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.
J Control Release ; 337: 458-471, 2021 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-34324895

RESUMEN

Focused ultrasound (FUS) in combination with systemically injected microbubbles can be used to non-invasively open the blood-brain barrier (BBB) in targeted regions for a variety of therapeutic applications. Over the past two decades, preclinical research into the safety and efficacy of FUS-induced BBB opening has proven this technique to be transient and efficacious, propelling FUS-induced BBB opening into several clinical trials in recent years. However, as clinical trials further progress, the neuroinflammatory response to FUS-induced BBB opening needs to be better understood. In this study, we provide further insight into the relationship of microbubble cavitation and the resulting innate immune response to FUS-induced BBB opening. By keeping ultrasound parameters fixed (i.e. frequency, pressure, pulse length, etc.), three groups of mice were sonicated using a real-time cavitation controller until a target cavitation dose was reached (1 x 107 V2•s, 5 x 107 V2•s, 1 x 108 V2•s). The change in relative gene expression of the mouse inflammatory cytokines and receptors were evaluated at three different time-points (6 h, 24 h, and 72 h) after FUS. At both 6 and 24 h time-points, significant changes in relative gene expression of inflammatory cytokines and receptors were observed across all cavitation groups. However, the degree of changes in relative expression levels and the number of genes with significant changes in expression varied across the cavitation groups. Groups with a higher cavitation dose exhibited both greater changes in relative expression levels and greater number of significant changes. By 72 h post-opening, the gene expression levels returned to baseline in all cavitation dose groups, signifying a transient inflammatory response to FUS-induced BBB opening at the targeted cavitation dose levels. Furthermore, the real-time cavitation controller was able to produce consistent and significantly different BBB permeability enhancement volumes across the three different cavitation dose groups. These results indicate that cavitation monitoring and controlling during FUS-induced BBB opening can be used to potentially modulate or limit the degree of neuroinflammation, further emphasizing the importance of implementing cavitation controllers as FUS-induced BBB opening is translated into the clinic.


Asunto(s)
Barrera Hematoencefálica , Inflamación , Sonicación/métodos , Animales , Sistemas de Liberación de Medicamentos , Imagen por Resonancia Magnética , Ratones , Microburbujas , Permeabilidad , Sonicación/efectos adversos
2.
Sci Rep ; 9(1): 19402, 2019 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-31852909

RESUMEN

Focused ultrasound-enhanced intranasal (IN + FUS) delivery is a noninvasive approach that utilizes the olfactory pathway to administer pharmacological agents directly to the brain, allowing for a more homogenous distribution in targeted locations compared to IN delivery alone. However, whether such a strategy has therapeutic values, especially in neurodegenerative disorders such as Parkinson's disease (PD), remains to be established. Herein, we evaluated whether the expression of tyrosine hydroxylase (TH), the rate limiting enzyme in dopamine catalysis, could be enhanced by IN + FUS delivery of brain-derived neurotrophic factor (BDNF) in a toxin-based PD mouse model. Mice were put on the subacute dosing regimen of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), producing bilateral degeneration of the nigrostriatal pathway consistent with early-stage PD. MPTP mice then received BDNF intranasally followed by multiple unilateral FUS-induced blood-brain barrier (BBB) openings in the left basal ganglia for three consecutive weeks. Subsequently, mice were survived for two months and were evaluated morphologically and behaviorally to determine the integrity of their nigrostriatal dopaminergic pathways. Mice receiving IN + FUS had significantly increased TH immunoreactivity in the treated hemisphere compared to the untreated hemisphere while mice receiving only FUS-induced BBB opening or no treatment at all did not show any differences. Additionally, behavioral changes were only observed in the IN + FUS treated mice, indicating improved motor control function in the treated hemisphere. These findings demonstrate the robustness of the method and potential of IN + FUS for the delivery of bioactive factors for treatment of neurodegenerative disorder.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Dopamina/genética , Enfermedad de Parkinson Secundaria/terapia , Enfermedad de Parkinson/terapia , Tirosina 3-Monooxigenasa/genética , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/efectos adversos , Administración Intranasal , Animales , Ganglios Basales/efectos de los fármacos , Ganglios Basales/patología , Ganglios Basales/efectos de la radiación , Barrera Hematoencefálica/diagnóstico por imagen , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/efectos de la radiación , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/efectos de la radiación , Cuerpo Estriado/diagnóstico por imagen , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/efectos de la radiación , Modelos Animales de Enfermedad , Dopamina/biosíntesis , Humanos , Ratones , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/genética , Enfermedad de Parkinson Secundaria/patología , Sustancia Negra/diagnóstico por imagen , Sustancia Negra/efectos de los fármacos , Sustancia Negra/efectos de la radiación , Ondas Ultrasónicas
3.
Front Neurosci ; 11: 607, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29187808

RESUMEN

Non-invasive brain stimulation using focused ultrasound has many potential applications as a research and clinical tool, including its incorporation as either an extracorporeal or implantable neural prosthetic. To this end, we investigated the effect of focused ultrasound (FUS) combined with systemically administered microbubbles on visual-motor decision-making behavior in monkeys. We applied FUS to the putamen in one hemisphere to open the blood-brain barrier (BBB), and then tested behavioral performance 3-4 h later. On days when the monkeys were treated with FUS, their decisions were faster and more accurate than days without sonication. The performance improvement suggested both a shift in the decision criterion and an enhancement of the use of sensory evidence in the decision process. FUS also interacted with the effect of a low dose of haloperidol. The findings indicate that a two-minute application of FUS can have a sustained impact on performance of complex cognitive tasks, and may increase the efficacy of psychoactive medications. The results lend further support to the idea that the dorsal striatum plays an integral role in evidence- and reward-based decision-making, and provide motivation for incorporating FUS into cognitive neural prosthetic devices.

4.
J Cereb Blood Flow Metab ; 37(2): 706-714, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26969468

RESUMEN

The delivery of most therapeutic agents is rendered ineffective for the treatment of brain diseases due to the presence of the blood-brain barrier (BBB). The goal of this study was to investigate the effect of pre-infusion focused ultrasound (FUS) and microbubbles on the distribution of direct brain infusion in vivo. A single-element FUS transducer was used in all sonications, which were carried out immediately prior to direct infusion procedures. Mice received direct infusion of either Gadolinium-labeled albumin (Gd-albumin, 74 kDa) or adeno-associated virus (AAV, ∼4 MDa). The volumes of Gd-albumin at 30 min were deemed comparable ( P = 0.334) between the direct infusion (DI)-only group and the FUS + DI group. At 120 min, the FUS + DI group showed significantly higher contrast-enhanced volume (9.76 ± 0.74 mm3) than the DI-only group (7.14 ± 0.34 mm3). For mice infused with AAV, the total volume of transduction was estimated as GFP-positive regions and FUS + DI group demonstrated significantly higher ( P = 0.017) transduction efficiency in vivo. In conclusion, enhanced bio-distribution of directly infused agents was observed when the targeted region was pre-conditioned with FUS and microbubbles. Focused ultrasound has the potential, as an adjuvant technique, to significantly enhance direct brain infusion and achieve the desired therapeutic outcomes.


Asunto(s)
Encéfalo/metabolismo , Medios de Contraste/administración & dosificación , Dependovirus/genética , Sistemas de Liberación de Medicamentos/instrumentación , Gadolinio/administración & dosificación , Técnicas de Transferencia de Gen , Microburbujas , Albúmina Sérica/administración & dosificación , Animales , Barrera Hematoencefálica/metabolismo , Medios de Contraste/farmacocinética , Diseño de Equipo , Gadolinio/farmacocinética , Terapia Genética , Bombas de Infusión , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Albúmina Sérica/farmacocinética , Sonicación/instrumentación , Transducción Genética
5.
J Cereb Blood Flow Metab ; 37(4): 1236-1250, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27278929

RESUMEN

Focused ultrasound in conjunction with lipid microbubbles has fully demonstrated its ability to induce non-invasive, transient, and reversible blood-brain barrier opening. This study was aimed at testing the feasibility of our lipid-coated microbubbles as a vector for targeted drug delivery in the treatment of central nervous system diseases. These microbubbles were labeled with the fluorophore 5-dodecanoylaminfluorescein. Focused ultrasound targeted mouse brains in vivo in the presence of these microbubbles for trans-blood-brain barrier delivery of 5-dodecanoylaminfluorescein. This new approach, compared to previously studies of our group, where fluorescently labeled dextrans and microbubbles were co-administered, represents an appreciable improvement in safety outcome and targeted drug delivery. This novel technique allows the delivery of 5-dodecanoylaminfluorescein at the region of interest unlike the alternative of systemic exposure. 5-dodecanoylaminfluorescein delivery was assessed by ex vivo fluorescence imaging and by in vivo transcranial passive cavitation detection. Stable and inertial cavitation doses were quantified. The cavitation dose thresholds for estimating, a priori, successful targeted drug delivery were, for the first time, identified with inertial cavitation were concluded to be necessary for successful delivery. The findings presented herein indicate the feasibility and safety of the proposed microbubble-based targeted drug delivery and that, if successful, can be predicted by cavitation detection in vivo.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Lípidos/química , Microburbujas , Vehículos Farmacéuticos/química , Sonicación/métodos , Animales , Medios de Contraste/administración & dosificación , Medios de Contraste/química , Estudios de Factibilidad , Fluoresceínas/administración & dosificación , Fluoresceínas/química , Imagen por Resonancia Magnética , Masculino , Ratones Endogámicos C57BL , Microscopía Confocal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA