Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
J Vasc Surg ; 58(3): 766-75.e12, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23755976

RESUMEN

OBJECTIVE: The present study is designed to understand the contribution of peripheral vascular disease and peripheral neuropathy to the wound-healing impairment associated with diabetes. Using a rabbit model of diabetic neuroischemic wound healing, we investigated rate of healing, leukocyte infiltration, and expression of cytokines, interleukin-8 and interleukin-6, and neuropeptides, substance P, and neuropeptide Y. METHODS: Diabetes was induced in New Zealand White rabbits by administering alloxan while control rabbits received saline. Ten days later, animals in both groups underwent surgery. One ear served as a sham, and the other was made ischemic (ligation of central+rostral arteries) or neuroischemic (ischemia+ resection of central+rostral nerves). Four 6-mm punch biopsy wounds were created in both ears and wound healing was followed for 10 days using computerized planimetry. RESULTS: Nondiabetic sham and ischemic wounds healed significantly more rapidly than diabetic sham and ischemic wounds. Healing was slowest in neuroischemic wounds, irrespective of diabetic status. A high M1/M2 macrophage ratio and a high proinflammatory cytokine expression, both indicators of chronic proinflammatory state, and low neuropeptide expression were seen in preinjury diabetic skin. Postinjury, in diabetic wounds, the M1/M2 ratio remained high, the reactive increase in cytokine expression was low, and neuropeptide expression was further decreased in neuroischemic wounds. CONCLUSIONS: This rabbit model illustrates how a combination of a high M1/M2 ratio, a failure to mount postinjury cytokine response as well as a diminished neuropeptide expression, contribute to wound-healing impairment in diabetes. The addition of neuropathy to ischemia leads to equivalently severe impaired wound-healing irrespective of diabetes status, suggesting that in the presence of ischemia, loss of neuropeptide function contributes to the impaired healing associated with diabetes.


Asunto(s)
Citocinas/metabolismo , Diabetes Mellitus Experimental/complicaciones , Angiopatías Diabéticas/etiología , Neuropatías Diabéticas/etiología , Mediadores de Inflamación/metabolismo , Isquemia/etiología , Neuropéptidos/metabolismo , Úlcera Cutánea/etiología , Piel , Cicatrización de Heridas , Animales , Citocinas/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/metabolismo , Angiopatías Diabéticas/genética , Angiopatías Diabéticas/inmunología , Angiopatías Diabéticas/metabolismo , Angiopatías Diabéticas/patología , Neuropatías Diabéticas/genética , Neuropatías Diabéticas/inmunología , Neuropatías Diabéticas/metabolismo , Neuropatías Diabéticas/patología , Regulación hacia Abajo , Isquemia/genética , Isquemia/inmunología , Isquemia/metabolismo , Isquemia/patología , Macrófagos/inmunología , Neuropéptidos/genética , Conejos , Piel/inmunología , Piel/metabolismo , Piel/patología , Úlcera Cutánea/genética , Úlcera Cutánea/inmunología , Úlcera Cutánea/metabolismo , Úlcera Cutánea/patología , Factores de Tiempo , Regulación hacia Arriba
2.
Biochem Biophys Res Commun ; 425(2): 261-5, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22842581

RESUMEN

BACKGROUND: RNAi technology is a promising tool for gene therapy of vascular disease. However, the biological heterogeneity between endothelial (EC) and vascular smooth muscle cells (SMC) and within different vascular beds make them differentially susceptible to siRNA. This is further complicated by the task of choosing the right transfection reagent that leads to consistent gene silencing across all cell types with minimal toxicity. The goal of this study was to investigate the intrinsic RNAi susceptibility of primary human aortic and coronary artery endothelial and vascular smooth muscle cells (AoEC, CoEC, AoSMC and CoSMC) using adherent cell cytometry. METHODS: Cells were seeded at a density of 5000cells/well of a 96well plate. Twenty four hours later cells were transfected with either non-targeting unlabeled control siRNA (50nM), or non-targeting red fluorescence labeled siRNA (siGLO Red, 5 or 50nM) using no transfection reagent, HiPerFect or Lipofectamine RNAiMAX. Hoechst nuclei stain was used to label cells for counting. For data analysis an adherent cell cytometer, Celigo was used. RESULTS: Red fluorescence counts were normalized to the cell count. EC displayed a higher susceptibility towards siRNA delivery than SMC from the corresponding artery. CoSMC were more susceptible than AoSMC. In all cell types RNAiMAX was more potent compared to HiPerFect or no transfection reagent. However, after 24h, RNAiMAX led to a significant cell loss in both AoEC and CoEC. None of the other transfection conditions led to a significant cell loss. CONCLUSION: This study confirms our prior observation that EC are more susceptible to siRNA than SMC based on intracellular siRNA delivery. RNAiMax treatment led to significant cell loss in AoEC and CoEC, but not in the SMC populations. Additionally, this study is the first to demonstrate that coronary SMC are more susceptible to siRNA than aortic SMC.


Asunto(s)
Aorta/citología , Vasos Coronarios/citología , Endotelio Vascular/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transfección , Adhesión Celular , Recuento de Células , Citometría de Flujo , Fluorescencia , Humanos , Músculo Liso Vascular/citología , ARN Interferente Pequeño/genética
3.
J Transl Med ; 9: 48, 2011 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-21518450

RESUMEN

BACKGROUND: siRNA technology is a promising tool for gene therapy of vascular disease. Due to the multitude of reagents and cell types, RNAi experiment optimization can be time-consuming. In this study adherent cell cytometry was used to rapidly optimize siRNA transfection in human aortic vascular smooth muscle cells (AoSMC). METHODS: AoSMC were seeded at a density of 3000-8000 cells/well of a 96 well plate. 24 hours later AoSMC were transfected with either non-targeting unlabeled siRNA (50 nM), or non-targeting labeled siRNA, siGLO Red (5 or 50 nM) using no transfection reagent, HiPerfect or Lipofectamine RNAiMax. For counting cells, Hoechst nuclei stain or Cell Tracker green were used. For data analysis an adherent cell cytometer, Celigo® was used. Data was normalized to the transfection reagent alone group and expressed as red pixel count/cell. RESULTS: After 24 hours, none of the transfection conditions led to cell loss. Red fluorescence counts were normalized to the AoSMC count. RNAiMax was more potent compared to HiPerfect or no transfection reagent at 5 nM siGLO Red (4.12 +/-1.04 vs. 0.70 +/-0.26 vs. 0.15 +/-0.13 red pixel/cell) and 50 nM siGLO Red (6.49 +/-1.81 vs. 2.52 +/-0.67 vs. 0.34 +/-0.19). Fluorescence expression results supported gene knockdown achieved by using MARCKS targeting siRNA in AoSMCs. CONCLUSION: This study underscores that RNAi delivery depends heavily on the choice of delivery method. Adherent cell cytometry can be used as a high throughput-screening tool for the optimization of RNAi assays. This technology can accelerate in vitro cell assays and thus save costs.


Asunto(s)
Citometría de Flujo/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Interferencia de ARN , Aorta/citología , Adhesión Celular , Recuento de Células , Muerte Celular , Células Cultivadas , Colorantes Fluorescentes/metabolismo , Humanos , Indicadores y Reactivos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada , ARN Interferente Pequeño/metabolismo , Transfección
4.
Biomaterials ; 57: 22-32, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25956194

RESUMEN

A principal challenge in wound healing is a lack of cell recruitment, cell infiltration, and vascularization, which occurs in the absence of temporal and spatial cues. We hypothesized that a scaffold that expands due to local changes in pH may alter oxygen and nutrient transport and the local cell density, leading to enhanced cell deposition and survival. In this study, we present a pH-responsive scaffold that increases oxygen transport, as confirmed by our finite element model analysis, and cell proliferation relative to a non-responsive scaffold. In vivo, responsive scaffolds induce a pro-healing gene expression profile indicative of enhanced angiogenesis, granulation tissue formation, and tissue remodeling. Scaffolds that stretch in response to their environment may be a hallmark for tissue regeneration.


Asunto(s)
Metacrilatos/química , Oxígeno/metabolismo , Andamios del Tejido/química , Cicatrización de Heridas , Animales , Proliferación Celular , Supervivencia Celular , Análisis de Elementos Finitos , Concentración de Iones de Hidrógeno , Ratones , Células 3T3 NIH , Porosidad , Ratas Wistar , Ingeniería de Tejidos
5.
Biomaterials ; 35(9): 3071-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24397987

RESUMEN

An excessive tissue response to prosthetic arterial graft material leads to intimal hyperplasia (IH), the leading cause of late graft failure. Seroma and abnormal capsule formation may also occur after prosthetic material implantation. The matricellular protein Thrombospondin-2 (TSP-2) has shown to be upregulated in response to biomaterial implantation. This study evaluates the uptake and release of small interfering RNA (siRNA) from unmodified and surface functionalized electrospun PET graft materials. ePET graft materials were synthesized using electrospinning technology. Subsets of the ePET materials were then chemically modified to create surface functional groups. Unmodified and surface-modified ePET grafts were dip-coated in siRNAs alone or siRNAs complexed with transfection reagents polyethyleneimine (PEI) or Lipofectamine RNAiMax. Further, control and TSP-2 siRNA-PEI complex treated ePET samples were placed onto a confluent layer of human aortic smooth muscle cells (AoSMCs). Complexation of all siRNAs with PEI led to a significant increase in adsorption to unmodified ePET. TSP-2 siRNA-PEI released from unmodified-ePET silenced TSP-2 in AoSMC. Regardless of the siRNA-PEI complex evaluated, AoSMC migrated into the ePET. siRNA-PEI complexes delivered to AoSMC from dip-coated ePET can result in gene knockdown. This methodology for siRNA delivery may improve the tissue response to vascular and other prosthetics.


Asunto(s)
Aorta/citología , Materiales Biocompatibles Revestidos/farmacología , Silenciador del Gen/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Polietilenglicoles/farmacología , Polietileneimina/farmacología , ARN Interferente Pequeño/metabolismo , Adsorción , Adhesión Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/ultraestructura , Tereftalatos Polietilenos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA