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











Base de datos
Intervalo de año de publicación
1.
Am J Infect Control ; 52(8): 915-918, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38657905

RESUMEN

BACKGROUND: The standard of care for disinfecting needleless connectors (NCs) of central venous catheters includes alcohol-containing caps or up to a 15-second scrub with alcohol or chlorhexidine. Due to the clinical impact and high cost of treating Central line-associated bloodstream infections (CLABSI), reducing the incidence of CLABSI is a priority for public health and of the Centers for Disease Control. Alcohol-containing caps have been demonstrated to disinfect external NC surfaces, but not the internal surface. Ultraviolet light (UV-C) is a strategy for disinfection of NC internal and external surfaces. METHODS: Four clinically relevant bacteria (Staphylococcus aureus, Staphylococcus epidermidis, Escherichia coli, Pseudomonas aeruginosa) and Candida albicans were inoculated on NCs. Disinfection efficacy was measured after exposure to one second of 285 nm UV-C light at 48 mW/cm2 in a proprietary handheld device and UV-C transparent NC or standard of care. Disinfection of internal and external surfaces of NC inoculated with S aureus using alcohol caps, and UV-C was also compared. RESULTS: A 4-log reduction in colony forming units (CFUs) on the interior and exterior surfaces of the UV-transparent NC of clinically relevant pathogens was observed with UV-C light at this power for 1 second. DISCUSSION: We demonstrated the efficacy of UV-C for the disinfection of NCs in one second using the UV-C device in benchtop studies. CONCLUSIONS: This device holds promise for reducing CLABSI, and clinical studies are planned.


Asunto(s)
Desinfección , Rayos Ultravioleta , Desinfección/métodos , Desinfección/instrumentación , Humanos , Bacterias/efectos de la radiación , Catéteres Venosos Centrales/microbiología , Recuento de Colonia Microbiana , Infecciones Relacionadas con Catéteres/prevención & control , Candida albicans/efectos de los fármacos , Candida albicans/efectos de la radiación
2.
Perit Dial Int ; 37(1): 109-111, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28153966

RESUMEN

We evaluated the microbiological performance of an ultraviolet (UV) light-based peritoneal dialysis catheter connection system. The system includes a UV light-generating device combined with a UV transmissive window incorporated into the transfer set. Each UV transparent transfer set was inoculated with 10 µL of cultured inoculum consisting of either S. aureus, E. coli, or C. albicans After being inoculated, we attached a solution set connector to the transfer catheter, and exposed that connection to a UV light dose of approximately 340 mJoules/cm2 After exposure to UV light, we broke the seal of the solution set and opened the plunger valve on the UV transmissive transfer catheter. We then flushed 10 mL of dialysate through the connection. The flushed solution was collected, diluted, plated on agar medium, and incubated for 24 hours. Results were compared to positive controls collected in an identical manner without exposure to UV light. Thirty test samples and 3 positive controls were collected for each organism. All test samples exposed to UV light had complete kill of bacteria except 1 colony on a single plate in the S. aureus group. Mean log reduction was 4.03 for C. albicans, 4.73 for S. aureus, and 5.29 for E. coli All positive control samples had significant bacterial growth. Our results demonstrate that the application of UV light within a UV transmissive transfer catheter window produces a germicidal effect upon microorganisms known to be associated with peritonitis.


Asunto(s)
Catéteres de Permanencia/microbiología , Desinfección/métodos , Contaminación de Equipos/prevención & control , Diálisis Peritoneal/instrumentación , Rayos Ultravioleta , Bacterias/crecimiento & desarrollo , Candida albicans/aislamiento & purificación , Escherichia coli/aislamiento & purificación , Humanos , Diálisis Peritoneal/métodos , Sensibilidad y Especificidad , Staphylococcus aureus/aislamiento & purificación
3.
Hum Gene Ther Methods ; 24(5): 279-88, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23937265

RESUMEN

Hypoxia-inducible factor-1 alpha (HIF-1α) gene therapy holds great promise for the treatment of myocardial ischemia. Both preclinical and clinical evaluations of this therapy are underway and can benefit from a vector strategy that allows noninvasive assessment of HIF-1α expression as an objective measure of gene delivery. We have developed a novel bidirectional plasmid vector (pcTnT-HIF-1α-VP2-TSTA-fluc), which employs the cardiac troponin T (cTnT) promoter in conjunction with a two-step transcriptional amplification (TSTA) system to drive the linked expression of a recombinant HIF-1α gene (HIF-1α-VP2) and the firefly luciferase gene (fluc). The firefly luciferase (FLuc) activity serves as a surrogate for HIF-1α-VP2 expression, and can be noninvasively assessed in mice using bioluminescence imaging after vector delivery. Transfection of cultured HL-1 cardiomyocytes with pcTnT-HIF-1α-VP2-TSTA-fluc led to a strong correlation between FLuc and HIF-1α-dependent vascular endothelial growth factor expression (r(2)=0.88). Intramyocardial delivery of pcTnT-HIF-1α-VP2-TSTA-fluc into infarcted mouse myocardium led to persistent HIF-1α-VP2 expression for 4 weeks, even though it improved neither CD31+ microvessel density nor echocardiographically determined left ventricular systolic function. These results lend support to recent findings of suboptimal efficacy associated with plasmid-mediated HIF-1α therapy. The imaging techniques developed herein should be useful for further optimizing HIF-1α-VP2 therapy in preclinical models of myocardial ischemia.


Asunto(s)
Terapia Genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Luciferasas de Luciérnaga/metabolismo , Isquemia Miocárdica/terapia , Animales , Línea Celular , Ecocardiografía , Femenino , Vectores Genéticos/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Luciferasas de Luciérnaga/genética , Ratones , Ratones Endogámicos BALB C , Contracción Miocárdica , Miocardio/citología , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica , Imagen Óptica/métodos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Regiones Promotoras Genéticas , Troponina T/genética , Factores de Crecimiento Endotelial Vascular/genética , Factores de Crecimiento Endotelial Vascular/metabolismo
4.
Diabetes ; 60(5): 1383-92, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21441442

RESUMEN

OBJECTIVE: ß-Cells that express an imaging reporter have provided powerful tools for studying ß-cell development, islet transplantation, and ß-cell autoimmunity. To further expedite diabetes research, we generated transgenic C57BL/6 "MIP-TF" mice that have a mouse insulin promoter (MIP) driving the expression of a trifusion (TF) protein of three imaging reporters (luciferase/enhanced green fluorescent protein/HSV1-sr39 thymidine kinase) in their ß-cells. This should enable the noninvasive imaging of ß-cells by charge-coupled device (CCD) and micro-positron emission tomography (PET), as well as the identification of ß-cells at the cellular level by fluorescent microscopy. RESEARCH DESIGN AND METHODS: MIP-TF mouse ß-cells were multimodality imaged in models of type 1 and type 2 diabetes. RESULTS: MIP-TF mouse ß-cells were readily identified in pancreatic tissue sections using fluorescent microscopy. We show that MIP-TF ß-cells can be noninvasively imaged using microPET. There was a correlation between CCD and microPET signals from the pancreas region of individual mice. After low-dose streptozotocin administration to induce type 1 diabetes, we observed a progressive reduction in bioluminescence from the pancreas region before the appearance of hyperglycemia. Although there have been reports of hyperglycemia inducing proinsulin expression in extrapancreatic tissues, we did not observe bioluminescent signals from extrapancreatic tissues of diabetic MIP-TF mice. Because MIP-TF mouse ß-cells express a viral thymidine kinase, ganciclovir treatment induced hyperglycemia, providing a new experimental model of type 1 diabetes. Mice fed a high-fat diet to model early type 2 diabetes displayed a progressive increase in their pancreatic bioluminescent signals, which were positively correlated with area under the curve-intraperitoneal glucose tolerance test (AUC-IPGTT). CONCLUSIONS: MIP-TF mice provide a new tool for monitoring ß-cells from the single cell level to noninvasive assessments of ß-cells in models of type 1 diabetes and type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Animales , Citometría de Flujo , Prueba de Tolerancia a la Glucosa , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Insulina/genética , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Tomografía de Emisión de Positrones , Regiones Promotoras Genéticas/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Timidina Quinasa/genética , Timidina Quinasa/metabolismo
5.
Mol Imaging Biol ; 13(6): 1124-32, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21267661

RESUMEN

PURPOSE: We tested a novel imaging strategy, in which both the survival of transplanted myoblasts and their therapeutic transgene expression, a recombinant hypoxia-inducible factor-1α (HIF-1α-VP2), can be monitored using firefly luciferase (fluc) and Renilla luciferase (hrl) bioluminescence reporter genes, respectively. PROCEDURES: The plasmid pUbi-hrl-pUbi-HIF-1α-VP2, which expresses both hrl and HIF-1α-VP2 using two ubiquitin promoters, was characterized in vitro. C2c12 myoblasts stably expressing fluc and transiently transfected with pUbi-hrl-pUbi-HIF-1α-VP2 were injected into the mouse hindlimb. Both hrl and fluc expression were monitored using bioluminescence imaging (BLI). RESULTS: Strong correlations existed between the expression of hRL and each of HIF-1α-VP2, VEGF, and PlGF (r(2) > 0.83, r(2) > 0.82, and r(2) > 0.97, respectively). In vivo, both transplanted cells and HIF-1α-VP2 transgene expression were successfully imaged using BLI. CONCLUSIONS: An objective evaluation of myoblast-mediated gene transfer in living mice can be performed by monitoring both the survival and the transgene expression of transplanted myoblasts using the techniques developed herein.


Asunto(s)
Técnicas de Transferencia de Gen , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Imagenología Tridimensional/métodos , Mediciones Luminiscentes/métodos , Mioblastos/metabolismo , Animales , Recuento de Células , Línea Celular , Trasplante de Células , Pruebas de Enzimas , Ensayo de Inmunoadsorción Enzimática , Estudios de Factibilidad , Genes Reporteros , Vectores Genéticos/genética , Humanos , Luz , Luciferasas de Renilla/metabolismo , Ratones , Neovascularización Fisiológica , Plásmidos/genética , Transfección , Transgenes/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA