Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38891875

ABSTRACT

Transcranial focused ultrasound stimulation (tFUS) has emerged as a promising neuromodulation technique that delivers acoustic energy with high spatial resolution for inducing long-term potentiation (LTP)- or depression (LTD)-like plasticity. The variability in the primary effects of tFUS-induced plasticity could be due to different stimulation patterns, such as intermittent versus continuous, and is an aspect that requires further detailed exploration. In this study, we developed a platform to evaluate the neuromodulatory effects of intermittent and continuous tFUS on motor cortical plasticity before and after tFUS application. Three groups of rats were exposed to either intermittent, continuous, or sham tFUS. We analyzed the neuromodulatory effects on motor cortical excitability by examining changes in motor-evoked potentials (MEPs) elicited by transcranial magnetic stimulation (TMS). We also investigated the effects of different stimulation patterns on excitatory and inhibitory neural biomarkers, examining c-Fos and glutamic acid decarboxylase (GAD-65) expression using immunohistochemistry staining. Additionally, we evaluated the safety of tFUS by analyzing glial fibrillary acidic protein (GFAP) expression. The current results indicated that intermittent tFUS produced a facilitation effect on motor excitability, while continuous tFUS significantly inhibited motor excitability. Furthermore, neither tFUS approach caused injury to the stimulation sites in rats. Immunohistochemistry staining revealed increased c-Fos and decreased GAD-65 expression following intermittent tFUS. Conversely, continuous tFUS downregulated c-Fos and upregulated GAD-65 expression. In conclusion, our findings demonstrate that both intermittent and continuous tFUS effectively modulate cortical excitability. The neuromodulatory effects may result from the activation or deactivation of cortical neurons following tFUS intervention. These effects are considered safe and well-tolerated, highlighting the potential for using different patterns of tFUS in future clinical neuromodulatory applications.


Subject(s)
Evoked Potentials, Motor , Motor Cortex , Neuronal Plasticity , Transcranial Magnetic Stimulation , Animals , Motor Cortex/physiology , Rats , Male , Evoked Potentials, Motor/physiology , Transcranial Magnetic Stimulation/methods , Proto-Oncogene Proteins c-fos/metabolism , Ultrasonic Waves , Rats, Sprague-Dawley , Glial Fibrillary Acidic Protein/metabolism , Glutamate Decarboxylase/metabolism
2.
ACS Chem Neurosci ; 15(9): 1728-1731, 2024 05 01.
Article in English | MEDLINE | ID: mdl-38634833

ABSTRACT

Ultrasound neuromodulation is a potential alternative therapy for suppressing epileptic discharges. Recently, several human clinical trials have reported promising results from repeated focused ultrasound (FUS) treatments for temporal lobe epilepsy. In this Viewpoint, we highlight the valuable guidance of preclinical validation methods for choosing the optimal FUS parameters, thus ensuring consistency with the outcomes of clinical trials and leading human trials to the safest and most effective approaches.


Subject(s)
Disease Models, Animal , Epilepsy , Animals , Humans , Epilepsy/therapy , Epilepsy, Temporal Lobe/therapy , Ultrasonic Therapy/methods
3.
Neurotherapeutics ; 20(2): 502-517, 2023 03.
Article in English | MEDLINE | ID: mdl-36917440

ABSTRACT

Focused ultrasound (FUS) has the potential to modulate regional brain excitability and possibly aid seizure control; however, effects on behavior of FUS used as a seizure therapy are unknown. This study explores behavioral effects and hippocampal restoration induced by pulsed FUS in a kainic acid (KA) animal model of temporal lobe epilepsy. Twenty-nine male Sprague-Dawley rats were observed for 20 weeks with anatomical magnetic resonance imaging (MRI) and behavioral performance evaluations, comprising measures of anxiety, limb usage, sociability, and memory. FUS targeted to the right hippocampus was given 9 and 14 weeks after KA was delivered to the right amygdala. Ultrasound pulsations were delivered with the acoustic settings of 0.25 of mechanical index, 0.5 W/cm2 of intensity spatial peak temporal average (ISPTA), 100 Hz of pulse repetition frequency, and 30% of duty cycle, during three consecutive pulse trains of 10 min separated by 5-min rests. Controls included normal animals with sham injections and KA-exposed animals without FUS exposure. Longitudinal MRI observations showed that FUS substantially protected hippocampal and striatal structures from KA-induced atrophy. KA alone increased anxiety, impaired contralateral limb usage, and reduced sociability and learning. Two courses of FUS sonications partially ameliorated these impairments by enhancing exploring and learning, balancing limb usage, and increasing social interaction. The histology results indicated that two sonications enhanced neuroprotection effect and decreased the inflammation markers induced by KA. This study supports existence of both neuroprotective and beneficial behavioral effects from low-intensity pulsed ultrasound in the KA animal model of epilepsy.


Subject(s)
Epilepsy , Kainic Acid , Rats , Male , Animals , Kainic Acid/toxicity , Rats, Sprague-Dawley , Hippocampus , Epilepsy/chemically induced , Epilepsy/diagnostic imaging , Epilepsy/therapy , Seizures , Disease Models, Animal
4.
Int J Mol Sci ; 24(3)2023 Jan 30.
Article in English | MEDLINE | ID: mdl-36768901

ABSTRACT

Transcranial focused ultrasound (tFUS) is a novel neuromodulating technique. It has been demonstrated that the neuromodulatory effects can be induced by weak ultrasound exposure levels (spatial-peak temporal average intensity, ISPTA < 10 mW/cm2) in vitro. However, fewer studies have examined the use of weak tFUS to potentially induce long-lasting neuromodulatory responses in vivo. The purpose of this study was to determine the lower-bound threshold of tFUS stimulation for inducing neuromodulation in the motor cortex of rats. A total of 94 Sprague-Dawley rats were used. The sonication region aimed at the motor cortex under weak tFUS exposure (ISPTA of 0.338-12.15 mW/cm2). The neuromodulatory effects of tFUS on the motor cortex were evaluated by the changes in motor-evoked potentials (MEPs) elicited by transcranial magnetic stimulation (TMS). In addition to histology analysis, the in vitro cell culture was used to confirm the neuromodulatory mechanisms following tFUS stimulation. In the results, the dose-dependent inhibitory effects of tFUS were found, showing increased intensities of tFUS suppressed MEPs and lasted for 30 min. Weak tFUS significantly decreased the expression of excitatory neurons and increased the expression of inhibitory GABAergic neurons. The PIEZO-1 proteins of GABAergic neurons were found to involve in the inhibitory neuromodulation. In conclusion, we show the use of weak ultrasound to induce long-lasting neuromodulatory effects and explore the potential use of weak ultrasound for future clinical neuromodulatory applications.


Subject(s)
Motor Cortex , Rats , Animals , Rats, Sprague-Dawley , Motor Cortex/diagnostic imaging , Motor Cortex/physiology , Ultrasonography , Transcranial Magnetic Stimulation , GABAergic Neurons , Evoked Potentials, Motor
5.
Neurotherapeutics ; 19(4): 1368-1380, 2022 07.
Article in English | MEDLINE | ID: mdl-35581489

ABSTRACT

Focused ultrasound (FUS) has potential utility for modulating regional brain excitability and possibly aiding seizure control; however, the duration of any beneficial effect is unknown. This study explores the efficacy and time course of a short series of pulsed FUS in suppressing EEG epileptiform spikes/bursts in a kainic acid (KA) animal model of temporal lobe epilepsy. Forty-four male Sprague-Dawley rats were recorded for 14 weeks with EEG while software calculated EEG numbers of epileptiform spikes and bursts (≥ 3 spikes/s). Four regimens of FUS given in a single session at week 7 were evaluated, with mechanical index (MI) ranging from 0.25 to 0.75, intensity spatial peak temporal average (ISPTA) from 0.1 to 2.8 W per cm2, duty cycle from 1 to 30%, and three consecutive pulse trains for 5 or 10 min each. Controls included sham injections in four and KA without FUS in eleven animals. Histological analysis investigated tissue effects. All animals receiving KA evidenced EEG spikes, averaging 10,378 ± 1651 spikes per 8 h and 1255 ± 199 bursts per 8 h by weeks 6-7. The KA-only group showed a 30% of increase in spikes and bursts by week 14. Compared to the KA-only group, spike counts were reduced by about 25%, burst counts by about 33%, and burst durations by about 50% with FUS. Behavioral seizures were not analyzed, but electrographic seizures longer than 10 s declined up to 70% after some FUS regimens. Repeated-measure ANOVA showed a significant effect of higher intensity and longer sonication duration FUS treatment using 0.75-MI, ISPTA 2.8 W/cm2, 30% duty cycle for 10-min sonications (group effect, F (4, 15) = 6.321, p < 0.01; interaction effect, F (44, 165) = 1.726, p < 0.01), with the hippocampal protective effect lasting to week 14, accompanied by decreased inflammation and gliosis effect. In contrast, spike and burst suppression were achieved using an FUS regimen with 0.25-MI ISPTA 0.5 W/cm2, 30% duty cycle for 10-min sonications. This regimen reduced inflammation and gliosis at weeks 8-14 and protected hippocampal tissue. This study demonstrates that low-intensity pulsed ultrasound can modulate epileptiform activity for up to 7 weeks and, if replicated in the clinical setting, might be a practical treatment for epilepsy.


Subject(s)
Epilepsy , Kainic Acid , Animals , Rats , Male , Kainic Acid/toxicity , Rats, Sprague-Dawley , Electroencephalography , Gliosis , Epilepsy/chemically induced , Epilepsy/therapy , Seizures/chemically induced , Seizures/therapy , Hippocampus , Inflammation
6.
Sci Rep ; 8(1): 4151, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29515222

ABSTRACT

Focused ultrasound (FUS) exposure with microbubbles can transiently open the blood-brain barrier (BBB) to deliver therapeutic molecules into CNS tissues. However, delivered molecular distribution/concentration at the target need to be controlled. Dynamic Contrast-Enhanced Magnetic-Resonance Imaging (DCE-MRI) is a well-established protocol for monitoring the pharmacokinetic/pharmacodynamic behavior of FUS-BBB opening. This study investigates the feasibility of using DCE-MRI to estimate molecular CNS penetration under various exposure conditions and molecule sizes. In the 1st stage, a relationship among the imaging index Ktrans, exposure level and molecular size was calibrated and established. In the 2nd stage, various exposure levels and distinct molecules were applied to evaluate the estimated molecular concentration discrepancy with the quantified ones. High correlation (r2 = 0.9684) between Ktrans and transcranial mechanical index (MI) implies Ktrans can serve as an in vivo imaging index to mirror FUS-BBB opening scale. When testing various molecules with the size ranging 1-149 kDa, an overall correlation of r2 = 0.9915 between quantified and predicted concentrations was reached, suggesting the established model can provide reasonably accurate estimation. Our work demonstrates the feasibility of estimating molecular penetration through FUS-BBB opening via DCE-MRI and may facilitate development of FUS-induced BBB opening in brain drug delivery.


Subject(s)
Blood-Brain Barrier , Contrast Media , Drug Delivery Systems/methods , Magnetic Resonance Imaging , Microbubbles , Ultrasonic Therapy , Animals , Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/metabolism , Contrast Media/pharmacokinetics , Contrast Media/pharmacology , Male , Mice , Rats, Sprague-Dawley
7.
Sci Rep ; 7: 46689, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28425493

ABSTRACT

Microbubbles (MBs) serve as a critical catalyst to amplify local cavitation in CNS capillary lumen to facilitate focused ultrasound (FUS) to transiently open the blood-brain barrier (BBB). However, limited understanding is available regarding the effect of different microbubbles to induce BBB opening. The aim of this study is to characterize different MBs on their effect in FUS-induced BBB opening. Three MBs, SonoVue, Definity, and USphere, were tested, with 0.4-MHz FUS exposure at 0.62-1.38 of mechanical index (MI) on rats. Evans blue, dynamic contrast-enhanced (DCE) MRI and small-animal ultrasound imaging were used as surrogates to allow molecule-penetrated quantification, BBB-opened observation, and MBs circulation/persistence. Cavitation activity was measured via the passive cavitation detection (PCD) setup to correlate with the exposure level and the histological effect. Under given and identical MB concentrations, the three MBs induced similar and equivalent BBB-opening effects and persistence. In addition, a treatment paradigm by adapting exposure time is proposed to compensate MB decay to retain the persistence of BBB-opening efficiency in multiple FUS exposures. The results potentially improve understanding of the equivalence among MBs in focused ultrasound CNS drug delivery, and provide an effective strategy for securing persistence in this treatment modality.


Subject(s)
Blood-Brain Barrier/drug effects , Contrast Media/administration & dosage , Microbubbles , Ultrasonic Therapy/methods , Animals , Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/metabolism , Capillary Permeability/drug effects , Magnetic Resonance Imaging/methods , Male , Rats, Sprague-Dawley
8.
Anal Chem ; 88(22): 10971-10978, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27750431

ABSTRACT

Chemical analysis of Chinese black ink on xuan paper is useful for the authentication of Asian artwork. The analysis has to be nondestructive and has to accommodate artworks of all sizes. We apply three analytical techniques, ArF laser-induced plume fluorescence, Fourier transform infrared (FTIR) spectroscopy, and portable X-ray fluorescence (pXRF) to analyze five commercial Chinese black inks on two kinds of xuan paper. The FTIR signal is found to be interfered by the substrate which is inevitable because the pigments diffuse extensively into the xuan fiber network. The XRF signal is shown to be feeble and no signal can be registered until the samples are stacked and when the analytes are present at tens of percent. In contrast, the plume fluorescence technique can detect the minor and trace signature elements. The method is based on a two-laser-pulse scheme performed on a high precision optical setup: the first 355 nm laser pulse ablates a thin layer of the ink to create a plume; the second 193 nm laser pulse induces multi analytes in the plume to fluoresce. Partial-least-squares discriminant analysis of the fluorescence spectra unambiguously sorts the ink-xuan combinations while the sampled area is not visibly damaged even under the microscope. The laser probe can handle samples of arbitrary size and shape, is air compatible, and no sample pretreatment is necessary.

9.
Sci Rep ; 6: 33264, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27630037

ABSTRACT

Focused ultrasound (FUS) with microbubbles can temporally open the blood-brain barrier (BBB), and the cavitation activities of microbubbles play a key role in the BBB-opening process. Previous attempts used contrast-enhanced magnetic resonance imaging (CE-MRI) to correlate the mechanical index (MI) with the scale of BBB-opening, but MI only partially gauged acoustic activities, and CE-MRI did not fully explore correlations of pharmacodynamic/pharmacokinetic behaviors. Recently, the cavitation index (CI) has been derived to serve as an indicator of microbubble-ultrasound stable cavitation, and may also serve as a valid indicator to gauge the level of FUS-induced BBB opening. This study investigates the feasibility of gauging FUS-induced BBB opened level via the two indexes, MI and CI, through dynamic contrast-enhanced (DCE)-MRI analysis as well as passive cavitation detection (PCD) analysis. Pharmacodynamic/pharmacokinetic parameters derived from DCE-MRI were characterized to identify the scale of FUS-induced BBB opening. Our results demonstrated that DCE-MRI can successfully access pharmacodynamic/pharmacokinetic BBB-opened behavior, and was highly correlated both with MI and CI, implying the feasibility in using these two indices to gauge the scale of FUS-induced BBB opening. The proposed finding may facilitate the design toward using focused ultrasound as a safe and reliable noninvasive CNS drug delivery.


Subject(s)
Blood-Brain Barrier/diagnostic imaging , Drug Delivery Systems , Magnetic Resonance Imaging/methods , Ultrasonic Therapy/methods , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Contrast Media/pharmacology , Disease Models, Animal , Humans , Microbubbles/therapeutic use , Rats
10.
Radiology ; 281(1): 99-108, 2016 10.
Article in English | MEDLINE | ID: mdl-27192459

ABSTRACT

Purpose To demonstrate that magnetic resonance (MR) imaging-monitored transcranial focused ultrasound can enhance the delivery of the antiangiogenic monoclonal antibody bevacizumab into the central nervous system (CNS) for glioblastoma multiforme (GBM) treatment. Materials and Methods All animal experiments were approved by the animal committee and adhered to experimental animal care guidelines. Transcranial focused ultrasound exposure in the presence of microbubbles was used to open the blood-brain barrier (BBB) to enhance bevacizumab penetration into the CNS in healthy and glioma-bearing mice. Bevacizumab concentration was quantitated with high-performance liquid chromatography, and Western blot testing was performed to confirm the specific biologic form in the CNS. Penetration of bevacizumab into brain tissue was estimated in vivo by means of contrast material-enhanced MR imaging and quantitative gallium 68 ((68)Ga)-bevacizumab micro-positron emission tomography, and glioma progression was longitudinally followed with T2-weighted MR imaging. Hematoxylin-eosin staining and cluster of differentiation 31 immunostaining were used to assess morphologic changes and vascular inhibition at histologic examination. The two-tailed Student t test and the Mantel-Cox log-rank test were used for statistical analyses, with a significance level of .05. Results Focused ultrasound significantly enhanced bevacizumab penetration into the CNS by 5.7- to 56.7-fold compared with that in nonexposed brain (both P < .0001). Contrast-enhanced MR imaging indexes correlated with bevacizumab concentration (r = 0.748-0.857) in vivo. Focused ultrasound-enhanced bevacizumab delivery significantly retarded glioma progression, with a significantly increased median survival (median increase in survival time = 135% in the group treated with bevacizumab and focused ultrasound, P < .0001; as compared with 48% in the group treated with bevacizumab alone, P = .0002). Conclusion Focused ultrasound-enhanced bevacizumab delivery can provide an antivascularization normalization effect to suppress glioma. (©) RSNA, 2016 Online supplemental material is available for this article.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Bevacizumab/therapeutic use , Blood-Brain Barrier , Brain Neoplasms/drug therapy , Glioma/drug therapy , Ultrasonic Therapy/methods , Animals , Blotting, Western , Brain Neoplasms/diagnostic imaging , Chromatography, High Pressure Liquid , Contrast Media , Disease Models, Animal , Disease Progression , Gadolinium DTPA , Glioma/diagnostic imaging , Longitudinal Studies , Magnetic Resonance Imaging , Mice , Microbubbles , Positron-Emission Tomography , Treatment Outcome
11.
Sci Rep ; 5: 15477, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26490653

ABSTRACT

Burst-mode focused ultrasound (FUS) induces microbubble cavitation in the vasculature and temporarily disrupts the blood-brain barrier (BBB) to enable therapeutic agent delivery. However, it remains unclear whether FUS-induced BBB opening is accompanied by neuromodulation. Here we characterized the functional effects of FUS-induced BBB opening by measuring changes in somatosensory evoked potentials (SSEPs) and blood-oxygen-level dependent (BOLD) responses. Rats underwent burst-mode FUS (mechanical index (MI) of 0.3, 0.55 or 0.8) to the forelimb region in the left primary somatosensory cortex to induce BBB opening. Longitudinal measurements were followed for up to 1 week to characterize the temporal dynamics of neuromodulation. We observed that 0.8-MI FUS profoundly suppressed SSEP amplitude and prolonged latency, and this effect lasted 7 days. 0.55-MI FUS resulted in minimal and short-term suppression of SSEP for less than 60 minutes and didn't affect latency. BOLD responses were also suppressed in an MI-dependent manner, mirroring the effect on SSEPs. Furthermore, repetitive delivery of 0.55-MI FUS every 3 days elicited no accumulative effects on SSEPs or tissue integrity. This is the first evidence that FUS-induced BBB opening is accompanied by reversible changes in neuron responses, and may provide valuable insight toward the development of FUS-induced BBB opening for clinical applications.


Subject(s)
Blood-Brain Barrier/diagnostic imaging , Drug Delivery Systems , Neurotransmitter Agents/therapeutic use , Ultrasonic Waves , Animals , Biological Transport , Blood-Brain Barrier/metabolism , Evoked Potentials, Somatosensory/radiation effects , Magnetic Resonance Imaging , Neurons/drug effects , Neurons/radiation effects , Radiography , Rats , Ultrasonography
12.
J Control Release ; 192: 1-9, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-24969355

ABSTRACT

Focused ultrasound (FUS) with the presence of microbubbles has been shown to induce transient and local opening of the blood-brain barrier (BBB) for the delivery of therapeutic molecules which normally cannot penetrate into the brain. The success of FUS brain-drug delivery relies on its integration with in-vivo imaging to monitor kinetic change of therapeutic molecules into the brain. In this study, we developed a dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) technique for kinetic analysis of delivered molecules during FUS-BBB opening. Three kinetic parameters (Ktrans, Ve, Kep) were characterized dynamically to describe BBB-permeability at two FUS exposure conditions (0.4 or 0.8MPa) over 24h. Ktrans, defined as the influx volume transfer constant from plasma to EES, and Ve, the EES volume fraction, were both found to be pressure-dependent. Ktrans and Ve showed a peak increase of 0.0086-0.0131min(-1) (for 0.4-0.8MPa pressure), and 0.0431-0.0692, respectively, immediately after FUS exposure. Both parameters subsequently decreased exponentially as a function of time, with estimated half-lives of decay of 2.89-5.3 and 2.2-4.93h, respectively. The kinetics of Kep, defined as the efflux rate constant from the extracellular extravascular space (EES) to the plasma, were complementary to Ktrans, with an initial decrease from 0.2010 to 0.1901min(-1) followed by a significantly longer recovery time (half-life of 17.39-99.92h). Our observations strongly supported the existence of imbalanced and mismatched kinetics of influx (Ktrans) and efflux (Kep) between the plasma and EES, indicating the existence of directional permeability during FUS-BBB opening. We further showed that kinetic change determined by DCE-MRI correlated well with the concentration of Evans Blue (EB)-albumin (coefficient of 0.74-0.89). These findings suggest that MRI kinetic monitoring may serve as an alternative method for in-vivo monitoring of pharmacokinetics and pharmacodynamics (PK/PD) change of therapeutic agents during drug delivery to the brain, and provide useful information for future optimization of FUS-BBB opening.


Subject(s)
Blood-Brain Barrier/metabolism , Coloring Agents/administration & dosage , Drug Delivery Systems/methods , Evans Blue/administration & dosage , Magnetic Resonance Imaging , Microbubbles , Ultrasonics/methods , Animals , Brain/metabolism , Brain/ultrastructure , Coloring Agents/pharmacokinetics , Evans Blue/pharmacokinetics , Male , Rats, Sprague-Dawley
13.
IEEE Trans Biomed Eng ; 61(4): 1350-60, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24658258

ABSTRACT

Focused ultrasound (FUS) in the presence of microbubbles can bring about transcranial and local opening of the blood-brain barrier (BBB) for potential noninvasive delivery of drugs to the brain. A phased-array ultrasound system is essential for FUS-BBB opening to enable electronic steering and correction of the focal beam which is distorted by cranial bone. Here, we demonstrate our prototype design of a 256-channel ultrasound phased-array system for large-region transcranial BBB opening in the brains of large animals. One of the unique features of this system is the capability of generating concurrent dual-frequency ultrasound signals from the driving system for potential enhancement of BBB opening. A wide range of signal frequencies can be generated (frequency = 0.2-1.2 MHz) with controllable driving burst patterns. Precise output power can be controlled for individual channels via 8-bit duty-cycle control of transistor-transistor logic signals and the 8-bit microcontroller-controlled buck converter power supply output voltage. The prototype system was found to be in compliance with the electromagnetic compatibility standard. Moreover, large animal experiments confirmed the phase switching effectiveness of this system, and induction of either a precise spot or large region of BBB opening through fast focal-beam switching. We also demonstrated the capability of dual-frequency exposure to potentially enhance the BBB-opening effect. This study contributes to the design of ultrasound phased arrays for future clinical applications, and provides a new direction toward optimizing FUS brain drug delivery.


Subject(s)
Drug Delivery Systems/methods , Ultrasonography, Interventional/methods , Animals , Blood-Brain Barrier/diagnostic imaging , Echoencephalography , Equipment Design , Humans , Skull/diagnostic imaging , Swine , Ultrasonography, Interventional/instrumentation
14.
Biomed Res Int ; 2013: 627496, 2013.
Article in English | MEDLINE | ID: mdl-23607093

ABSTRACT

Microbubble-enhanced focused ultrasound (FUS) can enhance the delivery of therapeutic agents into the brain for brain tumor treatment. The purpose of this study was to investigate the influence of brain tumor conditions on the distribution and dynamics of small molecule leakage into targeted regions of the brain after FUS-BBB opening. A total of 34 animals were used, and the process was monitored by 7T-MRI. Evans blue (EB) dye as well as Gd-DTPA served as small molecule substitutes for evaluation of drug behavior. EB was quantified spectrophotometrically. Spin-spin (R1) relaxometry and area under curve (AUC) were measured by MRI to quantify Gd-DTPA. We found that FUS-BBB opening provided a more significant increase in permeability with small tumors. In contrast, accumulation was much higher in large tumors, independent of FUS. The AUC values of Gd-DTPA were well correlated with EB delivery, suggesting that Gd-DTPA was a good indicator of total small-molecule accumulation in the target region. The peripheral regions of large tumors exhibited similar dynamics of small-molecule leakage after FUS-BBB opening as small tumors, suggesting that FUS-BBB opening may have the most significant permeability-enhancing effect on tumor peripheral. This study provides useful information toward designing an optimized FUS-BBB opening strategy to deliver small-molecule therapeutic agents into brain tumors.


Subject(s)
Blood-Brain Barrier/radiation effects , Brain Neoplasms/drug therapy , Drug Delivery Systems , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Evans Blue/pharmacology , Gadolinium DTPA/pharmacology , Humans , Magnetic Resonance Imaging , Radiography , Rats , Sound
15.
Anal Chem ; 85(9): 4311-5, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23570307

ABSTRACT

We demonstrated a minimally destructive two-tier approach for multielement forensic analysis of laser-printed ink. The printed document was first screened using a portable-X-ray fluorescence (XRF) probe. If the results were not conclusive, a laser microprobe was then deployed. The laser probe was based on a two-pulse scheme: the first laser pulse ablated a thin layer of the printed ink; the second laser pulse at 193 nm induced multianalytes in the desorbed ink to fluoresce. We analyzed four brands of black toners. The toners were printed on paper in the form of patches or letters or overprinted on another ink. The XRF probe could sort the four brands if the printed letters were larger than font 20. It could not tell the printing sequence in the case of overprints. The laser probe was more discriminatory; it could sort the toner brands and reveal the overprint sequence regardless of font size while the sampled area was not visibly different from neighboring areas even under the microscope. In terms of general analytical performance, the laser probe featured tens of micrometer lateral resolution and tens to hundreds of nm depth resolution and atto-mole mass detection limits. It could handle samples of arbitrary size and shape and was air compatible, and no sample pretreatment was necessary. It will prove useful whenever high-resolution and high sensitivity 3D elemental mapping is required.


Subject(s)
Fluorescence , Forensic Sciences/methods , Ink , Lasers , Printing , Fluorescent Dyes/chemistry , Particle Size , Spectrometry, X-Ray Emission/instrumentation , Surface Properties
16.
PLoS One ; 8(3): e58995, 2013.
Article in English | MEDLINE | ID: mdl-23527068

ABSTRACT

The purpose of this study is to assess the preclinical therapeutic efficacy of magnetic resonance imaging (MRI)-monitored focused ultrasound (FUS)-induced blood-brain barrier (BBB) disruption to enhance Temozolomide (TMZ) delivery for improving Glioblastoma Multiforme (GBM) treatment. MRI-monitored FUS with microbubbles was used to transcranially disrupt the BBB in brains of Fisher rats implanted with 9L glioma cells. FUS-BBB opening was spectrophotometrically determined by leakage of dyes into the brain, and TMZ was quantitated in cerebrospinal fluid (CSF) and plasma by LC-MS\MS. The effects of treatment on tumor progression (by MRI), animal survival and brain tissue histology were investigated. Results demonstrated that FUS-BBB opening increased the local accumulation of dyes in brain parenchyma by 3.8-/2.1-fold in normal/tumor tissues. Compared to TMZ alone, combined FUS treatment increased the TMZ CSF/plasma ratio from 22.7% to 38.6%, reduced the 7-day tumor progression ratio from 24.03 to 5.06, and extended the median survival from 20 to 23 days. In conclusion, this study provided preclinical evidence that FUS BBB-opening increased the local concentration of TMZ to improve the control of tumor progression and animal survival, suggesting its clinical potential for improving current brain tumor treatment.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/radiation effects , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Sound , Animals , Antineoplastic Agents, Alkylating/pharmacokinetics , Brain/pathology , Brain Neoplasms/diagnosis , Brain Neoplasms/mortality , Cell Line, Tumor , Dacarbazine/pharmacokinetics , Dacarbazine/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical , Glioblastoma/diagnosis , Glioblastoma/mortality , Magnetic Resonance Imaging , Male , Rats , Temozolomide
17.
J Biomed Sci ; 19: 61, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-22734518

ABSTRACT

BACKGROUND: Rabies is known to be lethal in human. Treatment with passive immunity for the rabies is effective only when the patients have not shown the central nerve system (CNS) signs. The blood-brain barrier (BBB) is a complex functional barrier that may compromise the therapeutic development in neurological diseases. The goal of this study is to determine the change of BBB integrity and to assess the therapeutic possibility of enhancing BBB permeability combined with passive immunity in the late stage of rabies virus infection. METHODS: The integrity of BBB permeability in rats was measured by quantitative ELISA for total IgG and albumin levels in the cerebrospinal fluid (CSF) and by exogenously applying Evans blue as a tracer. Western blotting of occludin and ZO-1, two tight junction proteins, was used to assess the molecular change of BBB structure.The breakdown of BBB with hypertonic arabinose, recombinant tumor necrosis factor-alpha (rTNF-γ), and focused ultrasound (FUS) were used to compare the extent of BBB disruption with rabies virus infection. Specific humoral immunity was analyzed by immunofluorescent assay and rapid fluorescent focus inhibition test. Virus-neutralizing monoclonal antibody (mAb) 8-10E was administered to rats with hypertonic breakdown of BBB as a passive immunotherapy to prevent the death from rabies. RESULTS: The BBB permeability was altered on day 7 post-infection. Increased BBB permeability induced by rabies virus infection was observed primarily in the cerebellum and spinal cord. Occludin was significantly decreased in both the cerebral cortex and cerebellum. The rabies virus-specific antibody was not strongly elicited even in the presence of clinical signs. Disruption of BBB had no direct association with the lethal outcome of rabies. Passive immunotherapy with virus-neutralizing mAb 8-10E with the hypertonic breakdown of BBB prolonged the survival of rabies virus-infected rats. CONCLUSIONS: We demonstrated that the BBB permeability was altered in a rat model with rabies virus inoculation. Delivery of neutralizing mAb to the infected site in brain combined with effective breakdown of BBB could be an aggressive but feasible therapeutic mode in rabies when the CNS infection has been established.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Blood-Brain Barrier , Central Nervous System , Rabies virus/pathogenicity , Rabies , Albumins/cerebrospinal fluid , Animals , Antibodies, Monoclonal/immunology , Blood-Brain Barrier/immunology , Blood-Brain Barrier/physiopathology , Blood-Brain Barrier/virology , Central Nervous System/pathology , Central Nervous System/virology , Humans , Immunoglobulin G/cerebrospinal fluid , Occludin/metabolism , Rabies/immunology , Rabies/pathology , Rabies/therapy , Rabies/virology , Rabies virus/immunology , Rats , Zonula Occludens-1 Protein/metabolism
18.
ACS Nano ; 6(2): 1795-805, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22248493

ABSTRACT

A key issue in cancer therapy is how to enhance the tumor-targeting efficacy of chemotherapeutic agents. In this study, we developed a cooperative dual-targeted delivery platform for paclitaxel (PTX) that has potential application as a powerful prostate cancer treatment. The nanomedicine was prepared by first conjugating PTX to nontoxic high-magnetization nanocarriers which can be actively guided and targeted by an external magnet. Next, the surface was functionalized with carboxylated o-(2-aminoethyl)polyethyleneglycol (NH(2)-EPEG-COOH) to enable uptake by the reticuloendothelial system. Antiprostate-specific membrane antigen antibodies (APSMAs) were then conjugated onto the carrier to recognize the extracellular domain of the prostate-cancer specific membrane antigen (PSMA), thus binding to cancer cells as a secondary active targeting mechanism. We found a significant enhancement of PTX concentration at the tumor site by nearly 20-fold. In addition, the drug half-life was prolonged more than 4.1-fold (from 24 to 99 h) at 37 °C. Low-dose (4.5 mg/kg) injection of the dual-targeted therapeutic nanomedicine in the presence of magnetic targeting significantly prolonged the median survival of nude mice from 35 to 58 days compared to mice that received a high dose (6 mg/kg) of free PTX. This report demonstrates the potential utility of targeted nanomedicine in the clinical treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Molecular Targeted Therapy/methods , Nanomedicine/methods , Paclitaxel/pharmacology , Prostatic Neoplasms/drug therapy , Animals , Antibodies, Neoplasm/chemistry , Antibodies, Neoplasm/immunology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Contrast Media , Drug Carriers/chemistry , Humans , Magnetic Resonance Imaging , Magnets/chemistry , Male , Mice , Paclitaxel/chemistry , Paclitaxel/therapeutic use , Polyethylene Glycols/chemistry , Prostate-Specific Antigen/immunology , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
19.
Angew Chem Int Ed Engl ; 50(35): 8086-9, 2011 Aug 22.
Article in English | MEDLINE | ID: mdl-21751316

ABSTRACT

Prepared to self-destruct: when poly(D, L-lactic-co-glycolic acid) (PLGA) hollow microspheres containing NaHCO(3) entered the endocytic organelles of a live cell, the NaHCO(3) in the aqueous core reacted with protons that infiltrated from the compartment to generate CO(2) gas. The evolution of CO(2) bubbles led to the formation of small holes in the PLGA shell and thus rapid release of the encapsulated drug doxorubicin.


Subject(s)
Lysosomes/chemistry , Microspheres , Pharmaceutical Preparations/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/chemistry , Doxorubicin/toxicity , Drug Carriers/chemistry , Humans , Hydrogen-Ion Concentration , Lactic Acid/chemistry , Lysosomes/metabolism , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Sodium Bicarbonate/chemistry
20.
Proc Natl Acad Sci U S A ; 107(34): 15205-10, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20696897

ABSTRACT

The superparamagnetic properties of magnetic nanoparticles (MNPs) allow them to be guided by an externally positioned magnet and also provide contrast for MRI. However, their therapeutic use in treating CNS pathologies in vivo is limited by insufficient local accumulation and retention resulting from their inability to traverse biological barriers. The combined use of focused ultrasound and magnetic targeting synergistically delivers therapeutic MNPs across the blood-brain barrier to enter the brain both passively and actively. Therapeutic MNPs were characterized and evaluated both in vitro and in vivo, and MRI was used to monitor and quantify their distribution in vivo. The technique could be used in normal brains or in those with tumors, and significantly increased the deposition of therapeutic MNPs in brains with intact or compromised blood-brain barriers. Synergistic targeting and image monitoring are powerful techniques for the delivery of macromolecular chemotherapeutic agents into the CNS under the guidance of MRI.


Subject(s)
Brain Neoplasms/drug therapy , Drug Delivery Systems/methods , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/therapeutic use , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/therapeutic use , Blood-Brain Barrier , Brain Neoplasms/blood supply , Brain Neoplasms/ultrastructure , Contrast Media , Epirubicin/administration & dosage , Epirubicin/therapeutic use , Magnetic Resonance Imaging , Magnetics , Metal Nanoparticles/ultrastructure , Microscopy, Electron, Transmission , Rats , Rats, Sprague-Dawley , Ultrasonic Therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...