Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 222
Filtrar
2.
bioRxiv ; 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38559149

RESUMEN

Focused ultrasound (FUS) is an emerging noinvasive technique for neuromodulation in the central nervous system (CNS). To evaluate the effects of FUS-induced neuromodulation, many studies used behavioral changes, functional magnetic resonance imaging (fMRI) or electroencephalography (EEG). However, behavioral readouts are often not easily mapped to specific brain activity, EEG has low spatial resolution limited to the surface of the brain and fMRI requires a large importable scanner that limits additional readouts and manipulations. In this context, functional ultrasound imaging (fUSI) holds promise to directly monitor the effects of FUS neuromodulation with high spatiotemporal resolution in a large field of view, with a comparatively simple and flexible setup. fUSI uses ultrafast Power Doppler Imaging (PDI) to measure changes in cerebral blood volume, which correlates well with neuronal activity and local field potentials. We designed a setup that aligns a FUS transducer with a linear array to allow immediate subsequent monitoring of the hemodynamic response with fUSI during and after FUS neuromodulation. We established a positive correlation between FUS pressure and the size of the activated area, as well as changes in cerebral blood volume (CBV) and found that unilateral sonications produce bilateral hemodynamic changes with ipsilateral accentuation in mice. We further demonstrated the ability to perform fully noninvasive, transcranial FUS-fUSI in nonhuman primates for the first time by using a lower-frequency transducer configuration.

3.
bioRxiv ; 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38617295

RESUMEN

Focused ultrasound (FUS) stimulation is a promising neuromodulation technique with the merits of non-invasiveness, high spatial resolution, and deep penetration depth. However, simultaneous imaging of FUS-induced brain tissue displacement and the subsequent effect of FUS stimulation on brain hemodynamics has proven challenging thus far. In addition, earlier studies lack in situ confirmation of targeting except for the magnetic resonance imaging-guided FUS system-based studies. The purpose of this study is 1) to introduce a fully ultrasonic approach to in situ target, modulate neuronal activity, and monitor the resultant neuromodulation effect by respectively leveraging displacement imaging, FUS, and functional ultrasound (fUS) imaging, and 2) to investigate FUS-evoked cerebral blood volume (CBV) response and the relationship between CBV and displacement. We performed displacement imaging on craniotomized mice to confirm the in targeting for neuromodulation site. We recorded hemodynamic responses evoked by FUS and fUS revealed an ipsilateral CBV increase that peaks at 4 s post-FUS. We saw a stronger hemodynamic activation in the subcortical region than cortical, showing good agreement with the brain elasticity map that can also be obtained using a similar methodology. We observed dose-dependent CBV response with peak CBV, activated area, and correlation coefficient increasing with ultrasonic dose. Furthermore, by mapping displacement and hemodynamic activation, we found that displacement colocalizes and linearly correlates with CBV increase. The findings presented herein demonstrated that FUS evokes ipsilateral hemodynamic activation in cortical and subcortical depths and the evoked hemodynamic responses colocalized and correlate with FUS-induced displacement. We anticipate that our findings will help consolidate accurate targeting as well as an understanding of how FUS displaces brain tissue and affects cerebral hemodynamics.

4.
J Transl Med ; 22(1): 320, 2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38555449

RESUMEN

BACKGROUND: Diffuse midline glioma (DMG) is a pediatric tumor with dismal prognosis. Systemic strategies have been unsuccessful and radiotherapy (RT) remains the standard-of-care. A central impediment to treatment is the blood-brain barrier (BBB), which precludes drug delivery to the central nervous system (CNS). Focused ultrasound (FUS) with microbubbles can transiently and non-invasively disrupt the BBB to enhance drug delivery. This study aimed to determine the feasibility of brainstem FUS in combination with clinical doses of RT. We hypothesized that FUS-mediated BBB-opening (BBBO) is safe and feasible with 39 Gy RT. METHODS: To establish a safety timeline, we administered FUS to the brainstem of non-tumor bearing mice concurrent with or adjuvant to RT; our findings were validated in a syngeneic brainstem murine model of DMG receiving repeated sonication concurrent with RT. The brainstems of male B6 (Cg)-Tyrc-2J/J albino mice were intracranially injected with mouse DMG cells (PDGFB+, H3.3K27M, p53-/-). A clinical RT dose of 39 Gy in 13 fractions (39 Gy/13fx) was delivered using the Small Animal Radiation Research Platform (SARRP) or XRAD-320 irradiator. FUS was administered via a 0.5 MHz transducer, with BBBO and tumor volume monitored by magnetic resonance imaging (MRI). RESULTS: FUS-mediated BBBO did not affect cardiorespiratory rate, motor function, or tissue integrity in non-tumor bearing mice receiving RT. Tumor-bearing mice tolerated repeated brainstem BBBO concurrent with RT. 39 Gy/13fx offered local control, though disease progression occurred 3-4 weeks post-RT. CONCLUSION: Repeated FUS-mediated BBBO is safe and feasible concurrent with RT. In our syngeneic DMG murine model, progression occurs, serving as an ideal model for future combination testing with RT and FUS-mediated drug delivery.


Asunto(s)
Barrera Hematoencefálica , Glioma , Humanos , Ratas , Niño , Masculino , Ratones , Animales , Modelos Animales de Enfermedad , Ratas Sprague-Dawley , Tronco Encefálico , Sistemas de Liberación de Medicamentos/métodos , Imagen por Resonancia Magnética , Glioma/radioterapia , Microburbujas , Encéfalo
5.
Sci Rep ; 14(1): 1488, 2024 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-38233480

RESUMEN

Focused ultrasound (FUS) is a non-invasive and non-ionizing technique which deploys ultrasound waves to induce bio-effects. When paired with acoustically active particles such as microbubbles (MBs), it can open the blood brain barrier (BBB) to facilitate drug delivery otherwise inhibited due to the presence of BBB. One of the parameters that affects the FUS beam propagation is the beam incidence angle on the skull. Prior work by our group has shown that, as incidence angles deviate from 90°, FUS focal pressures attenuate and result in a smaller BBB opening volume. The incidence angles calculated in our prior studies were in 2D and used skull information from CT. The study presented herein develops methods to calculate incidence angle in 3D in non-human primate (NHP) skull fragments using harmonic ultrasound imaging without using ionizing radiation. Our results show that ultrasound harmonic imaging is capable of accurately depicting features such as sutures and eye-sockets of the skull. Furthermore, we were able to reproduce previously reported relationships between the incidence angle and FUS beam attenuation. We also show feasibility of performing ultrasound harmonic imaging in in-vivo non-human primates. The all-ultrasound method presented herein combined with our neuronavigation system stands to increase more widespread adoption of FUS and render it accessible by eliminating the need for CT cranial mapping.


Asunto(s)
Barrera Hematoencefálica , Cráneo , Animales , Incidencia , Barrera Hematoencefálica/diagnóstico por imagen , Ultrasonografía , Cráneo/diagnóstico por imagen , Primates , Microburbujas , Sistemas de Liberación de Medicamentos/métodos , Imagen por Resonancia Magnética , Encéfalo/diagnóstico por imagen
6.
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
7.
medRxiv ; 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38196636

RESUMEN

Background: Focused ultrasound (FUS) in combination with microbubbles has recently shown great promise in facilitating blood-brain barrier (BBB) opening for drug delivery and immunotherapy in Alzheimer's disease (AD). However, it is currently limited to systems integrated within the MRI suites or requiring post-surgical implants, thus restricting its widespread clinical adoption. In this pilot study, we investigate the clinical safety and feasibility of a portable, non-invasive neuronavigation-guided FUS (NgFUS) system with integrated real-time 2-D microbubble cavitation mapping. Methods: A phase 1 clinical study with mild to moderate AD patients (N=6) underwent a single session of microbubble-mediated NgFUS to induce transient BBB opening (BBBO). Microbubble activity under FUS was monitored with real-time 2-D cavitation maps and dosing to ensure the efficacy and safety of the NgFUS treatment. Post-operative MRI was used for BBB opening and closure confirmation as well as safety assessment. Changes in AD biomarker levels in both blood serum and extracellular vesicles (EVs) were evaluated, while changes in amyloid-beta (Aß) load in the brain were assessed through 18F-Florbetapir PET. Results: BBBO was achieved in 5 out of 6 subjects with an average volume of 983±626 mm3 following FUS at the right frontal lobe both in white and gray matter regions. The outpatient treatment was completed within 34.8±10.7 min. Cavitation dose significantly correlated with the BBBO volume (R2>0.9, N=4), demonstrating the portable NgFUS system's capability of predicting opening volumes. The cavitation maps co-localized closely with the BBBO location, representing the first report of real-time transcranial 2-D cavitation mapping in the human brain. Larger opening volumes correlated with increased levels of AD biomarkers, including Aß42 (R2=0.74), Tau (R2=0.95), and P-Tau181 (R2=0.86), assayed in serum-derived EVs sampled 3 days after FUS (N=5). From PET scans, subjects showed a lower Aß load increase in the treated frontal lobe region compared to the contralateral region. Reduction in asymmetry standardized uptake value ratios (SUVR) correlated with the cavitation dose (R2>0.9, N=3). Clinical changes in the mini-mental state examination over 6 months were within the expected range of cognitive decline with no additional changes observed as a result of FUS. Conclusion: We showed the safety and feasibility of this cost-effective and time-efficient portable NgFUS treatment for BBBO in AD patients with the first demonstration of real-time 2-D cavitation mapping. The cavitation dose correlated with BBBO volume, a slowed increase in pathology, and serum detection of AD proteins. Our study highlights the potential for accessible FUS treatment in AD, with or without drug delivery.

8.
IEEE Trans Biomed Eng ; 71(2): 607-620, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37647191

RESUMEN

OBJECTIVE: To assess viscoelasticity, a pathologically relevant biomarker, shear wave elastography (SWE) generally uses phase velocity (PV) dispersion relationship generated via pulsed acoustic radiation force (ARF) excitation pulse. In this study, a multi-frequency oscillation (MFO)- excitation pulse with higher weight to higher frequencies is proposed to generate PV images via the generation of motion with energy concentrated at the target frequencies in contrast to the broadband frequency motion generated in pulsed SWE (PSWE). METHODS: The feasibility of MFO-SWE to generate PV images at 100 to 1000 Hz in steps of 100 Hz was investigated by imaging 6 and 70 kPa inclusions with 6.5 and 10.4 mm diameter and ex vivo bovine liver with and without the presence of an aberration layer and chicken muscle ex vivo, and 4T1 mouse breast tumor, in vivo with comparisons to PSWE. RESULTS: MFO-SWE-derived CNR was statistically higher than PSWE for 6 kPa (both with and without aberration) and 70 kPa (with aberration) inclusions and derived SNR of the liver was statistically higher than PSWE at higher frequency (600-1000 Hz). Quantitatively, at 600-1000 Hz, MFO-SWE improved CNR of inclusions (without and with) aberration on an average by (8.2 and 156)% and of the tumor by 122%, respectively, and improved SNR of the liver (without and with) aberration by (20.2 and 51.5)% and of chicken muscle by 72%, respectively compared to the PSWE. CONCLUSIONS AND SIGNIFICANCE: These results indicate the advantages of MFO-SWE to improve PV estimation at higher frequencies which could improve viscoelasticity quantification and feature delineation.


Asunto(s)
Diagnóstico por Imagen de Elasticidad , Animales , Bovinos , Ratones , Biomarcadores , Diagnóstico por Imagen de Elasticidad/métodos , Estudios de Factibilidad , Hígado/diagnóstico por imagen , Hígado/fisiología , Movimiento (Física) , Pollos
9.
Artículo en Inglés | MEDLINE | ID: mdl-37917522

RESUMEN

Harmonic motion imaging (HMI) is an ultrasound elastography technique that estimates the viscoelastic properties of tissues by inducing localized oscillatory motion using focused ultrasound (FUS). The resulting displacement, assumed to be inversely proportional to the tissue local stiffness, is estimated using an imaging array based on RF speckle tracking. In conventional HMI, this is accomplished with plane-wave (PW) imaging, which inherently suffers from low lateral resolution. Coherent PW compounding (PWC) leverages spatial and temporal resolution using synthetic focusing in transmit. In this study, we introduced focused imaging with parallel tracking in HMI and compared parallel tracking of various transmit F-numbers (F/2.6, 3, 4, and 5) qualitatively and quantitatively with PW and PWC imaging at various compounded angle ranges (6°, 12°, and 18°). An in silico model of a 56-kPa spherical inclusion (diameter: 3.6 mm) embedded in a 5.3-kPa background and a 5.3-kPa elastic phantom with cylindrical inclusions (Young's moduli: 22-56 kPa, diameters: 2.0-8.6 mm) were imaged to assess different tracking beam sequences. Speckle biasing in displacement estimation associated with parallel tracking was also investigated and concluded to be negligible in HMI. Parallel tracking in receive (Rx) resulted in 2%-7% and 8%-12% increase compared to PW imaging ( ) in HMI contrast and contrast-to-noise ratio in silico and phantoms. Focused imaging with parallel tracking in Rx was concluded to be most robust among PW and PWC imaging for displacement estimation, and its preclinical feasibility was demonstrated in postsurgical human cancerous breast tissue specimens and in vivo murine models of breast cancer.


Asunto(s)
Neoplasias de la Mama , Diagnóstico por Imagen de Elasticidad , Animales , Humanos , Ratones , Femenino , Ultrasonografía/métodos , Módulo de Elasticidad , Diagnóstico por Imagen de Elasticidad/métodos , Fantasmas de Imagen , Movimiento (Física)
10.
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
11.
Ultrasound Med Biol ; 50(1): 91-98, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37838523

RESUMEN

OBJECTIVE: Plaque characterization is essential for stroke prevention. In the study reported herein, we describe a heterogeneous phantom manufacturing technique with varying plaque compositions of different stiffness using polyvinyl alcohol (PVA) to emulate stenotic arteries and evaluated the use of pulse wave imaging (PWI) to assess plaque stiffness by comparing derived pulse wave velocities, with the goal of assessing plaque vulnerability and identifying high-risk patients for stroke. METHODS: Five stenotic phantoms (50% stenosis) were fabricated by pouring PVA solutions into 3-D-printed molds. Two of the phantoms had heterogeneous plaque compositions of soft (E0 = 13 kPa) and intermediate (E0 = 40 kPa) materials and of stiff (E0 = 54 kPa) and intermediate materials. Ultrasound sequences were acquired as the arterial phantoms were connected to a pulsating pump, and PWI was performed on the ultrasound acquisition using normalized cross-correlation to track the pulse-induced phantom wall distension propagations. Pulse wave velocities were estimated by fitting a linear regression line between the arrival time of the peak acceleration of the wall distension waveform and the corresponding location. RESULTS: Arterial phantoms with heterogeneous plaque stiffness were successfully fabricated. Pulse wave velocities of 2.06, 2.21, 2.49, 2.67 and 3.31 m/s were found in the phantom experiments using PWI for homogeneous soft plaque, the heterogeneous soft and intermediate plaque, homogeneous intermediate plaque, the heterogeneous stiff and intermediate plaque and homogeneous stiff plaque, respectively. CONCLUSION: A novel arterial phantom building technique was reported with varying heterogenous plaque compositions of different stiffness. The feasibility of using PWI to evaluate plaque stiffness in stenotic arteries was determined and found that PWI can distinguish between plaques of distinct stiffness and composition.


Asunto(s)
Placa Aterosclerótica , Accidente Cerebrovascular , Humanos , Alcohol Polivinílico , Constricción Patológica , Análisis de la Onda del Pulso/métodos , Fantasmas de Imagen , Placa Aterosclerótica/diagnóstico por imagen
12.
Nat Biomed Eng ; 2023 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-37857722

RESUMEN

The opening of the blood-brain barrier (BBB) by focused ultrasound (FUS) coupled with intravenously injected microbubbles can be leveraged as a form of immunotherapy for the treatment of neurodegenerative disorders. However, how FUS BBB opening affects brain macrophages is not well understood. Here by using single-cell sequencing to characterize the distinct responses of microglia and central nervous system-associated macrophages (CAMs) to FUS-mediated BBB opening in mice, we show that the treatment remodels the immune landscape via the recruitment of CAMs and the proliferation of microglia and via population size increases in disease-associated microglia. Both microglia and CAMs showed early and late increases in population sizes, yet only the proliferation of microglia increased at both timepoints. The population of disease-associated microglia also increased, accompanied by the upregulation of genes associated with gliogenesis and phagocytosis, with the depletion of brain macrophages significantly decreasing the duration of BBB opening.

13.
Proc Natl Acad Sci U S A ; 120(34): e2302910120, 2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37579143

RESUMEN

Gene editing in the brain has been challenging because of the restricted transport imposed by the blood-brain barrier (BBB). Current approaches mainly rely on local injection to bypass the BBB. However, such administration is highly invasive and not amenable to treating certain delicate regions of the brain. We demonstrate a safe and effective gene editing technique by using focused ultrasound (FUS) to transiently open the BBB for the transport of intravenously delivered CRISPR/Cas9 machinery to the brain.


Asunto(s)
Encéfalo , Edición Génica , Encéfalo/diagnóstico por imagen , Barrera Hematoencefálica , Transporte Biológico , Microburbujas
14.
Theranostics ; 13(12): 4102-4120, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37554284

RESUMEN

Rationale: Bilateral sonication with focused ultrasound (FUS) in conjunction with microbubbles has been shown to separately reduce amyloid plaques and hyperphosphorylated tau protein in the hippocampal formation and the entorhinal cortex in different mouse models of Alzheimer's disease (AD) without any therapeutic agents. However, the two pathologies are expressed concurrently in human disease. Therefore, the objective of this study is to investigate the effects of repeated bilateral sonications in the presence of both pathologies. Methods: Herein, we investigate its functional and morphological outcomes on brains bearing both pathologies simultaneously. Eleven transgenic mice of the 3xTg-AD line (14 months old) expressing human amyloid beta and human tau and eleven age-matched wild-type littermates received four weekly bilateral sonications covering the hippocampus followed by working memory testing. Afterwards, immunohistochemistry and immunoassays (western blot and ELISA) were employed to assess any changes in amyloid beta and human tau. Furthermore, we present preliminary data from our clinical trial using a neuronavigation-guided FUS system for sonications in AD patients (NCT04118764). Results: Interestingly, both wild-type and transgenic animals that received FUS experienced improved working memory and spent significantly more time in the escape platform-quadrant, with wild-type animals spending 43.2% (sham: 37.7%) and transgenic animals spending 35.3% (sham: 31.0%) of the trial in the target quadrant. Furthermore, this behavioral amelioration in the transgenic animals correlated with a 58.3% decrease in the neuronal length affected by tau and a 27.2% reduction in total tau levels. Amyloid plaque population, volume and overall load were also reduced overall. Consistently, preliminary data from a clinical trial involving AD patients showed a 1.8% decrease of amyloid PET signal 3-weeks after treatment in the treated hemisphere compared to baseline. Conclusion: For the first time, it is shown that bilateral FUS-induced BBB opening significantly and simultaneously ameliorates both coexistent pathologies, which translated to improvements in spatial memory of transgenic animals with complex AD, the human mimicking phenotype. The level of cognitive improvement was significantly correlated with the volume of BBB opening. Non-transgenic animals were also shown to exhibit similar memory amelioration for the first time, indicating that BBB opening results into benefits in the neuronal function regardless of the existence of AD pathology. A potential mechanism of action for the reduction of the both pathologies investigated was the cholesterol metabolism, specifically the LRP1b receptor, which exhibited increased expression levels in transgenic mice following FUS-induced BBB opening. Initial clinical evidence supported that the beta amyloid reduction shown in rodents could be translatable to humans with significant amyloid reduction shown in the treated hemisphere.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Ratones , Animales , Recién Nacido , Lactante , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Memoria Espacial , Encéfalo/metabolismo , Ratones Transgénicos , Modelos Animales de Enfermedad
15.
Brain Stimul ; 16(4): 1196-1204, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37558125

RESUMEN

BACKGROUND: Focused ultrasound (FUS) is a non-invasive neuromodulation technology that is being investigated for potential treatment of neurological and psychiatric disorders. FUS combined with microbubbles can temporarily open the intact blood-brain barrier (BBB) of animals and humans, and facilitate drug delivery. FUS exposure, either with or without microbubbles, has been demonstrated to alter the behavior of non-human primates (NHP), and previous studies have demonstrated the transient and long-term effects of FUS neuromodulation on functional connectivity using resting state functional MRI. The behavioral effects of FUS vary depending on whether or not it is applied in conjunction with microbubbles to open the BBB, but it is unknown whether opening the BBB affects functional connectivity differently than FUS alone. OBJECTIVE: To compare the effects of applying FUS alone (FUS neuromodulation) and FUS with microbubbles (FUS-BBB opening) on changes of resting state functional connectivity in NHP. METHODS: We applied 2 min FUS exposure without (neuromodulation) and with microbubbles (BBB opening) in the dorsal striatum of lightly anesthetized non-human primates, and acquired resting state functional MRI 40 min respectively after FUS exposure. The functional connectivity (FC) in the cortex and major brain networks between the two approaches were measured and compared. RESULTS: When applying FUS exposure to the caudate nucleus of NHP, we found that both FUS neuromodulation can activate FC between caudate and insular cortex, while inhibiting the FC between caudate and motor cortex. FUS-BBB opening can activate FC between the caudate and medial prefrontal cortex, and within the frontotemporal network (FTN). We also found both FUS and FUS-BBB opening can significantly activate FC within the default mode network (DMN). CONCLUSION: The results suggest applying FUS to a deep brain structure can alter functional connectivity in the DMN and FTN, and that FUS neuromodulation and FUS-mediated BBB opening can have different effects on patterns of functional connectivity.


Asunto(s)
Barrera Hematoencefálica , Encéfalo , Animales , Humanos , Encéfalo/diagnóstico por imagen , Encéfalo/fisiología , Primates , Corteza Cerebral/diagnóstico por imagen , Microburbujas , Imagen por Resonancia Magnética
16.
Front Neurosci ; 17: 1229683, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37575309

RESUMEN

The amyloid-ß (Aß) hypothesis implicates Aß protein accumulation in Alzheimer's disease (AD) onset and progression. However, therapies targeting Aß have proven insufficient in achieving disease reversal, prompting a shift to focus on early intervention and alternative therapeutic targets. Focused ultrasound (FUS) paired with systemically-introduced microbubbles (µB) is a non-invasive technique for targeted and transient blood-brain barrier opening (BBBO), which has demonstrated Aß and tau reduction, as well as memory improvement in models of late-stage AD. However, similar to drug treatments for AD, this approach is not sufficient for complete reversal of advanced, symptomatic AD. Here we aim to determine whether early intervention with FUS-BBBO in asymptomatic AD could delay disease onset. Thus, the objective of this study is to measure the protective effects of FUS-BBBO on anxiety, memory and AD-associated protein levels in female and male triple transgenic (3xTg) AD mice treated at an early age and disease state. Here we show that early, repeated intervention with FUS-BBBO decreased anxiety-associated behaviors in the open field test by 463.02 and 37.42% in male and female cohorts, respectively. FUS-BBBO preserved female aptitude for learning in the active place avoidance paradigm, reducing the shock quadrant time by 30.03 and 31.01% in the final long-term and reversal learning trials, respectively. Finally, FUS-BBBO reduced hippocampal accumulation of Aß40, Aß42, and total tau in females by 12.54, 13.05, and 3.57%, respectively, and reduced total tau in males by 18.98%. This demonstration of both cognitive and pathological protection could offer a solution for carriers of AD-associated mutations as a safe, non-invasive technique to delay the onset of the cognitive and pathological effects of AD.

17.
Res Sq ; 2023 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-37397998

RESUMEN

Focused ultrasound (FUS) is a non-invasive and non-ionizing technique which deploys ultrasound waves to induce bio-effects. When paired with acoustically active particles such as microbubbles (MBs), it can open the blood brain barrier (BBB) to facilitate drug delivery inhibited due to the presence of BBB. One of the parameters that affects the FUS beam propagation is the beam incidence angle on the skull. Prior work by our group has shown that, as incidence angles deviate from 90°, FUS focal pressures attenuate and result to a smaller BBB opening volume. The incidence angles calculated in our prior studies were in 2D and used skull information from CT. The study presented herein develops methods to calculate incidence angle in 3D in non-human primate (NHP) skull fragments using harmonic ultrasound imaging without using ionizing radiation. Our results show that ultrasound harmonic imaging is capable of accurately depicting features such as sutures and eye-sockets of the skull. Furthermore, we were able to reproduce previously reported relationships between the incidence angle and FUS beam attenuation. We also show feasibility of performing ultrasound harmonic imaging in in-vivo non-human primates. The all-ultrasound method presented herein combined with our neuronavigation system stands to increase more widespread adoption of FUS and render it accessible by eliminating the need for CT cranial mapping.

18.
Comput Biol Med ; 163: 107084, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37302374

RESUMEN

BACKGROUND: Direct current cardioversion (DCCV) is an established treatment to acutely convert atrial fibrillation (AF) to normal sinus rhythm. Yet, more than 70% of patients revert to AF shortly thereafter. Electromechanical Cycle Length Mapping (ECLM) is a high framerate, spectral analysis technique shown to non-invasively characterize electromechanical activation in paced canines and re-entrant flutter patients. This study assesses ECLM feasibility to map and quantify atrial arrhythmic electromechanical activation rates and inform on 1-day and 1-month DCCV response. METHODS: Forty-five subjects (30 AF; 15 healthy sinus rhythm (SR) controls) underwent transthoracic ECLM in four standard apical 2D echocardiographic views. AF patients were imaged within 1 h pre- and post-DCCV. 3D-rendered atrial ECLM cycle length (CL) maps and spatial CL histograms were generated. CL dispersion and percentage of arrhythmic CLs≤333ms across the entire atrial myocardium were computed transmurally. ECLM results were subsequently used as indicators of DCCV success. RESULTS: ECLM successfully confirmed the electrical atrial activation rates in 100% of healthy subjects (R2=0.96). In AF, ECLM maps localized the irregular activation rates pre-DCCV and confirmed successful post-DCCV with immediate reduction or elimination. ECLM metrics successfully distinguished DCCV 1-day and 1-month responders from non-responders, while pre-DCCV ECLM values independently predicted AF recurrence within 1-month post-DCCV. CONCLUSIONS: ECLM can characterize electromechanical activation rates in AF, quantify their extent, and identify and predict short- and long-term AF recurrence. ELCM constitutes thus a noninvasive arrhythmia imaging modality that can aid clinicians in simultaneous AF severity quantification, prediction of AF DCCV response, and personalized treatment planning.


Asunto(s)
Fibrilación Atrial , Cardioversión Eléctrica , Animales , Perros , Cardioversión Eléctrica/métodos , Fibrilación Atrial/diagnóstico por imagen , Fibrilación Atrial/terapia , Atrios Cardíacos/diagnóstico por imagen , Ecocardiografía/métodos , Resultado del Tratamiento
19.
Phys Med Biol ; 68(15)2023 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-37348487

RESUMEN

Objective. Arterial wall stiffness can provide valuable information on the proper function of the cardiovascular system. Ultrasound elasticity imaging techniques have shown great promise as a low-cost and non-invasive tool to enable localized maps of arterial wall stiffness. Such techniques rely upon motion detection algorithms that provide arterial wall displacement estimation.Approach. In this study, we propose an unsupervised deep learning-based approach, originally proposed for image registration, in order to enable improved quality arterial wall displacement estimation at high temporal and spatial resolutions. The performance of the proposed network was assessed through phantom experiments, where various models were trained by using ultrasound RF signals, or B-mode images, as well as different loss functions.Main results. Using the mean square error (MSE) for the training process provided the highest signal-to-noise ratio when training on the B-modes images (30.36 ± 1.14 dB) and highest contrast-to-noise ratio when training on the RF signals (32.84 ± 1.89 dB). In addition, training the model on RF signals demonstrated the capability of providing accurate localized pulse wave velocity (PWV) maps, with a mean relative error (MREPWV) of 3.32 ± 1.80% and anR2 of 0.97 ± 0.03. Finally, the developed model was tested in human common carotid arteriesin vivo, providing accurate tracking of the distension pulse wave propagation, with an MREPWV= 3.86 ± 2.69% andR2 = 0.95 ± 0.03.Significance. In conclusion, a novel displacement estimation approach was presented, showing promise in improving vascular elasticity imaging techniques.


Asunto(s)
Aprendizaje Profundo , Diagnóstico por Imagen de Elasticidad , Humanos , Análisis de la Onda del Pulso/métodos , Ultrasonografía/métodos , Diagnóstico por Imagen de Elasticidad/métodos , Arterias Carótidas/diagnóstico por imagen , Algoritmos , Elasticidad , Fantasmas de Imagen
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA