intimal formation (vehicle 23.7 +/- 4.6% vs. DMS infusion 8.92 +/- 2.9%, P < 0.05). CONCLUSIONS AND IMPLICATIONS: Taken together, these results demonstrate that local administration of the SK inhibitor, DMS, reduced neointimal formation, and this effect could involve inhibition of ERK-1/2 and Akt signalling, and modulation of smooth muscle growth.


Asunto(s)
Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Esfingosina/farmacología , Túnica Íntima/efectos de los fármacos , Túnica Íntima/patología , Animales , Apoptosis/efectos de los fármacos , Cateterismo , Proliferación Celular/efectos de los fármacos , Hiperplasia/patología , Lisofosfolípidos , Masculino , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/farmacología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol) , Proteína Quinasa C/metabolismo , Proteína Quinasa C/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Sus scrofa , Túnica Íntima/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA