Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
IEEE Trans Biomed Eng ; 71(6): 1770-1779, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38198257

RESUMEN

BACKGROUND: Nociceptive pain is required for healthy function, yet, neuropathic pain (disease or injury) can be severely debilitating. Though a wide-array of treatment options are available, they are often systemic and/or invasive. As a promising neuromodulation treatment, Focused ultrasound (FUS) is a noninvasive and highly spatially-targeted technique shown to stimulate neural activity, yet, effects on pain signaling are currently unknown. OBJECTIVE: Develop and validate a method for studying FUS nerve stimulation modulation of pain-evoked neural responses in vivo. METHODS: We developed a high-resolution functional ultrasound (fUS) method capable of mapping cortical responses in healthy and neuropathic pain mice in response to FUS neuromodulation treatment. RESULTS: FUS-evoked hemodynamic responses are correlated with the intensity of peripheral neuromodulation. We confirm functional connectivity is altered in neuropathic mice and demonstrate that FUS can modulate neuropathic pain-evoked hemodynamics. CONCLUSIONS: The findings presented herein provides evidence for an FUS-based nerve pain method and validates the fUS technique developed for monitoring pain-evoked hemodynamics. SIGNIFICANCE: We anticipate that the findings presented herein describe a noninvasive and flexible nerve modulation technique for pain mitigation, furthering evidence for clinical translation.


Asunto(s)
Neuralgia , Animales , Neuralgia/terapia , Neuralgia/diagnóstico por imagen , Neuralgia/fisiopatología , Ratones , Masculino , Ratones Endogámicos C57BL , Mapeo Encefálico/métodos , Terapia por Ultrasonido/métodos , Ultrasonografía/métodos
2.
Ultrasound Med Biol ; 50(3): 332-340, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38105118

RESUMEN

OBJECTIVE: The objective of this study was to investigate the effect of FUS on autonomic nervous system activity, including heart and respiratory rates, and to separate the thermal modulation from combined thermal and mechanical FUS effects. METHODS: The thalamus and hypothalamus of wild-type mice were sonicated with a continuous-wave, 2 MHz FUS transducer at pressures of 425 and 850 kPa for 60 seconds. Cardiac and respiratory rates were monitored as signs of autonomic nervous activity. FUS-induced changes in autonomic activity were compared to FUS targeted to a spatially-distant motor region and to laser-induced heating. RESULTS: FUS delivered to the primary target over the thalamus and hypothalamus at 850 kPa reversibly increased the respiratory rate by 6.5±3.2 breaths per minute and decreased the heart rate by 3.2±1.8 beats per minute. No significant changes occurred in this region at 425 kPa or when targeting the motor regions at 850 kPa. Laser heating with the same temperature rise profile produced by 850 kPa sonication resulted in cardiorespiratory modulation similar to that of FUS. CONCLUSIONS: FUS is capable of reversibly and non-invasively modulating cardiorespiratory activity in mice. Localized changes in temperature may constitute the main cause for this activity, though further investigation is warranted into the distinct and complementary mechanisms of mechanically- and thermally-induced FUS neuromodulation. Close monitoring of vital signs during FUS neuromodulation may be warranted to monitor systemic responses to stimulation.


Asunto(s)
Frecuencia Respiratoria , Tálamo , Ratones , Animales , Temperatura
3.
Ultrasound Med Biol ; 49(8): 1768-1779, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37202245

RESUMEN

OBJECTIVE: Ultrasound elasticity imaging techniques aim to provide a non-invasive characterization of tissue mechanical properties to detect pathological changes and monitor disease progression. Harmonic motion imaging (HMI) is an ultrasound-based elasticity imaging technique that utilizes an oscillatory acoustic radiation force to induce localized displacements and estimate relative tissue stiffness. Previous studies have applied a low amplitude modulation (AM) frequency of 25 or 50 Hz in HMI to assess the mechanical properties of different tissue types. In this study, we investigate the dependence of AM frequency in HMI and whether the frequency can be adjusted based on the size and mechanical properties of the underlying medium for enhanced image contrast and inclusion detection. METHODS: A tissue-mimicking phantom with embedded inclusions at different sizes and stiffnesses was imaged within a range of AM frequencies from 25 to 250 Hz at 25-Hz step size. DISCUSSION: The AM frequency at which the maximum contrast and CNR are achieved depends on the size and stiffness of the inclusions. A general trend shows that contrast and CNR peak at higher frequencies for smaller inclusions. In addition, for some inclusions with the same size but different stiffnesses, the optimized AM frequency increases with the stiffness of the inclusion. Nevertheless, there is a shift between the frequencies at which the contrast peaks and those with maximum CNR. Finally, in agreement with the phantom findings, imaging an ex-vivo human specimen with a 2.7-cm breast tumor at a range of AM frequencies showed that the highest contrast and CNR are achieved at the AM frequency of 50 Hz. CONCLUSION: These findings indicate that the AM frequency can be optimized in different applications of HMI, especially in the clinic, for improved detection and characterization of tumors with different geometries and mechanical properties.


Asunto(s)
Diagnóstico por Imagen de Elasticidad , Neoplasias , Humanos , Ultrasonografía , Elasticidad , Diagnóstico por Imagen de Elasticidad/métodos , Fantasmas de Imagen
4.
IEEE Trans Biomed Eng ; 70(4): 1150-1161, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36191094

RESUMEN

Harmonic motion imaging (HMI) is an ultrasound-based elasticity imaging technique that utilizes oscillatory acoustic radiation force to estimate the mechanical properties of tissues, as well as monitor high-intensity focused ultrasound (HIFU) treatment. Conventionally, in HMI, a focused ultrasound (FUS) transducer generates oscillatory tissue displacements, and an imaging transducer acquires channel data for displacement estimation, with each transducer being driven with a separate system. The fixed position of the FUS focal spot requires mechanical translation of the transducers, which can be a time-consuming and challenging procedure. In this study, we developed and characterized a new HMI system with a multi-element FUS transducer with the capability of electronic focal steering of ±5 mm and ±2 mm from the geometric focus in the axial and lateral directions, respectively. A pulse sequence was developed to drive both the FUS and imaging transducers using a single ultrasound data acquisition (DAQ) system. The setup was validated on a tissue-mimicking phantom with embedded inclusions. Integrating beam steering with the mechanical translation of the transducers resulted in a consistent high contrast-to-noise ratio (CNR) for the inclusions with Young's moduli of 22 and 44 kPa within a 5-kPa background while the data acquisition speed is increased by 4.5-5.2-fold compared to the case when only mechanical movements were applied. The feasibility of simultaneous generation of multiple foci and tracking the induced displacements is demonstrated in phantoms for applications where imaging or treatment of a larger region is needed. Moreover, preliminary feasibility is shown in a human subject with a breast tumor, where the mean HMI displacement within the tumor was about 4 times lower than that within perilesional tissues. The proposed HMI system facilitates data acquisition in terms of flexibility and speed and can be potentially used in the clinic for breast cancer imaging and treatment.


Asunto(s)
Diagnóstico por Imagen de Elasticidad , Humanos , Módulo de Elasticidad , Elasticidad , Electrónica , Movimiento
5.
Int J Mol Sci ; 23(24)2022 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-36555129

RESUMEN

The blood-brain barrier (BBB) controls brain homeostasis; it is formed by vascular endothelial cells that are physically connected by tight junctions (TJs). The BBB expresses efflux transporters such as P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP), which limit the passage of substrate molecules from blood circulation to the brain. Focused ultrasound (FUS) with microbubbles can create a local and reversible detachment of the TJs. However, very little is known about the effect of FUS on the expression of efflux transporters. We investigated the in vivo effects of moderate acoustic pressures on both P-gp and BCRP expression for up to two weeks after sonication. Magnetic resonance-guided FUS was applied in the striatum of 12 rats. P-gp and BCRP expression were determined by immunohistochemistry at 1, 3, 7, and 14 days postFUS. Our results indicate that FUS-induced BBB opening is capable of (i) decreasing P-gp expression up to 3 days after sonication in both the treated and in the contralateral brain regions and is capable of (ii) overexpressing BCRP up to 7 days after FUS in the sonicated regions only. Our findings may help improve FUS-aided drug delivery strategies by considering both the mechanical effect on the TJs and the regulation of P-gp and BCRP.


Asunto(s)
Barrera Hematoencefálica , Neoplasias , Ratas , Animales , Barrera Hematoencefálica/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Proyectos Piloto , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Células Endoteliales/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Encéfalo/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Microburbujas
6.
Artículo en Inglés | MEDLINE | ID: mdl-34713274

RESUMEN

Ultrasound attenuation through soft tissues can produce an acoustic radiation force (ARF) and heating. The ARF-induced displacements and temperature evaluations can reveal tissue properties and provide insights into focused ultrasound (FUS) bio-effects. In this study, we describe an interleaving pulse sequence tested in a tissue-mimicking phantom that alternates FUS and plane-wave imaging pulses at a 1 kHz frame rate. The FUS is amplitude modulated, enabling the simultaneous evaluation of tissue-mimicking phantom displacement using harmonic motion imaging (HMI) and temperature rise using thermal strain imaging (TSI). The parameters were varied with a spatial peak temporal average acoustic intensity (I spta ) ranging from 1.5 to 311 W.cm-2, mechanical index (MI) from 0.43 to 4.0, and total energy (E) from 0.24 to 83 J.cm-2. The HMI and TSI processing could estimate displacement and temperature independently for temperatures below 1.80°C and displacements up to ~117 µm (I spta <311 W.cm-2, MI<4.0, and E<83 J.cm-2) indicated by a steady-state tissue-mimicking phantom displacement throughout the sonication and a comparable temperature estimation with simulations in the absence of tissue-mimicking phantom motion. The TSI estimations presented a mean error of ±0.03°C versus thermocouple estimations with a mean error of ±0.24°C. The results presented herein indicate that HMI can operate at diagnostic-temperature levels (i.e., <1°C) even when exceeding diagnostic acoustic intensity levels (720 mW.cm-2 < I spta < 207 W.cm-2). In addition, the combined HMI and TSI can potentially be used for simultaneous evaluation of safety during tissue elasticity imaging as well as FUS mechanism involved in novel ultrasound applications such as ultrasound neuromodulation and tumor ablation.

7.
Sci Rep ; 11(1): 15043, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34294761

RESUMEN

An emerging approach with potential in improving the treatment of neurodegenerative diseases and brain tumors is the use of focused ultrasound (FUS) to bypass the blood-brain barrier (BBB) in a non-invasive and localized manner. A large body of pre-clinical work has paved the way for the gradual clinical implementation of FUS-induced BBB opening. Even though the safety profile of FUS treatments in rodents has been extensively studied, the histological and behavioral effects of clinically relevant BBB opening in large animals are relatively understudied. Here, we examine the histological and behavioral safety profile following localized BBB opening in non-human primates (NHPs), using a neuronavigation-guided clinical system prototype. We show that FUS treatment triggers a short-lived immune response within the targeted region without exacerbating the touch accuracy or reaction time in visual-motor cognitive tasks. Our experiments were designed using a multiple-case-study approach, in order to maximize the acquired data and support translation of the FUS system into human studies. Four NHPs underwent a single session of FUS-mediated BBB opening in the prefrontal cortex. Two NHPs were treated bilaterally at different pressures, sacrificed on day 2 and 18 post-FUS, respectively, and their brains were histologically processed. In separate experiments, two NHPs that were earlier trained in a behavioral task were exposed to FUS unilaterally, and their performance was tracked for at least 3 weeks after BBB opening. An increased microglia density around blood vessels was detected on day 2, but was resolved by day 18. We also detected signs of enhanced immature neuron presence within areas that underwent BBB opening, compared to regions with an intact BBB, confirming previous rodent studies. Logistic regression analysis showed that the NHP cognitive performance did not deteriorate following BBB opening. These preliminary results demonstrate that neuronavigation-guided FUS with a single-element transducer is a non-invasive method capable of reversibly opening the BBB, without substantial histological or behavioral impact in an animal model closely resembling humans. Future work should confirm the observations of this multiple-case-study work across animals, species and tasks.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/efectos de la radiación , Neuronavegación/métodos , Ondas Ultrasónicas , Animales , Conducta Animal , Transporte Biológico/efectos de la radiación , Biomarcadores , Barrera Hematoencefálica/diagnóstico por imagen , Cognición , Imagen por Resonancia Magnética , Microburbujas , Modelos Animales , Primates , Carácter Cuantitativo Heredable
8.
Ultrasound Med Biol ; 47(9): 2759-2767, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34176702

RESUMEN

Focused ultrasound (FUS) has been used to non-invasively elicit or inhibit motor neuronal activity in the mouse peripheral nervous system in vivo. However, less is known about whether FUS elicits immune system responses associated with peripheral sensory neuronal activity. In this study, we sought to determine that non-invasive ultrasound image-guided FUS can elicit the neurogenic axon reflex of peripheral nerves in the mouse sciatic nerve. The local vasodilation in the plantar view of the hind paw detected with a high-resolution laser Doppler imager indicated neurogenic flare responses after FUS stimulation. The effects of FUS were compared with control groups, where a distinct pattern of blood flow changes was observed only in FUS-elicited neurogenic flare responses. The findings indicate that image-guided FUS elicits local axon reflexes in vivo with a high degree of specificity and penetration depth.


Asunto(s)
Nervios Periféricos , Reflejo , Animales , Ratones , Neuronas , Nervios Periféricos/diagnóstico por imagen , Ultrasonografía , Vasodilatación
9.
Ultrasound Med Biol ; 47(4): 998-1013, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33455808

RESUMEN

Ultrasound can modulate activity in the central nervous system, including the induction of motor responses in rodents. Recent studies investigating ultrasound-induced motor movements have described mostly bilateral limb responses, but quantitative evaluations have failed to reveal lateralization or differences in response characteristics between separate limbs or how specific brain targets dictate distinct limb responses. This study uses high-resolution focused ultrasound (FUS) to elicit motor responses in anesthetized mice in vivo and four-limb electromyography (EMG) to evaluate the latency, duration and power of paired motor responses (n = 1768). The results indicate that FUS generates target-specific differences in electromyographic characteristics and that brain targets separated by as little as 1 mm can modulate the responses in individual limbs differentially. Exploiting these differences may provide a tool for quantifying the susceptibility of underlying neural volumes to FUS, understanding the functioning of the targeted neuroanatomy and aiding in mechanistic studies of this non-invasive neuromodulation technique.


Asunto(s)
Encéfalo/efectos de la radiación , Movimiento/efectos de la radiación , Ondas Ultrasónicas , Estimulación Acústica , Animales , Electromiografía , Miembro Anterior/fisiología , Miembro Posterior/fisiología , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/fisiología , Tiempo de Reacción
10.
Artículo en Inglés | MEDLINE | ID: mdl-32746182

RESUMEN

Passive acoustic mapping enables the spatiotemporal monitoring of cavitation with circulating microbubbles during focused ultrasound (FUS)-mediated blood-brain barrier opening. However, the computational load for processing large data sets of cavitation maps or more complex algorithms limit the visualization in real-time for treatment monitoring and adjustment. In this study, we implemented a graphical processing unit (GPU)-accelerated sparse matrix-based beamforming and time exposure acoustics in a neuronavigation-guided ultrasound system for real-time spatiotemporal monitoring of cavitation. The system performance was tested in silico through benchmarking, in vitro using nonhuman primate (NHP) and human skull specimens, and demonstrated in vivo in NHPs. We demonstrated the stability of the cavitation map for integration times longer than 62.5 [Formula: see text]. A compromise between real-time displaying and cavitation map quality obtained from beamformed RF data sets with a size of 2000 ×128 ×30 (axial [Formula: see text]) was achieved for an integration time of [Formula: see text], which required a computational time of 0.27 s (frame rate of 3.7 Hz) and could be displayed in real-time between pulses at PRF = 2 Hz. Our benchmarking tests show that the GPU sparse-matrix algorithm processed the RF data set at a computational rate of [Formula: see text]/pixel/sample, which enables adjusting the frame rate and the integration time as needed. The neuronavigation system with real-time implementation of cavitation mapping facilitated the localization of the cavitation activity and helped to identify distortions due to FUS phase aberration. The in vivo test of the method demonstrated the feasibility of GPU-accelerated sparse matrix computing in a close to a clinical condition, where focus distortions exemplify problems during treatment. These experimental conditions show the need for spatiotemporal monitoring of cavitation with real-time capability that enables the operator to correct or halt the sonication in case substantial aberrations are observed.


Asunto(s)
Acústica , Microburbujas , Algoritmos , Animales , Barrera Hematoencefálica/diagnóstico por imagen , Ultrasonografía
11.
Artículo en Inglés | MEDLINE | ID: mdl-32746236

RESUMEN

Focused ultrasound (FUS)-based viscoelastic imaging techniques using high frame rate (HFR) ultrasound to track tissue displacement can be used for mechanistic monitoring of FUS neuromodulation. However, a majority of techniques avoid imaging during the active push transmit (interleaved or postpush acquisitions) to mitigate ultrasound interference, which leads to missing temporal information of ultrasound effects when FUS is being applied. Furthermore, critical for clinical translation, use of both axial steering and real-time (<1 s) capabilities for optimizing acoustic parameters for tissue engagement are largely missing. In this study, we describe a method of noninterleaved, single Vantage imaging displacement within an active FUS push with simultaneous axial steering and real-time capabilities using a single ultrasound acquisition machine. Results show that the pulse sequence can track micron-sized displacements using frame rates determined by the calculated time-of-flight (TOF), without interleaving the FUS pulses and imaging acquisition. Decimation by 3-7 frames increases signal-to-noise ratio (SNR) by 15.09±7.03 dB. Benchmarking tests of CUDA-optimized code show increase in processing speed of 35- and 300-fold in comparison with MATLAB parallel processing GPU and CPU functions, respectively, and we can estimate displacement from steered push beams ±10 mm from the geometric focus. Preliminary validation of displacement imaging in humans shows that the same driving pressures led to variable nerve engagement, demonstrating important feedback to improve transducer coupling, FUS incident angle, and targeting. Regarding the use of our technique for neuromodulation, we found that FUS altered thermal perception of thermal pain by 0.9643 units of pain ratings in a single trial. Additionally, 5 [Formula: see text] of nerve displacement was shown in on-target versus off-target sonications. The initial feasibility in healthy volunteers warrants further study for potential clinical translation of FUS for pain suppression.


Asunto(s)
Nervio Mediano , Transductores , Humanos , Dolor , Sensación , Ultrasonografía
12.
Pharmaceutics ; 12(11)2020 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-33233374

RESUMEN

The blood-brain barrier is the primary obstacle to efficient intracerebral drug delivery. Focused ultrasound, in conjunction with microbubbles, is a targeted and non-invasive way to disrupt the blood-brain barrier. Many commercially available ultrasound contrast agents and agents specifically designed for therapeutic purposes have been investigated in ultrasound-mediated blood-brain barrier opening studies. The new generation of sono-sensitive agents, such as liquid-core droplets, can also potentially disrupt the blood-brain barrier after their ultrasound-induced vaporization. In this review, we describe the different compositions of agents used for ultrasound-mediated blood-brain barrier opening in recent studies, and we discuss the challenges of the past five years related to the optimal formulation of agents.

13.
Sci Rep ; 10(1): 15254, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32943648

RESUMEN

Non-invasive diagnosis of breast cancer is still challenging due to the low specificity of the imaging modalities that calls for unnecessary biopsies. The diagnostic accuracy can be improved by assessing the breast tissue mechanical properties associated with pathological changes. Harmonic motion imaging (HMI) is an elasticity imaging technique that uses acoustic radiation force to evaluate the localized mechanical properties of the underlying tissue. Herein, we studied the in vivo feasibility of a clinical HMI system to differentiate breast tumors based on their relative HMI displacements, in human subjects. We performed HMI scans in 10 female subjects with breast masses: five benign and five malignant masses. Results revealed that both benign and malignant masses were stiffer than the surrounding tissues. However, malignant tumors underwent lower mean HMI displacement (1.1 ± 0.5 µm) compared to benign tumors (3.6 ± 1.5 µm) and the adjacent non-cancerous tissue (6.4 ± 2.5 µm), which allowed to differentiate between tumor types. Additionally, the excised breast specimens of the same patients (n = 5) were imaged post-surgically, where there was an excellent agreement between the in vivo and ex vivo findings, confirmed with histology. Higher displacement contrast between cancerous and non-cancerous tissue was found ex vivo, potentially due to the lower nonlinearity in the elastic properties of ex vivo tissue. This preliminary study lays the foundation for the potential complementary application of HMI in clinical practice in conjunction with the B-mode to classify suspicious breast masses.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Mama/diagnóstico por imagen , Diagnóstico por Imagen de Elasticidad/métodos , Estudios de Factibilidad , Femenino , Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Humanos , Persona de Mediana Edad , Movimiento (Física) , Transductores
14.
Front Phys ; 82020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32509757

RESUMEN

Focused ultrasound (FUS) neuromodulation has shown that mechanical waves can interact with cell membranes and mechanosensitive ion channels, causing changes in neuronal activity. However, the thorough understanding of the mechanisms involved in these interactions are hindered by different experimental conditions for a variety of animal scales and models. While the lack of complete understanding of FUS neuromodulation mechanisms does not impede benefiting from the current known advantages and potential of this technique, a precise characterization of its mechanisms of action and their dependence on experimental setup (e.g., tuning acoustic parameters and characterizing safety ranges) has the potential to exponentially improve its efficacy as well as spatial and functional selectivity. This could potentially reach the cell type specificity typical of other, more invasive techniques e.g., opto- and chemogenetics or at least orientation-specific selectivity afforded by transcranial magnetic stimulation. Here, the mechanisms and their potential overlap are reviewed along with discussions on the potential insights into mechanisms that magnetic resonance imaging sequences along with a multimodal stimulation approach involving electrical, magnetic, chemical, light, and mechanical stimuli can provide.

15.
IEEE Trans Med Imaging ; 39(11): 3391-3402, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32406828

RESUMEN

Focused ultrasound (FUS) is an emerging technique for neuromodulation due to its noninvasive application and high depth penetration. Recent studies have reported success in modulation of brain circuits, peripheral nerves, ion channels, and organ structures. In particular, neuromodulation of peripheral nerves and the underlying mechanisms remain comparatively unexplored in vivo. Lack of methodologies for FUS targeting and monitoring impede further research in in vivo studies. Thus, we developed a method that non-invasively measures nerve engagement, via tissue displacement, during FUS neuromodulation of in vivo nerves using simultaneous high frame-rate ultrasound imaging. Using this system, we can validate, in real-time, FUS targeting of the nerve and characterize subsequent compound muscle action potentials (CMAPs) elicited from sciatic nerve activation in mice using 0.5 to 5 ms pulse durations and 22 - 28 MPa peak positive stimulus pressures at 4 MHz. Interestingly, successful motor excitation from FUS neuromodulation required a minimum interframe nerve displacement of 18 µm without any displacement incurred at the skin or muscle levels. Moreover, CMAPs detected in mice monotonically increased with interframe nerve displacements within the range of 18 to 300 µm . Thus, correlation between nerve displacement and motor activation constitutes strong evidence FUS neuromodulation is driven by a mechanical effect given that tissue deflection is a result of highly focused acoustic radiation force.


Asunto(s)
Nervios Periféricos , Terapia por Ultrasonido , Animales , Ratones , Nervios Periféricos/diagnóstico por imagen , Ultrasonografía
16.
J Neural Eng ; 17(2): 026026, 2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-31940596

RESUMEN

OBJECTIVE: Focused ultrasound (FUS) has recently been demonstrated capable of exciting motor neuronal activity. However, comprehensive understanding of elucidated excitatory and inhibitory effects is required to better assess FUS-mediated modulation. In this study, we demonstrate that image-guided FUS can selectively modulate motor neuron activity in the mouse sciatic nerve in vivo and attribute motor responses to thermal effects. APPROACH: FUS was applied on the sciatic nerve of anesthetized mice in vivo through the intact skin and muscle using ultrasound imaging for targeting. Both excitatory and inhibitory effects were recorded using electromyography (EMG) along with muscle response of the hind limb. The effects of FUS modulation versus heating by invasive alternative heating source (AHS) on electrically evoked EMG responses in the sciatic nerve in vivo were also investigated. The safety and reversibility of the technique were validated using histology and EMG recovery. MAIN RESULTS: The FUS was capable of eliciting motor neuronal activity comparable to electrical stimulation ES, and facilitating motor neuronal response on electrically evoked potentials with temperature elevation up to 11.5 °C ± 0.3 °C (PRF ⩽ 40 Hz). On the other hand, FUS-induced temperature elevations above 15.1 °C ± 1.6 °C (PRF ⩾ 100 Hz) resulted in the suppression of electrically-evoked motor neuronal activity along with a decrease in EMG latency and area under the curve (AUC), which was validated against the invasive AHS with temperature elevation of 18.1 °C ± 8.5 °C. Histological findings along with EMG responses after FUS modulation demonstrated a reversible or irreversible modulation. SIGNIFICANCE: The findings reported herein indicate that image-guided FUS (PRF ⩽ 100 Hz) induces safe and controllable modulation of involuntarily evoked motor neuron activity in vivo.


Asunto(s)
Neuronas , Nervio Ciático , Animales , Estimulación Eléctrica , Electromiografía , Miembro Posterior , Ratones
17.
Ultrasound Med Biol ; 46(1): 73-89, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31668690

RESUMEN

Focused ultrasound (FUS)-mediated blood-brain barrier (BBB) opening is currently being investigated in clinical trials. Here, we describe a portable clinical system with a therapeutic transducer suitable for humans, which eliminates the need for in-line magnetic resonance imaging (MRI) guidance. A neuronavigation-guided 0.25-MHz single-element FUS transducer was developed for non-invasive clinical BBB opening. Numerical simulations and experiments were performed to determine the characteristics of the FUS beam within a human skull. We also validated the feasibility of BBB opening obtained with this system in two non-human primates using U.S. Food and Drug Administration (FDA)-approved treatment parameters. Ultrasound propagation through a human skull fragment caused 44.4 ± 1% pressure attenuation at a normal incidence angle, while the focal size decreased by 3.3 ± 1.4% and 3.9 ± 1.8% along the lateral and axial dimension, respectively. Measured lateral and axial shifts were 0.5 ± 0.4 mm and 2.1 ± 1.1 mm, while simulated shifts were 0.1 ± 0.2 mm and 6.1 ± 2.4 mm, respectively. A 1.5-MHz passive cavitation detector transcranially detected cavitation signals of Definity microbubbles flowing through a vessel-mimicking phantom. T1-weighted MRI confirmed a 153 ± 5.5 mm3 BBB opening in two non-human primates at a mechanical index of 0.4, using Definity microbubbles at the FDA-approved dose for imaging applications, without edema or hemorrhage. In conclusion, we developed a portable system for non-invasive BBB opening in humans, which can be achieved at clinically relevant ultrasound exposures without the need for in-line MRI guidance. The proposed FUS system may accelerate the adoption of non-invasive FUS-mediated therapies due to its fast application, low cost and portability.


Asunto(s)
Barrera Hematoencefálica/diagnóstico por imagen , Neuronavegación/métodos , Transductores , Animales , Diseño de Equipo , Humanos , Neuronavegación/instrumentación , Primates , Ultrasonografía
18.
Artículo en Inglés | MEDLINE | ID: mdl-31514131

RESUMEN

Temperature measurements with thin thermocouples embedded in ultrasound fields are strongly subjected to a viscous heating artifact (VHA). The artifact contribution decays over time; therefore, it can be minimized at late temperature readings. However, previous studies have failed to demonstrate a rigorous method for determining the optimal time point at which the artifact contribution is negligible. In this study, we present an iterative processing method based on successive curve fittings using an artifact-independent model. The fitting starting point moves at each iteration until the maximum R2 indicates where the viscous heating is minimum. A solution of the bioheat transfer equation is used to account for blood perfusion, thus enabling in vivo measurements. Three T-type thermocouples with different diameters and sensitivities were assessed in an excised canine liver and in the mouse brain in vivo. We found that the artifact constitutes up to 81% ± 5% of wire thermocouple readings. The best-fit time varied in the liver samples ( n = 3 ) from 0 to 3.335 ± 0.979 s and in the mouse brain ( n = 5 ) from 0 to 0.498 ± 0.457 s at variable experimental conditions, which clearly demonstrates the need of the method for finding the appropriate starting time point of the fit. This study introduces a statistical method to determine the best time to fit a curve that can back-estimate temperature in tissues under ultrasound exposure using thermocouples. This method allows temperature evaluation in vivo and in vitro during a validation and safety assessment of a wide range of therapeutic and diagnostic ultrasound modalities.


Asunto(s)
Termometría/instrumentación , Termometría/métodos , Terapia por Ultrasonido/instrumentación , Terapia por Ultrasonido/métodos , Algoritmos , Animales , Artefactos , Encéfalo/efectos de la radiación , Perros , Diseño de Equipo , Calor , Hígado/efectos de la radiación , Ratones , Fantasmas de Imagen , Viscosidad
19.
Appl Phys Lett ; 114(20): 203702, 2019 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-31148844

RESUMEN

Measuring temperature during focused ultrasound (FUS) procedures is critical for characterization, calibration, and monitoring to ultimately ensure safety and efficacy. Despite the low cost and the high spatial and temporal resolutions of temperature measurements using thermocouples, the viscous heating (VH) artifact at the thermocouple-tissue interface requires reading corrections for correct thermometric analysis. In this study, a simulation pipeline is proposed to correct the VH artifact arising from temperature measurements using thermocouples in FUS fields. The numerical model consists of simulating a primary source of heating due to ultrasound absorption and a secondary source of heating from viscous forces generated by the thermocouple in the FUS field. Our numerical validation found that up to 90% of the measured temperature rise was due to VH effects. Experimental temperature measurements were performed using thermocouples embedded in fresh chicken breast samples. Temperature corrections were demonstrated for single high-intensity FUS pulses at 3.1 MHz and for multiple pulses (3.1 MHz, 100 Hz, and 500 Hz pulse repetition frequency). The VH accumulated during sonications and produced a temperature increase of 3.1 °C and 15.3 °C for the single and multiple pulse sequences, respectively. The methodology presented here enables the decoupling of the temperature increase generated by absorption and VH. Thus, more reliable temperature measurements can be extracted from thermocouple measurements by correcting for VH.

20.
J Control Release ; 303: 289-301, 2019 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-30953664

RESUMEN

The blood-brain barrier (BBB) prevents most drugs from gaining access to the brain parenchyma, which is a recognized impediment to the treatment of neurodegenerative disorders like Parkinson's disease (PD). Focused ultrasound (FUS), in conjunction with systemically administered microbubbles, opens the BBB locally, reversibly and non-invasively. Herein, we show that neither FUS applied over both the striatum and the ventral midbrain, without neurotrophic factors, nor intravenous administration of neurotrophic factors (either through protein or gene delivery) without FUS, ameliorates the damage to the nigrostriatal dopaminergic pathway in the sub-acute MPTP mouse model of early-stage PD. Conversely, the combination of FUS and intravenous neurotrophic (protein or gene) delivery attenuates the damage to the nigrostriatal dopaminergic pathway, by allowing the entry of these agents into the brain parenchyma. Our findings provide evidence that the application of FUS at the early stages of PD facilitates critical neurotrophic delivery that can curb the rapid progression of neurodegeneration while improving the neuronal function, seemingly opening new therapeutic avenues for the early treatment of diseases of the central nervous system.


Asunto(s)
Terapia Genética , Trastornos Parkinsonianos/terapia , Terapia por Ultrasonido , Animales , Encéfalo/metabolismo , Vectores Genéticos , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Masculino , Ratones Endogámicos C57BL , Microburbujas , Neurturina/administración & dosificación , Proteínas Recombinantes/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA