Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 893, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38291050

ABSTRACT

Subanesthetic ketamine is increasingly used for the treatment of varied psychiatric conditions, both on- and off-label. While it is commonly classified as an N-methyl D-aspartate receptor (NMDAR) antagonist, our picture of ketamine's mechanistic underpinnings is incomplete. Recent clinical evidence has indicated, controversially, that a component of the efficacy of subanesthetic ketamine may be opioid dependent. Using pharmacological functional ultrasound imaging in rats, we found that blocking opioid receptors suppressed neurophysiologic changes evoked by ketamine, but not by a more selective NMDAR antagonist, in limbic regions implicated in the pathophysiology of depression and in reward processing. Importantly, this opioid-dependent response was strongly sex-dependent, as it was not evident in female subjects and was fully reversed by surgical removal of the male gonads. We observed similar sex-dependent effects of opioid blockade affecting ketamine-evoked postsynaptic density and behavioral sensitization, as well as in opioid blockade-induced changes in opioid receptor density. Together, these results underscore the potential for ketamine to induce its affective responses via opioid signaling, and indicate that this opioid dependence may be strongly influenced by subject sex. These factors should be more directly assessed in future clinical trials.


Subject(s)
Ketamine , Mental Disorders , Humans , Rats , Male , Female , Animals , Ketamine/pharmacology , Ketamine/therapeutic use , Analgesics, Opioid/pharmacology , Mental Disorders/drug therapy , Signal Transduction , Receptors, N-Methyl-D-Aspartate/metabolism
2.
Brain Stimul ; 16(6): 1743-1752, 2023.
Article in English | MEDLINE | ID: mdl-38052373

ABSTRACT

Transcranial ultrasound neuromodulation is a promising potential therapeutic tool for the noninvasive treatment of neuropsychiatric disorders. However, the expansive parameter space and difficulties in controlling for peripheral auditory effects make it challenging to identify ultrasound sequences and brain targets that may provide therapeutic efficacy. Careful preclinical investigations in clinically relevant behavioral models are critically needed to identify suitable brain targets and acoustic parameters. However, there is a lack of ultrasound devices allowing for multi-target experimental investigations in awake and unrestrained rodents. We developed a miniaturized 64-element ultrasound array that enables neurointerventional investigations with within-trial active control targets in freely behaving rats. We first characterized the acoustic field with measurements in free water and with transcranial propagation. We then confirmed in vivo that the array can target multiple brain regions via electronic steering, and verified that wearing the device does not cause significant impairments to animal motility. Finally, we demonstrated the performance of our system in a high-throughput neuromodulation experiment, where we found that ultrasound stimulation of the rat central medial thalamus, but not an active control target, promotes arousal and increases locomotor activity.


Subject(s)
Brain , Wakefulness , Rats , Animals , Ultrasonography , Brain/diagnostic imaging , Brain/physiology , Arousal
3.
bioRxiv ; 2023 Oct 25.
Article in English | MEDLINE | ID: mdl-37961368

ABSTRACT

Ultrasound-activatable drug-loaded nanocarriers enable noninvasive and spatiotemporally-precise on-demand drug delivery throughout the body. However, most systems for ultrasonic drug uncaging utilize cavitation or heating as the drug release mechanism and often incorporate relatively exotic excipients into the formulation that together limit the drug-loading potential, stability, and clinical translatability and applicability of these systems. Here we describe an alternate strategy for the design of such systems in which the acoustic impedance and osmolarity of the internal liquid phase of a drug-loaded particle is tuned to maximize ultrasound-induced drug release. No gas phase, cavitation, or medium heating is necessary for the drug release mechanism. Instead, a non-cavitation-based mechanical response to ultrasound mediates the drug release. Importantly, this strategy can be implemented with relatively common pharmaceutical excipients, as we demonstrate here by implementing this mechanism with the inclusion of a few percent sucrose into the internal buffer of a liposome. Further, the ultrasound protocols sufficient for in vivo drug uncaging with this system are achievable with current clinical therapeutic ultrasound systems and with intensities that are within FDA and society guidelines for safe transcranial ultrasound application. Finally, this current implementation of this mechanism should be versatile and effective for the loading and uncaging of any therapeutic that may be loaded into a liposome, as we demonstrate for four different drugs in vitro, and two in vivo. These acoustomechanically activatable liposomes formulated with common pharmaceutical excipients promise a system with high clinical translational potential for ultrasonic drug uncaging of myriad drugs of clinical interest.

4.
Biol Psychiatry ; 93(12): 1118-1126, 2023 06 15.
Article in English | MEDLINE | ID: mdl-36841701

ABSTRACT

BACKGROUND: (S)-ketamine is an NMDA receptor antagonist, but it also binds to and activates mu opioid receptors (MORs) and kappa opioid receptors in vitro. However, the extent to which these receptors contribute to (S)-ketamine's in vivo pharmacology is unknown. METHODS: We investigated the extent to which (S)-ketamine interacts with opioid receptors in rats by combining in vitro and in vivo pharmacological approaches, in vivo molecular and functional imaging, and behavioral procedures relevant to human abuse liability. RESULTS: We found that the preferential opioid receptor antagonist naltrexone decreased (S)-ketamine self-administration and (S)-ketamine-induced activation of the nucleus accumbens, a key brain reward region. A single reinforcing dose of (S)-ketamine occupied brain MORs in vivo, and repeated doses decreased MOR density and activity and decreased heroin reinforcement without producing changes in NMDA receptor or kappa opioid receptor density. CONCLUSIONS: These results suggest that (S)-ketamine's abuse liability in humans is mediated in part by brain MORs.


Subject(s)
Ketamine , Rats , Humans , Animals , Ketamine/pharmacology , Receptors, Opioid, mu/physiology , Receptors, N-Methyl-D-Aspartate , Heroin , Receptors, Opioid/metabolism , Receptors, Opioid, kappa/metabolism
5.
Brain Connect ; 13(1): 28-38, 2023 02.
Article in English | MEDLINE | ID: mdl-35678063

ABSTRACT

Objective: In recent years, transcranial magnetic resonance-guided focused ultrasound (tcMRgFUS) has been established as a potential treatment option for movement disorders, including essential tremor (ET). So far, however, little is known about the impact of tcMRgFUS on structural connectivity. The objective of this study was to detect microstructural changes in tremor- and motor-related white matter tracts in ET patients treated with tcMRgFUS thalamotomy. Methods: Eleven patients diagnosed with ET were enrolled in this tcMRgFUS thalamotomy study. For each patient, 3 Tesla magnetic resonance imaging (3T MRI) including structural and diffusion MRI were acquired and the Clinical Rating Scale for Tremor was assessed before the procedure as well as 1 year after the treatment. Diffusion MRI tractography was performed to identify the cerebello-thalamo-cortical tract (CTCT), the medial lemniscus, and the corticospinal tract in both hemispheres on pre-treatment data. Pre-treatment tractography results were co-registered to post-treatment diffusion data. Diffusion tensor imaging (DTI) metrics, including fractional anisotropy (FA), mean diffusivity (MD) and radial diffusivity (RD), were averaged across the tracts in the pre- and post-treatment data. Results: The mean value of tract-specific DTI metrics changed significantly within the thalamic lesion and in the CTCT on the treated side (p < 0.05). Changes of DTI-derived indices within the CTCT correlated well with lesion overlap (FA: r = -0.54, p = 0.04; MD: r = 0.57, p = 0.04); RD: r = 0.67, p = 0.036). Further, a trend was seen for the correlation between changes of DTI-derived indices within the CTCT and clinical improvement (FA: r = 0.58; p = 0.062; MD: r = -0.52, p = 0.64; RD: r = -0.61 p = 0.090). Conclusions: Microstructural changes were detected within the CTCT after tcMRgFUS, and these changes correlated well with lesion-tract overlap. Our results show that diffusion MRI is able to detect the microstructural effects of tcMRgFUS, thereby further elucidating the treatment mechanism, and ultimately to improve targeting prospectively. Impact statement The results of this study demonstrate microstructural changes within the cerebello-thalamo-cortical pathways 1 year after MR-guided focused ultrasound thalamotomy. Even more, microstructural changes within the cerebello-thalamo-cortical pathways correlated significantly with clinical outcome. These findings do not only highly emphasize the need of new targeting strategies for MR-guided focused ultrasound thalamotomy but also help to elucidate the treatment mechanism of it.


Subject(s)
Essential Tremor , Magnetic Resonance Imaging , Humans , Magnetic Resonance Imaging/methods , Diffusion Tensor Imaging/methods , Tremor , Brain , Essential Tremor/diagnostic imaging , Essential Tremor/surgery , Magnetic Resonance Spectroscopy
6.
J Control Release ; 349: 434-442, 2022 09.
Article in English | MEDLINE | ID: mdl-35798095

ABSTRACT

Intrathecal drug delivery is routinely used in the treatment and prophylaxis of varied central nervous system conditions, as doing so allows drugs to directly bypass the blood-brain barrier. However, the utility of this route of administration is limited by poor brain and spinal cord parenchymal drug uptake from the cerebrospinal fluid. We demonstrate that a simple noninvasive transcranial ultrasound protocol can significantly increase influx of cerebrospinal fluid into the perivascular spaces of the brain, to enhance the uptake of intrathecally administered drugs. Specifically, we administered small (~1 kDa) and large (~155 kDa) molecule agents into the cisterna magna of rats and then applied low, diagnostic-intensity focused ultrasound in a scanning protocol throughout the brain. Using real-time magnetic resonance imaging and ex vivo histologic analyses, we observed significantly increased uptake of small molecule agents into the brain parenchyma, and of both small and large molecule agents into the perivascular space from the cerebrospinal fluid. Notably, there was no evidence of brain parenchymal damage following this intervention. The low intensity and noninvasive approach of transcranial ultrasound in this protocol underscores the ready path to clinical translation of this technique. In this manner, this protocol can be used to directly bypass the blood-brain barrier for whole-brain delivery of a variety of agents. Additionally, this technique can potentially be used as a means to probe the causal role of the glymphatic system in the variety of disease and physiologic processes to which it has been correlated.


Subject(s)
Glymphatic System , Ultrasonics , Animals , Blood-Brain Barrier , Brain/blood supply , Brain/diagnostic imaging , Drug Delivery Systems , Rats
7.
IEEE Trans Med Imaging ; 41(7): 1813-1825, 2022 07.
Article in English | MEDLINE | ID: mdl-35108201

ABSTRACT

Functional ultrasound (fUS) is a rapidly emerging modality that enables whole-brain imaging of neural activity in awake and mobile rodents. To achieve sufficient blood flow sensitivity in the brain microvasculature, fUS relies on long ultrasound data acquisitions at high frame rates, posing high demands on the sampling and processing hardware. Here we develop an image reconstruction method based on deep learning that significantly reduces the amount of data necessary while retaining imaging performance. We trained convolutional neural networks to learn the power Doppler reconstruction function from sparse sequences of ultrasound data with compression factors of up to 95%. High-quality images from in vivo acquisitions in rats were used for training and performance evaluation. We demonstrate that time series of power Doppler images can be reconstructed with sufficient accuracy to detect the small changes in cerebral blood volume (~10%) characteristic of task-evoked cortical activation, even though the network was not formally trained to reconstruct such image series. The proposed platform may facilitate the development of this neuroimaging modality in any setting where dedicated hardware is not available or in clinical scanners.


Subject(s)
Deep Learning , Animals , Brain/diagnostic imaging , Brain/physiology , Image Processing, Computer-Assisted/methods , Neural Networks, Computer , Rats , Ultrasonography
8.
Front Neurosci ; 14: 675, 2020.
Article in English | MEDLINE | ID: mdl-32760238

ABSTRACT

A long-standing goal of translational neuroscience is the ability to noninvasively deliver therapeutic agents to specific brain regions with high spatiotemporal resolution. Focused ultrasound (FUS) is an emerging technology that can noninvasively deliver energy up the order of 1 kW/cm2 with millimeter and millisecond resolution to any point in the human brain with Food and Drug Administration-approved hardware. Although FUS is clinically utilized primarily for focal ablation in conditions such as essential tremor, recent breakthroughs have enabled the use of FUS for drug delivery at lower intensities (i.e., tens of watts per square centimeter) without ablation of the tissue. In this review, we present strategies for image-guided FUS-mediated pharmacologic neurointerventions. First, we discuss blood-brain barrier opening to deliver therapeutic agents of a variety of sizes to the central nervous system. We then describe the use of ultrasound-sensitive nanoparticles to noninvasively deliver small molecules to millimeter-sized structures including superficial cortical regions and deep gray matter regions within the brain without the need for blood-brain barrier opening. We also consider the safety and potential complications of these techniques, with attention to temporal acuity. Finally, we close with a discussion of different methods for mapping the ultrasound field within the brain and describe future avenues of research in ultrasound-targeted drug therapies.

9.
Nanotheranostics ; 4(4): 210-223, 2020.
Article in English | MEDLINE | ID: mdl-32802731

ABSTRACT

Rationale: Localized blood-brain barrier (BBB) opening can be achieved with minimal to no tissue damage by applying pulsed focused ultrasound alongside a low microbubble (MB) dose. However, relatively little is known regarding how varying treatment parameters affect the degree of neuroinflammation following BBB opening. The goal of this study was to evaluate the activation of an inflammatory response following BBB opening as a function of applied acoustic pressure using two different microbubble doses. Methods: Mice were treated with 650 kHz ultrasound using varying acoustic peak negative pressures (PNPs) using two different MB doses, and activation of an inflammatory response, in terms of microglial and astrocyte activation, was assessed one hour following BBB opening using immunohistochemical staining. Harmonic and subharmonic acoustic emissions (AEs) were monitored for all treatments with a passive cavitation detector, and contrast-enhanced magnetic resonance imaging (CE-MRI) was performed following BBB opening to quantify the degree of opening. Hematoxylin and eosin-stained slides were assessed for the presence of microhemorrhage and edema. Results: For each MB dose, BBB opening was achieved with minimal activation of microglia and astrocytes using a PNP of 0.15 MPa. Higher PNPs were associated with increased activation, with greater increases associated with the use of the higher MB dose. Additionally, glial activation was still observed in the absence of histopathological findings. We found that CE-MRI was most strongly correlated with the degree of activation. While acoustic emissions were not predictive of microglial or astrocyte activation, subharmonic AEs were strongly associated with marked and severe histopathological findings. Conclusions: Our study demonstrated that there were mild histologic changes and activation of the acute inflammatory response using PNPs ranging from 0.15 MPa to 0.20 MPa, independent of MB dose. However, when higher PNPs of 0.25 MPa or above were applied, the same applied PNP resulted in more severe and widespread histological findings and activation of the acute inflammatory response when using the higher MB dose. The potential activation of the inflammatory response following ultrasound-mediated BBB opening should be considered when treating patients to maximize therapeutic benefit.


Subject(s)
Blood-Brain Barrier/radiation effects , Drug Delivery Systems/methods , Inflammation/metabolism , Microbubbles , Ultrasonic Therapy/methods , Animals , Astrocytes/metabolism , Brain Chemistry/radiation effects , Female , Mice , Microglia/metabolism , Ultrasonic Waves
10.
Radiology ; 294(1): 149-157, 2020 01.
Article in English | MEDLINE | ID: mdl-31714192

ABSTRACT

Background MRI performed with echo-planar imaging (EPI) sequences is sensitive to susceptibility artifacts in the presence of metallic objects, which presents a substantial barrier for performing functional MRI and diffusion tensor imaging (DTI) in patients with metallic orthodontic material and other head implants. Purpose To evaluate the ability to reduce susceptibility artifacts in healthy human participants wearing metallic orthodontic braces for two alternative approaches: T2-prepared functional MRI and diffusion-prepared DTI with three-dimensional fast gradient-echo readout. Materials and Methods In this prospective study conducted from February to September 2018, T2-prepared functional MRI and diffusion-prepared DTI were performed in healthy human participants. Removable dental braces with bonding trays were used so that MRI could be performed with braces and without braces in the same participants. Results were evaluated in regions with strong (EPI dropout regions for functional MRI and the inferior fronto-occipital fasciculus for DTI) and minimal (motor cortex for functional MRI and the posterior limb of internal capsule for DTI) susceptibility artifacts. Signal-to-noise ratio (SNR), contrast-to-noise ratio for functional MRI, apparent diffusion coefficient and fractional anisotropy for DTI, and degree of distortion (quantified with the Jaccard index, which measures the similarity of geometric shapes) were compared in regions with strong or minimal susceptibility effects between the current standard EPI sequences and the proposed alternatives by using paired t test. Results Six participants were evaluated (mean age ± standard deviation, 40 years ± 6; three women). In brain regions with strong susceptibility effects from the metallic braces, T2-prepared functional MRI showed significantly higher SNR (37.8 ± 2.4 vs 15.5 ± 5.3; P < .001) and contrast-to-noise ratio (0.83 ± 0.16 vs 0.29 ± 0.10; P < .001), whereas diffusion-prepared DTI showed higher SNR (5.8 ± 1.5 vs 3.8 ± 0.7; P = .03) than did conventional EPI methods. Apparent diffusion coefficient and fractional anisotropy were consistent with the literature. Geometric distortion was substantially reduced throughout the brain with the proposed methods (significantly higher Jaccard index, 0.95 ± 0.12 vs 0.81 ± 0.61; P < .001). Conclusion T2-prepared functional MRI and diffusion-prepared diffusion tensor imaging can acquire functional and diffusion MRI, respectively, in healthy human participants wearing metallic dental braces with less susceptibility artifacts and geometric distortion than with conventional echo-planar imaging. © RSNA, 2019 Online supplemental material is available for this article. See also the editorial by Dietrich in this issue.


Subject(s)
Artifacts , Brain/diagnostic imaging , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Orthodontic Brackets , Adult , Diffusion Tensor Imaging/methods , Female , Humans , Male , Prospective Studies , Reproducibility of Results
11.
Neuron ; 100(3): 728-738.e7, 2018 11 07.
Article in English | MEDLINE | ID: mdl-30408444

ABSTRACT

Being able to noninvasively modulate brain activity, where and when an experimenter desires, with an immediate path toward human translation is a long-standing goal for neuroscience. To enable robust perturbation of brain activity while leveraging the ability of focused ultrasound to deliver energy to any point of the brain noninvasively, we have developed biocompatible and clinically translatable nanoparticles that allow ultrasound-induced uncaging of neuromodulatory drugs. Utilizing the anesthetic propofol, together with electrophysiological and imaging assays, we show that the neuromodulatory effect of ultrasonic drug uncaging is limited spatially and temporally by the size of the ultrasound focus, the sonication timing, and the pharmacokinetics of the uncaged drug. Moreover, we see secondary effects in brain regions anatomically distinct from and functionally connected to the sonicated region, indicating that ultrasonic drug uncaging could noninvasively map the changes in functional network connectivity associated with pharmacologic action at a particular brain target.


Subject(s)
Anesthetics/administration & dosage , Brain Mapping/methods , Brain/physiology , Nanoparticles/administration & dosage , Nerve Net/physiology , Ultrasonic Waves , Anesthetics/metabolism , Animals , Brain/diagnostic imaging , Brain/drug effects , Magnetic Resonance Imaging/methods , Male , Nanoparticles/metabolism , Nerve Net/diagnostic imaging , Nerve Net/drug effects , Propofol/metabolism , Rats , Rats, Long-Evans
12.
Neuron ; 98(5): 875-877, 2018 06 06.
Article in English | MEDLINE | ID: mdl-29879389

ABSTRACT

Many neuroscientists are excited regarding the potential of ultrasound to yield spatiotemporally precise and noninvasive modulation of arbitrary brain regions. Here, Guo et al. (2018) and Sato et al. (2018) show that applying ultrasound to rodent brains activates acoustic responses more prominently than eliciting neuromodulation directly, suggesting potential confounds of ultrasound neuromodulation experiments.


Subject(s)
Brain , Cochlea
13.
Hum Brain Mapp ; 38(6): 3001-3010, 2017 06.
Article in English | MEDLINE | ID: mdl-28295805

ABSTRACT

OBJECTIVE: The aim of this study is to evaluate the correlation between resting state functional MRI (RS-fMRI) activity and motor and cognitive impairment in spinocerebellar ataxia type 6 (SCA6). METHODS: Twelve patients with genetically confirmed SCA6 and 14 age matched healthy controls were imaged with RS-fMRI. Whole brain gray matter was automatically parcellated into 1000 regions of interest (ROIs). For each ROI, the first eigenvariate of voxel time courses was extracted. For each patient, Pearson correlation coefficients between each pair of ROI time courses were calculated across the 1000 ROIs. The set of average control correlation coefficients were fed as an undirected weighted adjacency matrix into the Rubinov and Sporns (2010) modularity algorithm. The intranetwork global efficiency of the thresholded adjacency sub-matrix was calculated and correlated with ataxia scores and cognitive performance. RESULTS: SCA6 patients showed mild cognitive impairments in executive function and visual-motor processing compared to control subjects. These neuropsychological impairments were correlated with decreased RS functional connectivity (FC) in the attention network. CONCLUSIONS: Mild cognitive executive functions and visual-motor coordination impairments seen in SCA6 patients correlate with decreased resting-state connectivity in the attention network, suggesting a possible metric for the study of cognitive dysfunction in cerebellar disease. Hum Brain Mapp 38:3001-3010, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Brain Mapping , Brain/physiopathology , Cognition Disorders/etiology , Cognition Disorders/pathology , Neural Pathways/physiopathology , Spinocerebellar Ataxias/complications , Adult , Aged , Aged, 80 and over , Brain/diagnostic imaging , Female , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Middle Aged , Nerve Net/diagnostic imaging , Nerve Net/physiopathology , Neural Pathways/diagnostic imaging , Neuropsychological Tests , Oxygen/blood , Psychomotor Disorders/etiology , Rest , Statistics as Topic
14.
Nano Lett ; 17(2): 652-659, 2017 02 08.
Article in English | MEDLINE | ID: mdl-28094959

ABSTRACT

Targeted, noninvasive neuromodulation of the brain of an otherwise awake subject could revolutionize both basic and clinical neuroscience. Toward this goal, we have developed nanoparticles that allow noninvasive uncaging of a neuromodulatory drug, in this case the small molecule anesthetic propofol, upon the application of focused ultrasound. These nanoparticles are composed of biodegradable and biocompatible constituents and are activated using sonication parameters that are readily achievable by current clinical transcranial focused ultrasound systems. These particles are potent enough that their activation can silence seizures in an acute rat seizure model. Notably, there is no evidence of brain parenchymal damage or blood-brain barrier opening with their use. Further development of these particles promises noninvasive, focal, and image-guided clinical neuromodulation along a variety of pharmacological axes.


Subject(s)
Brain/drug effects , Emulsions/chemistry , Nanoparticles/chemistry , Neurotransmitter Agents/administration & dosage , Anesthetics/administration & dosage , Anesthetics/chemistry , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Drug Carriers , Drug Liberation , Fluorocarbons/chemistry , Magnetic Resonance Imaging , Neurotransmitter Agents/chemistry , Optical Imaging , Propofol/administration & dosage , Propofol/chemistry , Rats , Seizures/drug therapy , Tissue Distribution , Ultrasonic Waves
15.
Mol Imaging Biol ; 19(1): 24-30, 2017 02.
Article in English | MEDLINE | ID: mdl-27481359

ABSTRACT

PURPOSE: A wide variety of hydrophilic imaging and therapeutic agents are unable to gain access to the central nervous system (CNS) due to the blood-brain barrier (BBB). In particular, unless a particular transporter exists that may transport the agent across the BBB, most agents that are larger than 500 Da or that are hydrophilic will be excluded by the BBB. Glutamate carboxypeptidase II (GCPII), also known as the prostate-specific membrane antigen (PSMA) in the periphery, has been implicated in various neuropsychiatric conditions. As all agents that target GCPII are hydrophilic and thereby excluded from the CNS, we used GCPII as a platform for demonstrating our MR-guided focused ultrasound (MRgFUS) technique for delivery of GCPII/PSMA-specific imaging agents to the brain. PROCEDURES: Female rats underwent MRgFUS-mediated opening of the BBB. After opening of the BBB, either a radio- or fluorescently labeled ureido-based ligand for GCPII/PSMA was administered intravenously. Brain uptake was assessed for 2-(3-{1-carboxy-5-[(6-[18F]fluoro-pyridine-3-carbonyl)-amino]-pentyl}-ureido)-pentanedioic acid ([18F]DCFPyL) and YC-27, two compounds known to bind GCPII/PSMA with high affinity, using positron emission tomography (PET) and near-infrared fluorescence (NIRF) imaging, respectively. Specificity of ligand binding to GCPII/PSMA in the brain was determined with co-administration of a molar excess of ZJ-43, a compound of the same chemical class but different structure from either [18F]DCFPyL or YC-27, which competes for GCPII/PSMA binding. RESULTS: Dynamic PET imaging using [18F]DCFPyL demonstrated that target uptake reached a plateau by ∼1 h after radiotracer administration, with target/background ratios continuing to increase throughout the course of imaging, from a ratio of ∼4:1 at 45 min to ∼7:1 by 80 min. NIRF imaging likewise demonstrated delivery of YC-27 to the brain, with clear visualization of tracer in the brain at 24 h. Tissue uptake of both ligands was greatly diminished by ZJ-43 co-administration, establishing specificity of binding of each to GCPII/PSMA. On gross and histological examination, animals showed no evidence for hemorrhage or other deleterious consequences of MRgFUS. CONCLUSIONS: MRgFUS provided safe opening of the BBB to enable specific delivery of two hydrophilic agents to target tissues within the brain. This platform might facilitate imaging and therapy using a variety of agents that have heretofore been excluded from the CNS.


Subject(s)
Antigens, Surface/metabolism , Blood-Brain Barrier/metabolism , Glutamate Carboxypeptidase II/metabolism , Hydrophobic and Hydrophilic Interactions , Magnetic Resonance Imaging , Molecular Imaging/methods , Ultrasonography/methods , Animals , Autoradiography , Brain/diagnostic imaging , Female , Fluorescence , Glutamates , Lysine/analogs & derivatives , Positron Emission Tomography Computed Tomography , Rats, Inbred F344 , Urea/analogs & derivatives
16.
Hum Brain Mapp ; 37(5): 1986-97, 2016 May.
Article in English | MEDLINE | ID: mdl-27012314

ABSTRACT

Much recent attention has been paid to quantifying anatomic and functional neuroimaging on the individual subject level. For optimal individual subject characterization, specific acquisition and analysis features need to be identified that maximize interindividual variability while concomitantly minimizing intra-subject variability. We delineate the effect of various acquisition parameters (length of acquisition, sampling frequency) and analysis methods (time course extraction, region of interest parcellation, and thresholding of connectivity-derived network graphs) on characterizing individual subject differentiation. We utilize a non-parametric statistical metric that quantifies the degree to which a parameter set allows this individual subject differentiation by both maximizing interindividual variance and minimizing intra-individual variance. We apply this metric to analysis of four publicly available test-retest resting-state fMRI (rs-fMRI) data sets. We find that for the question of maximizing individual differentiation, (i) for increasing sampling, there is a relative tradeoff between increased sampling frequency and increased acquisition time; (ii) for the sizes of the interrogated data sets, only 3-4 min of acquisition time was sufficient to maximally differentiate each subject with an algorithm that utilized no a priori information regarding subject identification; and (iii) brain regions that most contribute to this individual subject characterization lie in the default mode, attention, and executive control networks. These findings may guide optimal rs-fMRI experiment design and may elucidate the neural bases for subject-to-subject differences. Hum Brain Mapp 37:1986-1997, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Brain Mapping , Brain/diagnostic imaging , Individuality , Magnetic Resonance Imaging , Algorithms , Humans , Image Processing, Computer-Assisted , Neural Pathways , Statistics, Nonparametric
17.
Hum Brain Mapp ; 37(3): 913-23, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26663615

ABSTRACT

PURPOSE: To compare language networks derived from resting-state fMRI (rs-fMRI) with task-fMRI in patients with brain tumors and investigate variables that affect rs-fMRI vs task-fMRI concordance. MATERIALS AND METHODS: Independent component analysis (ICA) of rs-fMRI was performed with 20, 30, 40, and 50 target components (ICA20 to ICA50) and language networks identified for patients presenting for presurgical fMRI mapping between 1/1/2009 and 7/1/2015. 49 patients were analyzed fulfilling criteria for presence of brain tumors, no prior brain surgery, and adequate task-fMRI performance. Rs-vs-task-fMRI concordance was measured using Dice coefficients across varying fMRI thresholds before and after noise removal. Multi-thresholded Dice coefficient volume under the surface (DiceVUS) and maximum Dice coefficient (MaxDice) were calculated. One-way Analysis of Variance (ANOVA) was performed to determine significance of DiceVUS and MaxDice between the four ICA order groups. Age, Sex, Handedness, Tumor Side, Tumor Size, WHO Grade, number of scrubbed volumes, image intensity root mean square (iRMS), and mean framewise displacement (FD) were used as predictors for VUS in a linear regression. RESULTS: Artificial elevation of rs-fMRI vs task-fMRI concordance is seen at low thresholds due to noise. Noise-removed group-mean DiceVUS and MaxDice improved as ICA order increased, however ANOVA demonstrated no statistically significant difference between the four groups. Linear regression demonstrated an association between iRMS and DiceVUS for ICA30-50, and iRMS and MaxDice for ICA50. CONCLUSION: Overall there is moderate group level rs-vs-task fMRI language network concordance, however substantial subject-level variability exists; iRMS may be used to determine reliability of rs-fMRI derived language networks.


Subject(s)
Brain Mapping/methods , Brain Neoplasms/physiopathology , Brain/physiopathology , Magnetic Resonance Imaging/methods , Adolescent , Adult , Aged , Analysis of Variance , Brain/surgery , Brain Neoplasms/surgery , Female , Humans , Language , Linear Models , Male , Mental Processes/physiology , Middle Aged , Neuropsychological Tests , Preoperative Care/methods , Rest , Software , Young Adult
18.
Cold Spring Harb Protoc ; 2015(8): 715-24, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26240415

ABSTRACT

Brain reward systems play a central role in the cognitive and hedonic behaviors of mammals. Multiple neuron types and brain regions are involved in reward processing, posing fascinating scientific questions, and major experimental challenges. Using diverse approaches including genetics, electrophysiology, imaging, and behavioral analysis, a large body of research has focused on both normal functioning of the reward circuitry and on its potential significance in neuropsychiatric diseases. In this introduction, we illustrate a real-world application of optogenetics to mammalian behavior and physiology, delineating procedures and technologies for optogenetic control of individual components of the reward circuitry. We describe the experimental setup and protocol for integrating optogenetic modulation of dopamine neurons with fast-scan cyclic voltammetry, conditioned place preference, and operant conditioning to assess the causal role of well-defined electrical and biochemical signals in reward-related behavior.


Subject(s)
Behavior, Animal , Brain/physiology , Dopaminergic Neurons/physiology , Optogenetics/methods , Reward , Animals , Behavior Observation Techniques , Electrophysiological Phenomena , Mammals , Optical Imaging
19.
Nat Commun ; 6: 6719, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25813863

ABSTRACT

Alterations in mucin expression and glycosylation are associated with cancer development. Underglycosylated mucin-1 (uMUC1) is overexpressed in most malignant adenocarcinomas of epithelial origin (for example, colon, breast and ovarian cancer). Its counterpart MUC1 is a large polymer rich in glycans containing multiple exchangeable OH protons, which is readily detectable by chemical exchange saturation transfer (CEST) MRI. We show here that deglycosylation of MUC1 results in >75% reduction in CEST signal. Three uMUC1(+) human malignant cancer cell lines overexpressing uMUC1 (BT20, HT29 and LS174T) show a significantly lower CEST signal compared with the benign human epithelial cell line MCF10A and the uMUC1(-) tumour cell line U87. Furthermore, we demonstrate that in vivo CEST MRI is able to make a distinction between LS174T and U87 tumour cells implanted in the mouse brain. These results suggest that the mucCEST MRI signal can be used as a label-free surrogate marker to non-invasively assess mucin glycosylation and tumour malignancy.


Subject(s)
Carcinoma/metabolism , Glycosylation , Magnetic Resonance Imaging/methods , Mucin-1/metabolism , Animals , Carcinoma/pathology , Cell Line, Tumor , HT29 Cells , Humans , Mice , Mice, SCID , Molecular Imaging/methods , Neoplasm Transplantation
20.
Cell ; 157(7): 1535-51, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24949967

ABSTRACT

Social interaction is a complex behavior essential for many species and is impaired in major neuropsychiatric disorders. Pharmacological studies have implicated certain neurotransmitter systems in social behavior, but circuit-level understanding of endogenous neural activity during social interaction is lacking. We therefore developed and applied a new methodology, termed fiber photometry, to optically record natural neural activity in genetically and connectivity-defined projections to elucidate the real-time role of specified pathways in mammalian behavior. Fiber photometry revealed that activity dynamics of a ventral tegmental area (VTA)-to-nucleus accumbens (NAc) projection could encode and predict key features of social, but not novel object, interaction. Consistent with this observation, optogenetic control of cells specifically contributing to this projection was sufficient to modulate social behavior, which was mediated by type 1 dopamine receptor signaling downstream in the NAc. Direct observation of deep projection-specific activity in this way captures a fundamental and previously inaccessible dimension of mammalian circuit dynamics.


Subject(s)
Neural Pathways , Nucleus Accumbens/physiology , Social Behavior , Ventral Tegmental Area/physiology , Animals , Calcium Signaling , Female , Mice , Nucleus Accumbens/cytology , Photometry/methods , Receptors, Dopamine/chemistry , Receptors, Dopamine/metabolism , Reward , Rhodopsin/chemistry , Rhodopsin/metabolism , Ventral Tegmental Area/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...