Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-38625763

RESUMO

Transcranial focused ultrasound (tFUS) has emerged as a new mode of non-invasive brain stimulation (NIBS), with its exquisite spatial precision and capacity to reach the deep regions of the brain. The placement of the acoustic focus onto the desired part of the brain is critical for successful tFUS procedures; however, acoustic wave propagation is severely affected by the skull, distorting the focal location/shape and the pressure level. High-resolution (HR) numerical simulation allows for monitoring of acoustic pressure within the skull but with a considerable computational burden. To address this challenge, we employed a 4x super-resolution (SR) Swin Transformer method to improve the precision of estimating tFUS acoustic pressure field, targeting operator-defined brain areas. The training datasets were obtained through numerical simulations at both ultra-low (2.0 mm) and high (0.5 mm) resolutions, conducted on in vivo CT images of 12 human skulls. Our multivariable datasets, which incorporate physical properties of the acoustic pressure field, wave velocity, and skull CT images, were utilized to train three-dimensional SR models. We found that our method yielded 87.99?4.28% accuracy in terms of focal volume conformity under foreseen skull data, and accuracy of 82.32?5.83% for unforeseen skulls, respectively. Moreover, a significant improvement of 99.4% in computational efficiency compared to the traditional 0.5 mm HR numerical simulation was shown. The presented technique, when adopted in guiding the placement of the FUS transducer to engage specific brain targets, holds great potential in enhancing the safety and effectiveness of tFUS therapy.

2.
Ultrasonography ; 43(1): 35-46, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38029736

RESUMO

PURPOSE: Acoustic streaming induced by applying transcranial focused ultrasound (FUS) promotes localized advective solute transport in the brain and has recently garnered research interest for drug delivery and enhancement of brain waste clearance. The acoustic streaming behavior in brain tissue is difficult to model numerically and thus warrants an in vitro examination of the effects of using different sonication parameters, in terms of frequency, intensity, and pulse duration (PD). METHODS: Melamine and polyvinyl alcohol (PVA) foams were used to mimic the porous brain tissue, which contains leptomeningeal fenestrations and perivascular space, while agar hydrogel was used to emulate denser neuropil. FUS was delivered to these media, which were immersed in a phosphate-buffered saline containing toluidine blue O dye, across various frequencies (400, 500, and 600 kHz; applicable to transcranial delivery) in a pulsed mode at two different spatialpeak pulse-average intensities (3 and 4 W/cm2). RESULTS: Image analysis showed that the use of 400 kHz yielded the greatest dye infiltration in melamine foam, while sonication had no impact on infiltration in the agar hydrogel due to the dominance of diffusional transport. Using a fixed spatial-peak temporal-average intensity of 0.4 W/cm2 at 400 kHz, a PD of 75 ms resulted in the greatest infiltration depth in both melamine and PVA foams among the tested range (50-150 ms). CONCLUSION: These findings suggest the existence of a specific frequency and PD that induce greater enhancement of solute/fluid movement, which may contribute to eventual in vivo applications in promoting waste clearance from the brain.

3.
Anticancer Res ; 43(11): 4793-4800, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37909989

RESUMO

BACKGROUND/AIM: We examined the effect of low-intensity focused ultrasound (FUS) on unbinding cisplatin from plasma proteins and enhancing its chemotherapeutic efficacy using a mouse model of xenograft human cervical cancer. MATERIALS AND METHODS: FUS, operating in a pulsed mode, was applied to a dialysis cassette immersed in a normal saline bath containing both bovine serum albumin (BSA) and cisplatin, and the unbound level of cisplatin diffused into the cassette was measured. To assess the in vivo efficacy of the technique, athymic nu/nu mice were inoculated with human cervical cancer cells under four different combinatory conditions, with and without the administration of cisplatin and FUS. FUS was delivered to the tumor mass for 1 h across four separate sessions spanning a period of 10 days, following the intraperitoneal injection of cisplatin. RESULTS: In vitro equilibrium dialysis revealed that non-thermal application of FUS increased the concentration of unbound cisplatin compared to cassettes that were not exposed to sonication, suggesting successful unbinding. Assessment of tumor growth in vivo showed that FUS following cisplatin administration resulted in a significant reduction in tumor growth, whereas the administration of cisplatin alone exhibited plateau growth. Without administration of cisplatin, equivalent rates of aggressive tumor growth were observed regardless of the application of FUS. CONCLUSION: Pulsed application of FUS can unbind cisplatin from albumin and enhance its tumoricidal effects in cervical cancer. Further assessment of intratumoral/systemic cisplatin concentration is required to quantify its selective delivery to the tumor.


Assuntos
Neoplasias do Colo do Útero , Humanos , Feminino , Neoplasias do Colo do Útero/tratamento farmacológico , Xenoenxertos , Cisplatino/farmacologia , Transplante Heterólogo , Acústica
4.
Sci Rep ; 13(1): 17002, 2023 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-37813871

RESUMO

Cerebrospinal fluid (CSF) is crucial for maintaining neuronal homeostasis, providing nutrition, and removing metabolic waste from the brain. However, the relationship between neuronal activity and CSF solute transport remains poorly understood. To investigate the effect of regional neuronal activity on CSF solute transport, Sprague-Dawley rats (all male, n = 30) under anesthesia received an intracisternal injection of a fluorescent tracer (Texas Red ovalbumin) and were subjected to unilateral electrical stimulation of a forelimb. Two groups (n = 10 each) underwent two different types of stimulation protocols for 90 min, one including intermittent 7.5-s resting periods and the other without rest. The control group was not stimulated. Compared to the control, the stimulation without resting periods led to increased transport across most of the cortical areas, including the ventricles. The group that received intermittent stimulation showed an elevated level of solute uptake in limited areas, i.e., near/within the ventricles and on the ventral brain surface. Interhemispheric differences in CSF solute transport were also found in the cortical regions that overlap with the forelimb sensorimotor area. These findings suggest that neuronal activity may trigger local and brain-wide increases in CSF solute transport, contributing to waste clearance.


Assuntos
Encéfalo , Roedores , Ratos , Animais , Masculino , Ratos Sprague-Dawley , Encéfalo/metabolismo , Homeostase/fisiologia , Transporte Biológico/fisiologia , Líquido Cefalorraquidiano/metabolismo
5.
Sci Rep ; 13(1): 12339, 2023 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-37524783

RESUMO

Transport of interstitial fluid and solutes plays a critical role in clearing metabolic waste from the brain. Transcranial application of focused ultrasound (FUS) has been shown to promote localized cerebrospinal fluid solute uptake into the brain parenchyma; however, its effects on the transport and clearance of interstitial solutes remain unknown. We demonstrate that pulsed application of low-intensity FUS to the rat brain enhances the transport of intracortically injected fluorescent tracers (ovalbumin and high molecular-weight dextran), yielding greater parenchymal tracer volume distribution compared to the unsonicated control group (ovalbumin by 40.1% and dextran by 34.6%). Furthermore, FUS promoted the drainage of injected interstitial ovalbumin to both superficial and deep cervical lymph nodes (cLNs) ipsilateral to sonication, with 78.3% higher drainage observed in the superficial cLNs compared to the non-sonicated hemisphere. The application of FUS increased the level of solute transport visible from the dorsal brain surface, with ~ 43% greater area and ~ 19% higher fluorescence intensity than the unsonicated group, especially in the pial surface ipsilateral to sonication. The sonication did not elicit tissue-level neuronal excitation, measured by an electroencephalogram, nor did it alter the molecular weight of the tracers. These findings suggest that nonthermal transcranial FUS can enhance advective transport of interstitial solutes and their subsequent removal in a completely non-invasive fashion, offering its potential non-pharmacological utility in facilitating clearance of waste from the brain.


Assuntos
Encéfalo , Dextranos , Ratos , Animais , Ratos Sprague-Dawley , Ovalbumina/metabolismo , Dextranos/metabolismo , Encéfalo/fisiologia , Sonicação
6.
PLoS One ; 18(7): e0288654, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37478086

RESUMO

The effects of transcranial focused ultrasound (FUS) stimulation of the primary somatosensory cortex and its thalamic projection (i.e., ventral posterolateral nucleus) on the generation of electroencephalographic (EEG) responses were evaluated in healthy human volunteers. Stimulation of the unilateral somatosensory circuits corresponding to the non-dominant hand generated EEG evoked potentials across all participants; however, not all perceived stimulation-mediated tactile sensations of the hand. These FUS-evoked EEG potentials (FEP) were observed from both brain hemispheres and shared similarities with somatosensory evoked potentials (SSEP) from median nerve stimulation. Use of a 0.5 ms pulse duration (PD) sonication given at 70% duty cycle, compared to the use of 1 and 2 ms PD, elicited more distinctive FEP peak features from the hemisphere ipsilateral to sonication. Although several participants reported hearing tones associated with FUS stimulation, the observed FEP were not likely to be confounded by the auditory sensation based on a separate measurement of auditory evoked potentials (AEP) to tonal stimulation (mimicking the same repetition frequency as the FUS stimulation). Off-line changes in resting-state functional connectivity (FC) associated with thalamic stimulation revealed that the FUS stimulation enhanced connectivity in a network of sensorimotor and sensory integration areas, which lasted for at least more than an hour. Clinical neurological evaluations, EEG, and neuroanatomical MRI did not reveal any adverse or unintended effects of sonication, attesting its safety. These results suggest that FUS stimulation may induce long-term neuroplasticity in humans, indicating its neurotherapeutic potential for various neurological and neuropsychiatric conditions.


Assuntos
Mãos , Sensação , Humanos , Sensação/fisiologia , Potenciais Evocados Auditivos , Potenciais Somatossensoriais Evocados/fisiologia , Sonicação/métodos , Córtex Somatossensorial/diagnóstico por imagem , Córtex Somatossensorial/fisiologia
7.
Brain Cogn ; 169: 106001, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37235929

RESUMO

We systematically investigated the link between trait mindfulness scores and functional connectivity (FC) features or behavioral data, to emphasize the importance of the reliability of self-report mindfulness scores. Sixty healthy young male participants underwent two functional MRI runs with three mindfulness or mind-wandering task blocks with an N-back task (NBT) block. The data from 49 participants (age: 23.3 ± 2.8) for whom two sets of the self-reported Mindfulness Attention Awareness Scale (MAAS) and NBT performance were available were analyzed. We divided participants into two groups based on the consistency level of their MAAS scores (i.e., a "consistent" and an "inconsistent" group). Then, the association between the MAAS scores and FC features or NBT performance was investigated using linear regression analysis with p-value correction and bootstrapping. Meaningful associations (a) between MAAS and NBT accuracy (slope = 0.41, CI = [0.10, 0.73], corrected p < 0.05), (b) between MAAS and the FC edges in the frontoparietal network, and (c) between the FC edges and NBT performance were only observed in the consistent group (n = 26). Our findings demonstrate the importance of appropriate screening mechanisms for self-report-based dispositional mindfulness scores when trait mindfulness scores are combined with neuronal features and behavioral data.


Assuntos
Memória de Curto Prazo , Atenção Plena , Humanos , Masculino , Adulto Jovem , Adulto , Autorrelato , Reprodutibilidade dos Testes , Atenção/fisiologia
8.
Comput Methods Programs Biomed ; 237: 107591, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37182263

RESUMO

BACKGROUND AND OBJECTIVE: Transcranial focused ultrasound (tFUS) has emerged as a new non-invasive brain stimulation (NIBS) modality, with its exquisite ability to reach deep brain areas at a high spatial resolution. Accurate placement of an acoustic focus to a target region of the brain is crucial during tFUS treatment; however, the distortion of acoustic wave propagation through the intact skull casts challenges. High-resolution numerical simulation allows for monitoring of the acoustic pressure field in the cranium but also demands extensive computational loads. In this study, we adopt a super-resolution residual network technique based on a deep convolution to enhance the prediction quality of the FUS acoustic pressure field in the targeted brain regions. METHODS: The training dataset was acquired by numerical simulations performed at low-(1.0 mm) and high-resolutions (0.5mm) on three ex vivo human calvariae. Five different super-resolution (SR) network models were trained by using a multivariable dataset in 3D, which incorporated information on the acoustic pressure field, wave velocity, and localized skull computed tomography (CT) images. RESULTS: The accuracy of 80.87±4.50% in predicting the focal volume with a substantial improvement of 86.91% in computational cost compared to the conventional high-resolution numerical simulation was achieved. The results suggest that the method can greatly reduce the simulation time without sacrificing accuracy and improve the accuracy further with the use of additional inputs. CONCLUSIONS: In this research, we developed multivariable-incorporating SR neural networks for transcranial focused ultrasound simulation. Our super-resolution technique may contribute to promoting the safety and efficacy of tFUS-mediated NIBS by providing on-site feedback information on the intracranial pressure field to the operator.


Assuntos
Encéfalo , Crânio , Humanos , Encéfalo/diagnóstico por imagem , Crânio/diagnóstico por imagem , Simulação por Computador , Acústica , Cabeça
9.
Sci Rep ; 13(1): 4128, 2023 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-36914775

RESUMO

The efficacy of many anti-epileptic drugs, including phenytoin (PHT), is reduced by plasma protein binding (PPB) that sequesters therapeutically active drug molecules within the bloodstream. An increase in systemic dose elevates the risk of drug side effects, which demands an alternative technique to increase the unbound concentration of PHT in a region-specific manner. We present a low-intensity focused ultrasound (FUS) technique that locally enhances the efficacy of PHT by transiently disrupting its binding to albumin. We first identified the acoustic parameters that yielded the highest PHT unbinding from albumin among evaluated parameter sets using equilibrium dialysis. Then, rats with chronic mesial temporal lobe epilepsy (mTLE) received four sessions of PHT injection, each followed by 30 min of FUS delivered to the ictal region, across 2 weeks. Two additional groups of mTLE rats underwent the same procedure, but without receiving PHT or FUS. Assessment of electrographic seizure activities revealed that FUS accompanying administration of PHT effectively reduced the number and mean duration of ictal events compared to other conditions, without damaging brain tissue or the blood-brain barrier. Our results demonstrated that the FUS technique enhanced the anti-epileptic efficacy of PHT in a chronic mTLE rodent model by region-specific PPB disruption.


Assuntos
Epilepsia do Lobo Temporal , Fenitoína , Animais , Ratos , Anticonvulsivantes/uso terapêutico , Proteínas Sanguíneas/metabolismo , Epilepsia do Lobo Temporal/induzido quimicamente , Epilepsia do Lobo Temporal/tratamento farmacológico , Fenitoína/farmacologia , Fenitoína/uso terapêutico
10.
Brain Stimul ; 16(1): 48-55, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36549480

RESUMO

Transcranial ultrasound stimulation (TUS) has been shown to be a safe and effective technique for non-invasive superficial and deep brain stimulation. Safe and efficient translation to humans requires estimating the acoustic attenuation of the human skull. Nevertheless, there are no international guidelines for estimating the impact of the skull bone. A tissue independent, arbitrary derating was developed by the U.S. Food and Drug Administration to take into account tissue absorption (0.3 dB/cm-MHz) for diagnostic ultrasound. However, for the case of transcranial ultrasound imaging, the FDA model does not take into account the insertion loss induced by the skull bone, nor the absorption by brain tissue. Therefore, the estimated absorption is overly conservative which could potentially limit TUS applications if the same guidelines were to be adopted. Here we propose a three-layer model including bone absorption to calculate the maximum pressure transmission through the human skull for frequencies ranging between 100 kHz and 1.5 MHz. The calculated pressure transmission decreases with the frequency and the thickness of the bone, with peaks for each thickness corresponding to a multiple of half the wavelength. The 95th percentile maximum transmission was calculated over the accessible surface of 20 human skulls for 12 typical diameters of the ultrasound beam on the skull surface, and varies between 40% and 78%. To facilitate the safe adjustment of the acoustic pressure for short ultrasound pulses, such as transcranial imaging or transcranial ultrasound stimulation, a table summarizes the maximum pressure transmission for each ultrasound beam diameter and each frequency.


Assuntos
Encéfalo , Crânio , Humanos , Crânio/diagnóstico por imagem , Ultrassonografia , Acústica , Cabeça
11.
PLoS One ; 17(12): e0278865, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36512563

RESUMO

Low-intensity transcranial focused ultrasound (tFUS) offers new functional neuromodulation opportunities, enabling stimulation of cortical as well as deep brain areas with high spatial resolution. Brain stimulation of awake sheep, in the absence of the confounding effects of anesthesia on brain function, provides translational insight into potential human applications with safety information supplemented by histological analyses. We examined the effects of tFUS pulsing parameters, particularly regarding pulse durations (PDs), on stimulating the cortical motor area (M1) and its thalamic projection in unanesthetized, awake sheep (n = 8). A wearable tFUS headgear, custom-made for individual sheep, enabled experiments to be conducted without using anesthesia. FUS stimuli, each 200 ms long, were delivered to the M1 and the thalamus using three different PDs (0.5, 1, and 2 ms) with the pulse repetition frequency (PRF) adjusted to maintain a 70% duty cycle at a derated in situ spatial-peak temporal-average intensity (Ispta) of 3.6 W/cm2. Efferent electromyography (EMG) responses to stimulation were quantified from both hind limbs. Group-averaged EMG responses from each of the hind limbs across the experimental conditions revealed selective responses from the hind limb contralateral to sonication. The use of 0.5 and 1 ms PDs generated higher EMG signal amplitudes compared to those obtained using a 2 ms PD. Faster efferent response was also observed from thalamic stimulation than that from stimulating the M1. Post-sonication behavioral observation and histological assessment performed 24 h and 1 month after sonication were not indicative of any abnormalities. The results suggest the presence of pulsing scheme-dependent effects of tFUS on brain stimulation and attest its safety in awake large animals.


Assuntos
Córtex Motor , Vigília , Humanos , Animais , Ovinos , Mapeamento Encefálico/métodos , Encéfalo/fisiologia , Córtex Motor/fisiologia , Frequência Cardíaca
12.
Pharmaceutics ; 14(10)2022 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-36297554

RESUMO

Low-intensity transcranial focused ultrasound (FUS) has gained momentum as a non-/minimally-invasive modality that facilitates the delivery of various pharmaceutical agents to the brain. With the additional ability to modulate regional brain tissue excitability, FUS is anticipated to confer potential neurotherapeutic applications whereby a deeper insight of its safety is warranted. We investigated the effects of FUS applied to the rat brain (Sprague-Dawley) shortly after an intracortical injection of fluorescent interstitial solutes, a widely used convection-enhanced delivery technique that directly (i.e., bypassing the blood-brain-barrier (BBB)) introduces drugs or interstitial tracers to the brain parenchyma. Texas Red ovalbumin (OA) and fluorescein isothiocyanate-dextran (FITC-d) were used as the interstitial tracers. Rats that did not receive sonication showed an expected interstitial distribution of OA and FITC-d around the injection site, with a wider volume distribution of OA (21.8 ± 4.0 µL) compared to that of FITC-d (7.8 ± 2.7 µL). Remarkably, nearly half of the rats exposed to the FUS developed intracerebral hemorrhaging (ICH), with a significantly higher volume of bleeding compared to a minor red blood cell extravasation from the animals that were not exposed to sonication. This finding suggests that the local cerebrovascular injury inflicted by the micro-injection was further exacerbated by the application of sonication, particularly during the acute stage of injury. Smaller tracer volume distributions and weaker fluorescent intensities, compared to the unsonicated animals, were observed for the sonicated rats that did not manifest hemorrhaging, which may indicate an enhanced degree of clearance of the injected tracers. Our results call for careful safety precautions when ultrasound sonication is desired among groups under elevated risks associated with a weakened or damaged vascular integrity.

13.
IEEE J Biomed Health Inform ; 26(11): 5653-5664, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35969551

RESUMO

Transcranial focused ultrasound (tFUS) has gained attention in the field of brain stimulation owing to its non-invasive neurotherapeutic potentials. However, complex interactions between acoustic waves and the cranium may introduce misalignment of the acoustic focus from a geometric target location, thus, necessitate on-site feedback of real-time navigational information of the transducer (spatial coordinates and angular orientation) for the operators to accurately place the acoustic focus to the desired brain area. In this study, we propose a deep-learning-based network model that can provide spatial navigational information of a single-element FUS transducer with respect to the targeted brain region. The training dataset were acquired through forward simulations that reflect the different tFUS transmissions for each skull structure using cranial computed tomography (CT) image data. The performance of the network was evaluated through three ex vivo calvaria. As a result show that the presented neural network-based method can an accurately navigate the FUS transducer with the conformity of ∼99.59% in placement of the transducer and ∼74.49% in the focal volume and an average difference of ∼0.96 mm in the focal point, all capable of real-time operation (∼10 ms).


Assuntos
Encéfalo , Transdutores , Humanos , Encéfalo/fisiologia , Crânio/diagnóstico por imagem , Acústica , Redes Neurais de Computação
14.
Sci Rep ; 12(1): 12940, 2022 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-35902724

RESUMO

Efficient transport of solutes in the cerebrospinal fluid (CSF) plays a critical role in their clearance from the brain. Convective bulk flow of solutes in the CSF in the perivascular space (PVS) is considered one of the important mechanisms behind solute movement in the brain, before their ultimate drainage to the systemic lymphatic system. Acoustic pressure waves can impose radiation force on a medium in its path, inducing localized and directional fluidic flow, known as acoustic streaming. We transcranially applied low-intensity focused ultrasound (FUS) to rats that received an intracisternal injection of fluorescent CSF tracers (dextran and ovalbumin, having two different molecular weights-Mw). The sonication pulsing parameter was determined on the set that propelled the aqueous solution of toluidine blue O dye into a porous media (melamine foam) at the highest level of infiltration. Fluorescence imaging of the brain showed that application of FUS increased the uptake of ovalbumin at the sonicated plane, particularly around the ventricles, whereas the uptake of high-Mw dextran was unaffected. Numerical simulation showed that the effects of sonication were non-thermal. Sonication did not alter the animals' behavior or disrupt the blood-brain barrier (BBB) while yielding normal brain histology. The results suggest that FUS may serve as a new non-invasive means to promote interstitial CSF solute transport in a region-specific manner without disrupting the BBB, providing potential for enhanced clearance of waste products from the brain.


Assuntos
Barreira Hematoencefálica , Dextranos , Animais , Barreira Hematoencefálica/diagnóstico por imagem , Encéfalo/diagnóstico por imagem , Ovalbumina , Ratos , Ratos Sprague-Dawley
15.
J Clin Med ; 11(13)2022 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-35807094

RESUMO

Transcranial focused ultrasound (tFUS) is a promising technique of non-invasive brain stimulation for modulating neuronal activity with high spatial specificity. The medial prefrontal cortex (mPFC) has been proposed as a potential target for neuromodulation to prove emotional and sleep qualities. We aim to set up an appropriate clinical protocol for investigating the effects of tFUS stimulation of the bilateral mPFC for modulating the function of the brain-wide network using different sonication parameters. Seven participants received 20 min of 250 kHz tFUS to the bilateral mPFC with excitatory (70% duty cycle with sonication interval at 5 s) or suppressive (5% duty cycle with no interval) sonication protocols, which were compared to a sham condition. By placing the cigar-shaped sonication focus on the falx between both mPFCs, it was possible to simultaneously stimulate the bilateral mPFCs. Brain activity was analyzed using continuous electroencephalographic (EEG) recording during, before, and after tFUS. We investigated whether tFUS stimulation under the different conditions could lead to distinctive changes in brain activity in local brain regions where tFUS was directly delivered, and also in adjacent or remote brain areas that were not directly stimulated. This kind of study setting suggests that dynamic changes in brain cortical responses can occur within short periods of time, and that the distribution of these responses may differ depending on local brain states and functional brain architecture at the time of tFUS administration, or perhaps, at least temporarily, beyond the stimulation time. If so, tFUS could be useful for temporarily modifying regional brain activity, modulating functional connectivity, or reorganizing brain functions associated with various neuropsychiatric diseases, such as insomnia and depression.

16.
J Pers Med ; 12(2)2022 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-35207738

RESUMO

Preclinical studies have suggested that low-intensity transcranial focused ultrasound (tFUS) may have therapeutic potential for Alzheimer's disease (AD) by opening the blood-brain barrier (BBB), reducing amyloid pathology, and improving cognition. This study investigated the effects of tFUS on BBB opening, regional cerebral metabolic rate of glucose (rCMRglu), and cognitive function in AD patients. Eight patients with AD received image-guided tFUS to the right hippocampus immediately after intravenous injection of microbubble ultrasound contrast agents. Patients completed magnetic resonance imaging (MRI), 18F-fluoro-2-deoxyglucose positron emission tomography (PET), and cognitive assessments before and after the sonication. No evidence of transient BBB opening was found on T1 dynamic contrast-enhanced MRI. However, immediate recall (p = 0.03) and recognition memory (p = 0.02) were significantly improved on the verbal learning test. PET image analysis demonstrated increased rCMRglu in the right hippocampus (p = 0.001). In addition, increases of hippocampal rCMRglu were correlated with improvement in recognition memory (Spearman's ρ = 0.77, p = 0.02). No adverse event was observed. Our results suggest that tFUS to the hippocampus of AD patients may improve rCMRglu of the target area and memory in the short term, even without BBB opening. Further larger sham-controlled trials with loger follow-up are warranted to evaluate the efficacy and safety of tFUS in patients with AD.

17.
Ultrasonography ; 41(2): 382-393, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34963257

RESUMO

PURPOSE: The purpose of this study was to evaluate if transcutaneous application of low-intensity ultrasound can locally enhance the effects of finasteride on hair growth in a murine model of androgenic alopecia (AA). METHODS: AA mice (injected twice per week with testosterone enanthate, n=11), under daily oral administration of finasteride, received 1-MHz ultrasound for 1 hour at the unilateral thigh area five times per week for 5 weeks. Non-thermal and non-cavitational ultrasound was delivered in a pulsed manner (55-ms pulse duration with a repetition frequency of 4 Hz). Skin temperature was measured during sonication, and the measurements were validated with numerical simulations of sonication-induced tissue temperature changes. Hair growth was assessed both photographically and histologically. RESULTS: We found more hair growth on the sonicated thigh area than on the unsonicated thigh, beginning from week 3 through the end of the experiment. Histological analyses showed that the number of hair follicles doubled in the skin sections that received sonication compared to the unsonicated zone, with thicker follicular diameter and skin. An over five-fold increase was also observed in the anagen/telogen ratio in the sonicated area, suggesting an enhanced anagen phase. Skin temperature was unaltered by the administered sonication. CONCLUSION: The findings of the present study suggest that pulsed application of ultrasound promotes hair growth, potentially by disrupting the binding of albumin to finasteride. This may suggest further applications to enhance the pharmacological effects of other relevant drugs exhibiting high plasma protein binding.

18.
iScience ; 24(11): 103372, 2021 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-34825139

RESUMO

To address current unmet needs in terms of scalability and material biocompatibility for future photocrosslinking-based additive manufacturing technologies, emergent platform designs are in inexorable demand. In particular, a shift from the present use of cell-damaging UV light sources in light-based three-dimensional (3D) bioprinting methods demands new platforms. We adopted an organic light-emitting diode (OLED) microdisplay as a digital visible light modulator to create a 3D printing platform modality that offers scalability and multi-material capability while forgoing the need for UV photocrosslinking. We formulate biocompatible inks that are visible light-crosslinkable with relatively quick photoinitiation rates. We demonstrated successful attachment and rapid growth of primary human dermal fibroblast-adult (HDF-a) cells on biological substrates fabricated using the OLED platform. This platform incites new possibilities by providing a simple-yet-effective means for low-cost, high-throughput, and multi-material 3D fabrication of functional structures made of polymers, ceramic composites, and biomaterials.

19.
Sci Rep ; 11(1): 19274, 2021 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-34588588

RESUMO

Transcranial application of pulsed low-intensity focused ultrasound (FUS) modulates the excitability of region-specific brain areas, and anesthetic confounders on brain activity warrant the evaluation of the technique in awake animals. We examined the neuromodulatory effects of FUS in unanesthetized sheep by developing a custom-fit headgear capable of reproducibly placing an acoustic focus on the unilateral motor cortex (M1) and corresponding thalamic area. The efferent responses to sonication, based on the acoustic parameters previously identified in anesthetized sheep, were measured using electromyography (EMG) from both hind limbs across three experimental conditions: on-target sonication, off-target sonication, and without sonication. Excitatory sonication yielded greater amplitude of EMG signals obtained from the hind limb contralateral to sonication than that from the ipsilateral limb. Spurious appearance of motion-related EMG signals limited the amount of analyzed data (~ 10% selection of acquired data) during excitatory sonication, and the averaged EMG response rates elicited by the M1 and thalamic stimulations were 7.5 ± 1.4% and 6.7 ± 1.5%, respectively. Suppressive sonication, while sheep walked on the treadmill, temporarily reduced the EMG amplitude from the limb contralateral to sonication. No significant change was found in the EMG amplitudes during the off-target sonication. Behavioral observation throughout the study and histological analysis showed no sign of brain tissue damage caused by the acoustic stimulation. Marginal response rates observed during excitatory sonication call for technical refinement to reduce motion artifacts during EMG acquisitions as well as acoustic aberration correction schemes to improve spatial accuracy of sonication. Yet, our results indicate that low-intensity FUS modulated the excitability of regional brain tissues reversibly and safely in awake sheep, supporting its potential in theragnostic applications.


Assuntos
Atividade Motora/efeitos da radiação , Córtex Motor/efeitos da radiação , Tálamo/efeitos da radiação , Terapia por Ultrassom/métodos , Animais , Eletromiografia , Feminino , Modelos Animais , Atividade Motora/fisiologia , Córtex Motor/fisiologia , Ovinos , Tálamo/fisiologia , Terapia por Ultrassom/efeitos adversos , Ondas Ultrassônicas/efeitos adversos , Vigília
20.
BMC Anesthesiol ; 21(1): 158, 2021 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-34020595

RESUMO

BACKGROUND: High-intensity ultrasound has been used to induce acoustic cavitation in the skin and subsequently enhances skin permeability to deliver hydrophobic topical medications including lidocaine. In contrast, instead of changing skin permeability, pulsed application of low-intensity focused ultrasound (FUS) has shown to non-invasively and temporarily disrupt drug-plasma protein binding, thus has potential to enhance the anesthetic effects of hydrophilic lidocaine hydrochloride through unbinding it from serum/interstitial α1-acid glycoprotein (AAG). METHODS: FUS, operating at fundamental frequency of 500 kHz, was applied pulse-mode (55-ms pulse duration, 4-Hz pulse repetition frequency) at a spatial-peak pulse-average intensity of 5 W/cm2. In vitro equilibrium dialysis was performed to measure the unbound concentration of lidocaine (lidocaine hydrochloride) from dialysis cassettes, one located at the sonication focus and the other outside the sonication path, all immersed in phosphate-buffered saline solution containing both lidocaine (10 µg/mL) and human AAG (5 mg/mL). In subsequent animal experiments (Sprague-Dawley rats, n = 10), somatosensory evoked potential (SSEP), elicited by electrical stimulations to the unilateral hind leg, was measured under three experimental conditions-applications of FUS to the unilateral thigh area at the site of administered topical lidocaine, FUS only, and lidocaine only. Skin temperature was measured before and after sonication. Passive cavitation detection was also performed during sonication to evaluate the presence of FUS-induced cavitation. RESULTS: Sonication increased the unbound lidocaine concentration (8.7 ± 3.3 %) from the dialysis cassette, compared to that measured outside the sonication path (P < 0.001). Application of FUS alone did not alter the SSEP while administration of lidocaine reduced its P23 component (i.e., a positive peak at 23 ms latency). The FUS combined with lidocaine resulted in a further reduction of the P23 component (in a range of 21.8 - 23.4 ms after the electrical stimulations; F(2,27) = 3.2 - 4.0, P < 0.05), indicative of the enhanced anesthetic effect of the lidocaine. Administration of FUS neither induced cavitation nor altered skin conductance or temperature, suggesting that skin permeability was unaffected. CONCLUSIONS: Unbinding lidocaine from the plasma proteins by exposure to non-thermal low-intensity ultrasound is attributed as the main mechanism behind the observation.


Assuntos
Anestésicos Locais/farmacologia , Tratamento por Ondas de Choque Extracorpóreas/métodos , Lidocaína/farmacologia , Pele/efeitos dos fármacos , Administração Tópica , Anestésicos Locais/administração & dosagem , Animais , Lidocaína/administração & dosagem , Modelos Animais , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA