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










Publication year range
1.
Stroke ; 55(2): 519-522, 2024 02.
Article in English | MEDLINE | ID: mdl-38095119

ABSTRACT

Implantable vagus nerve stimulation, paired with high-dose occupational therapy, has been shown to be effective in improving upper limb function among patients with stroke and received regulatory approval from the US Food and Drug Administration and the Centers for Medicare & Medicaid Services. Combining nonsurgical and surgical approaches of vagus nerve stimulation in recent meta-analyses has resulted in misleading reports on the efficacy of each type of stimulation among patients with stroke. This article aims to clarify the confusion surrounding implantable vagus nerve stimulation as a poststroke treatment option, highlighting the importance of distinguishing between transcutaneous auricular vagus nerve stimulation and implantable vagus nerve stimulation. Recent meta-analyses on vagus nerve stimulation have inappropriately combined studies of fundamentally different interventions, outcome measures, and participant selection, which do not conform to methodological best practices and, hence, cannot be used to deduce the relative efficacy of the different types of vagus nerve stimulation for stroke rehabilitation. Health care providers, patients, and insurers should rely on appropriately designed research to guide well-informed decisions.


Subject(s)
Stroke Rehabilitation , Stroke , Vagus Nerve Stimulation , Aged , United States , Humans , Vagus Nerve Stimulation/methods , Treatment Outcome , Medicare , Stroke/therapy , Stroke Rehabilitation/methods
2.
Commun Med (Lond) ; 2: 37, 2022.
Article in English | MEDLINE | ID: mdl-35603289

ABSTRACT

Background: Functional recovery of arm movement typically plateaus following a stroke, leaving chronic motor deficits. Brain-computer interfaces (BCI) may be a potential treatment for post-stroke deficits. Methods: In this n-of-1 trial (NCT03913286), a person with chronic subcortical stroke with upper-limb motor impairment used a powered elbow-wrist-hand orthosis that opened and closed the affected hand using cortical activity, recorded from a percutaneous BCI comprised of four microelectrode arrays implanted in the ipsilesional precentral gyrus, based on decoding of spiking patterns and high frequency field potentials generated by imagined hand movements. The system was evaluated in a home setting for 12 weeks. Results: Robust single unit activity, modulating with attempted or imagined movement, was present throughout the precentral gyrus. The participant acquired voluntary control over a hand-orthosis, achieving 10 points on the Action Research Arm Test using the BCI, compared to 0 without any device, and 5 using myoelectric control. Strength, spasticity, the Fugl-Meyer scores improved. Conclusions: We demonstrate in a human being that ensembles of individual neurons in the cortex overlying a chronic supratentorial, subcortical stroke remain active and engaged in motor representation and planning and can be used to electrically bypass the stroke and promote limb function. The participant's ability to rapidly acquire control over otherwise paralyzed hand opening, more than 18 months after a stroke, may justify development of a fully implanted movement restoration system to expand the utility of fully implantable BCI to a clinical population that numbers in the tens of millions worldwide.

3.
J Orthop Traumatol ; 23(1): 2, 2022 Jan 06.
Article in English | MEDLINE | ID: mdl-34989884

ABSTRACT

Electrical stimulation is used to elicit muscle contraction and can be utilized for neurorehabilitation following spinal cord injury when paired with voluntary motor training. This technology is now an important therapeutic intervention that results in improvement in motor function in patients with spinal cord injuries. The purpose of this review is to summarize the various forms of electrical stimulation technology that exist and their applications. Furthermore, this paper addresses the potential future of the technology.


Subject(s)
Spinal Cord Injuries , Electric Stimulation , Humans , Spinal Cord Injuries/therapy
5.
Artif Organs ; 45(8): 804-812, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34156104

ABSTRACT

Despite remarkable advances in the treatment of numerous medical conditions, neurological disease and injury remains an outstanding challenge and cause of disability worldwide. The decreased regenerative capacity and extreme complexity and heterogeneity of nervous tissue, in particular the brain, and the fact that the brain remains the least understood organ, have hampered our ability to provide definitive treatments for prevalent conditions such as stroke. Stroke is the second-leading cause of death worldwide, and the nervous system is intimately involved in other prevalent conditions including ischemic heart disease, diabetes mellitus, and hypertension. Advances in neuromodulation, electroceuticals, microsurgical techniques, optogenetics, brain-computer interfaces, and autologous constructs offer potential solutions to address the otherwise permanent neurological deficits of stroke and other conditions. Here we review these various approaches to build an "artificial nervous system" that could restore function and independence in people living with these conditions. We focus on stroke both because it is the leading cause of neurological disability worldwide and because we anticipate that advances in the reversal of stroke-related deficits will have ripple effects benefiting people with other neurological conditions including spinal cord injury, traumatic brain injury, ALS, and muscular dystrophy.


Subject(s)
Stroke Rehabilitation/methods , Animals , Brain-Computer Interfaces , Chronic Disease , Humans , Nerve Transfer , Neurofeedback , Neuronal Plasticity , Prostheses and Implants , Recovery of Function , Wearable Electronic Devices
6.
Sci Adv ; 7(4)2021 01.
Article in English | MEDLINE | ID: mdl-33523957

ABSTRACT

For implantable neural interfaces, functional/clinical outcomes are challenged by limitations in specificity and stability of inorganic microelectrodes. A biological intermediary between microelectrical devices and the brain may improve specificity and longevity through (i) natural synaptic integration with deep neural circuitry, (ii) accessibility on the brain surface, and (iii) optogenetic manipulation for targeted, light-based readout/control. Accordingly, we have developed implantable "living electrodes," living cortical neurons, and axonal tracts protected within soft hydrogel cylinders, for optobiological monitoring/modulation of brain activity. Here, we demonstrate fabrication, rapid axonal outgrowth, reproducible cytoarchitecture, and simultaneous optical stimulation and recording of these tissue engineered constructs in vitro. We also present their transplantation, survival, integration, and optical recording in rat cortex as an in vivo proof of concept for this neural interface paradigm. The creation and characterization of these functional, optically controllable living electrodes are critical steps in developing a new class of optobiological tools for neural interfacing.


Subject(s)
Brain-Computer Interfaces , Animals , Axons , Electrodes, Implanted , Microelectrodes , Neurons/physiology , Rats
7.
Neuroimage ; 225: 117514, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33137477

ABSTRACT

The role of the left ventral lateral parietal cortex (VPC) in episodic memory is hypothesized to include bottom-up attentional orienting to recalled items, according to the dual-attention model (Cabeza et al., 2008). However, its role in memory encoding could be further clarified, with studies showing both positive and negative subsequent memory effects (SMEs). Furthermore, few studies have compared the relative contributions of sub-regions in this functionally heterogeneous area, specifically the anterior VPC (supramarginal gyrus/BA40) and the posterior VPC (angular gyrus/BA39), on a within-subject basis. To elucidate the role of the VPC in episodic encoding, we compared SMEs in the intracranial EEG across multiple frequency bands in the supramarginal gyrus (SmG) and angular gyrus (AnG), as twenty-four epilepsy patients with indwelling electrodes performed a free recall task. We found a significant SME of decreased theta power and increased high gamma power in the VPC overall, and specifically in the SmG. Furthermore, SmG exhibited significantly greater spectral tilt SME from 0.5 to 1.6 s post-stimulus, in which power spectra slope differences between recalled and unrecalled words were greater than in the AnG (p = 0.04). These results affirm the contribution of VPC to episodic memory encoding, and suggest an anterior-posterior dissociation within VPC with respect to its electrophysiological underpinnings.


Subject(s)
Attention/physiology , Memory, Episodic , Mental Recall/physiology , Parietal Lobe/physiology , Drug Resistant Epilepsy , Electrocorticography , Electrodes, Implanted , Humans , Memory/physiology
8.
NPJ Regen Med ; 5: 11, 2020.
Article in English | MEDLINE | ID: mdl-32550009

ABSTRACT

Innervation plays a pivotal role as a driver of tissue and organ development as well as a means for their functional control and modulation. Therefore, innervation should be carefully considered throughout the process of biofabrication of engineered tissues and organs. Unfortunately, innervation has generally been overlooked in most non-neural tissue engineering applications, in part due to the intrinsic complexity of building organs containing heterogeneous native cell types and structures. To achieve proper innervation of engineered tissues and organs, specific host axon populations typically need to be precisely driven to appropriate location(s) within the construct, often over long distances. As such, neural tissue engineering and/or axon guidance strategies should be a necessary adjunct to most organogenesis endeavors across multiple tissue and organ systems. To address this challenge, our team is actively building axon-based "living scaffolds" that may physically wire in during organ development in bioreactors and/or serve as a substrate to effectively drive targeted long-distance growth and integration of host axons after implantation. This article reviews the neuroanatomy and the role of innervation in the functional regulation of cardiac, skeletal, and smooth muscle tissue and highlights potential strategies to promote innervation of biofabricated engineered muscles, as well as the use of "living scaffolds" in this endeavor for both in vitro and in vivo applications. We assert that innervation should be included as a necessary component for tissue and organ biofabrication, and that strategies to orchestrate host axonal integration are advantageous to ensure proper function, tolerance, assimilation, and bio-regulation with the recipient post-implant.

9.
NPJ Parkinsons Dis ; 6: 4, 2020.
Article in English | MEDLINE | ID: mdl-31934611

ABSTRACT

Parkinson's disease (PD) is the second most common progressive neurodegenerative disease, affecting 1-2% of people over 65. The classic motor symptoms of PD result from selective degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc), resulting in a loss of their long axonal projections to the striatum. Current treatment strategies such as dopamine replacement and deep brain stimulation (DBS) can only minimize the symptoms of nigrostriatal degeneration, not directly replace the lost pathway. Regenerative medicine-based solutions are being aggressively pursued with the goal of restoring dopamine levels in the striatum, with several emerging techniques attempting to reconstruct the entire nigrostriatal pathway-a key goal to recreate feedback pathways to ensure proper dopamine regulation. Although many pharmacological, genetic, and optogenetic treatments are being developed, this article focuses on the evolution of transplant therapies for the treatment of PD, including fetal grafts, cell-based implants, and more recent tissue-engineered constructs. Attention is given to cell/tissue sources, efficacy to date, and future challenges that must be overcome to enable robust translation into clinical use. Emerging regenerative medicine therapies are being developed using neurons derived from autologous stem cells, enabling the construction of patient-specific constructs tailored to their particular extent of degeneration. In the upcoming era of restorative neurosurgery, such constructs may directly replace SNpc neurons, restore axon-based dopaminergic inputs to the striatum, and ameliorate motor deficits. These solutions may provide a transformative and scalable solution to permanently replace lost neuroanatomy and improve the lives of millions of people afflicted by PD.

10.
Front Neurosci ; 13: 269, 2019.
Article in English | MEDLINE | ID: mdl-30983957

ABSTRACT

Within the neural engineering field, next-generation implantable neuroelectronic interfaces are being developed using biologically-inspired and/or biologically-derived materials to improve upon the stability and functional lifetime of current interfaces. These technologies use biomaterials, bioactive molecules, living cells, or some combination of these, to promote host neuronal survival, reduce the foreign body response, and improve chronic device-tissue integration. This article provides a general overview of the different strategies, milestones, and evolution of bioactive neural interfaces including electrode material properties, biological coatings, and "decoration" with living cells. Another such biohybrid approach developed in our lab uses preformed implantable micro-tissue featuring long-projecting axonal tracts encased within carrier biomaterial micro-columns. These so-called "living electrodes" have been engineered with carefully tailored material, mechanical, and biological properties to enable natural, synaptic based modulation of specific host circuitry while ultimately being under computer control. This article provides an overview of these living electrodes, including design and fabrication, performance attributes, as well as findings to date characterizing in vitro and in vivo functionality.

11.
J Neural Eng ; 15(5): 056008, 2018 10.
Article in English | MEDLINE | ID: mdl-29855432

ABSTRACT

OBJECTIVE: Micro-tissue engineered neural networks (micro-TENNs) are anatomically-inspired constructs designed to structurally and functionally emulate white matter pathways in the brain. These 3D neural networks feature long axonal tracts spanning discrete neuronal populations contained within a tubular hydrogel, and are being developed to reconstruct damaged axonal pathways in the brain as well as to serve as physiologically-relevant in vitro experimental platforms. The goal of the current study was to characterize the functional properties of these neuronal and axonal networks. APPROACH: Bidirectional micro-TENNs were transduced to express genetically-encoded calcium indicators, and spontaneous fluorescence activity was recorded using real-time microscopy at 20 Hz from specific regions-of-interest in the neuronal populations. Network activity patterns and functional connectivity across the axonal tracts were then assessed using various techniques from statistics and information theory including Pearson cross-correlation, phase synchronization matrices, power spectral analysis, directed transfer function, and transfer entropy. MAIN RESULTS: Pearson cross-correlation, phase synchronization matrices, and power spectral analysis revealed high values of correlation and synchronicity between the spatially segregated neuronal clusters connected by axonal tracts. Specifically, phase synchronization revealed high synchronicity of >0.8 between micro-TENN regions of interest. Normalized directed transfer function and transfer entropy matrices suggested robust information flow between the neuronal populations. Time varying power spectrum analysis revealed the strength of information propagation at various frequencies. Signal power strength was visible at elevated peak levels for dominant delta (1-4 Hz) and theta (4-8 Hz) frequency bands and progressively weakened at higher frequencies. These signal power strength results closely matched normalized directed transfer function analysis where near synchronous information flow was detected between frequencies of 2-5 Hz. SIGNIFICANCE: To our knowledge, this is the first report using directed transfer function and transfer entropy methods based on fluorescent calcium activity to estimate functional connectivity of distinct neuronal populations via long-projecting, 3D axonal tracts in vitro. These functional data will further improve the design and optimization of implantable neural networks that could ultimately be deployed to reconstruct the nervous system to treat neurological disease and injury.


Subject(s)
Axons/physiology , Calcium/chemistry , Neural Networks, Computer , Neural Pathways/physiology , Neuroimaging/methods , Tissue Engineering/methods , Animals , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Delta Rhythm/physiology , Entropy , Female , Fluorescence , Neural Pathways/cytology , Neurons/physiology , Pregnancy , Rats , Theta Rhythm/physiology
12.
Adv Funct Mater ; 28(12)2018 Mar 21.
Article in English | MEDLINE | ID: mdl-34045935

ABSTRACT

Brain-computer interface and neuromodulation strategies relying on penetrating non-organic electrodes/optrodes are limited by an inflammatory foreign body response that ultimately diminishes performance. A novel "biohybrid" strategy is advanced, whereby living neurons, biomaterials, and microelectrode/optical technology are used together to provide a biologically-based vehicle to probe and modulate nervous-system activity. Microtissue engineering techniques are employed to create axon-based "living electrodes", which are columnar microstructures comprised of neuronal population(s) projecting long axonal tracts within the lumen of a hydrogel designed to chaperone delivery into the brain. Upon microinjection, the axonal segment penetrates to prescribed depth for synaptic integration with local host neurons, with the perikaryal segment remaining externalized below conforming electrical-optical arrays. In this paradigm, only the biological component ultimately remains in the brain, potentially attenuating a chronic foreign-body response. Axon-based living electrodes are constructed using multiple neuronal subtypes, each with differential capacity to stimulate, inhibit, and/or modulate neural circuitry based on specificity uniquely afforded by synaptic integration, yet ultimately computer controlled by optical/electrical components on the brain surface. Current efforts are assessing the efficacy of this biohybrid interface for targeted, synaptic-based neuromodulation, and the specificity, spatial density and long-term fidelity versus conventional microelectronic or optical substrates alone.

13.
Front Neurosci ; 11: 373, 2017.
Article in English | MEDLINE | ID: mdl-28713235

ABSTRACT

Pilot clinical trials of human patients implanted with devices that can chronically record and stimulate ensembles of hundreds to thousands of individual neurons offer the possibility of expanding the substrate of cognition. Parallel trains of firing rate activity can be delivered in real-time to an array of intermediate external modules that in turn can trigger parallel trains of stimulation back into the brain. These modules may be built in software, VLSI firmware, or biological tissue as in vitro culture preparations or in vivo ectopic construct organoids. Arrays of modules can be constructed as early stage whole brain emulators, following canonical intra- and inter-regional circuits. By using machine learning algorithms and classic tasks known to activate quasi-orthogonal functional connectivity patterns, bedside testing can rapidly identify ensemble tuning properties and in turn cycle through a sequence of external module architectures to explore which can causatively alter perception and behavior. Whole brain emulation both (1) serves to augment human neural function, compensating for disease and injury as an auxiliary parallel system, and (2) has its independent operation bootstrapped by a human-in-the-loop to identify optimal micro- and macro-architectures, update synaptic weights, and entrain behaviors. In this manner, closed-loop brain-computer interface pilot clinical trials can advance strong artificial intelligence development and forge new therapies to restore independence in children and adults with neurological conditions.

14.
Crit Rev Biomed Eng ; 44(1-2): 123-52, 2016.
Article in English | MEDLINE | ID: mdl-27652455

ABSTRACT

The ideal neuroprosthetic interface permits high-quality neural recording and stimulation of the nervous system while reliably providing clinical benefits over chronic periods. Although current technologies have made notable strides in this direction, significant improvements must be made to better achieve these design goals and satisfy clinical needs. This article provides an overview of the state of neuroprosthetic interfaces, starting with the design and placement of these interfaces before exploring the stimulation and recording platforms yielded from contemporary research. Finally, we outline emerging research trends in an effort to explore the potential next generation of neuroprosthetic interfaces.


Subject(s)
Brain-Computer Interfaces/trends , Neural Prostheses/trends , Prosthesis Design/trends , Prosthesis Implantation/methods , Biomedical Research/trends , Electrodes, Implanted , Humans
15.
Front Syst Neurosci ; 10: 1, 2016.
Article in English | MEDLINE | ID: mdl-26834579

ABSTRACT

Restoring neurological and cognitive function in individuals who have suffered brain damage is one of the principal objectives of modern translational neuroscience. Electrical stimulation approaches, such as deep-brain stimulation, have achieved the most clinical success, but they ultimately may be limited by the computational capacity of the residual cerebral circuitry. An alternative strategy is brain substrate expansion, in which the computational capacity of the brain is augmented through the addition of new processing units and the reconstitution of network connectivity. This latter approach has been explored to some degree using both biological and electronic means but thus far has not demonstrated the ability to reestablish the function of large-scale neuronal networks. In this review, we contend that fulfilling the potential of brain substrate expansion will require a significant shift from current methods that emphasize direct manipulations of the brain (e.g., injections of cellular suspensions and the implantation of multi-electrode arrays) to the generation of more sophisticated neural tissues and neural-electric hybrids in vitro that are subsequently transplanted into the brain. Drawing from neural tissue engineering, stem cell biology, and neural interface technologies, this strategy makes greater use of the manifold techniques available in the laboratory to create biocompatible constructs that recapitulate brain architecture and thus are more easily recognized and utilized by brain networks.

17.
Front Syst Neurosci ; 8: 226, 2014.
Article in English | MEDLINE | ID: mdl-25520632

ABSTRACT

Despite several decades of research into novel brain-implantable devices to treat a range of diseases, only two-cochlear implants for sensorineural hearing loss and deep brain stimulation for movement disorders-have yielded any appreciable clinical benefit. Obstacles to translation include technical factors (e.g., signal loss due to gliosis or micromotion), lack of awareness of current clinical options for patients that the new therapy must outperform, traversing between federal and corporate funding needed to support clinical trials, and insufficient management expertise. This commentary reviews these obstacles preventing the translation of promising new neurotechnologies into clinical application and suggests some principles that interdisciplinary teams in academia and industry could adopt to enhance their chances of success.

18.
Cereb Cortex ; 24(2): 403-13, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23081881

ABSTRACT

The first events in a series exert a powerful influence on cognition and behavior in both humans and animals. This is known as the law of primacy. Here, we analyze the neural correlates of primacy in humans by analyzing electrocorticographic recordings in 84 neurosurgical patients as they studied and subsequently recalled lists of common words. We found that spectral power in the gamma frequency band (28-100 Hz) was elevated at the start of the list and gradually subsided, whereas lower frequency (2-8 Hz) delta and theta band power exhibited the opposite trend. This gradual shift in the power spectrum was found across a widespread network of brain regions. The degree to which the subsequent memory effect was modulated by list (serial) position was most pronounced in medial temporal lobe structures. These results suggest that globally increased gamma and decreased delta-theta spectral powers reflect a brain state that predisposes medial temporal lobe structures to enhance the encoding and maintenance of early list items.


Subject(s)
Brain/physiology , Memory, Episodic , Adolescent , Adult , Brain/surgery , Brain Mapping , Child , Delta Rhythm , Electrodes, Implanted , Electroencephalography , Female , Humans , Male , Middle Aged , Neuropsychological Tests , Prefrontal Cortex/physiology , Temporal Lobe/physiology , Theta Rhythm , Vocabulary , Young Adult
19.
Crit Rev Biomed Eng ; 39(1): 5-28, 2011.
Article in English | MEDLINE | ID: mdl-21488812

ABSTRACT

Brain-machine interfaces (BMI) seek to directly communicate with the human nervous system in order to diagnose and treat intrinsic neurological disorders. While the first generation of these devices has realized significant clinical successes, they often rely on gross electrical stimulation using empirically derived parameters through open-loop mechanisms of action that are not yet fully understood. Their limitations reflect the inherent challenge in developing the next generation of these devices. This review identifies lessons learned from the first generation of BMI devices (chiefly deep brain stimulation), identifying key problems for which the solutions will aid the development of the next generation of technologies. Our analysis examines four hypotheses for the mechanism by which brain stimulation alters surrounding neurophysiologic activity. We then focus on motor prosthetics, describing various approaches to overcoming the problems of decoding neural signals. We next turn to visual prosthetics, an area for which the challenges of signal coding to match neural architecture has been partially overcome. Finally, we close with a review of cortical stimulation, examining basic principles that will be incorporated into the design of future devices. Throughout the review, we relate the issues of each specific topic to the common thread of BMI research: translating new knowledge of network neuroscience into improved devices for neuromodulation.


Subject(s)
Deep Brain Stimulation , Man-Machine Systems , Neural Prostheses , Signal Processing, Computer-Assisted , Cerebral Cortex/physiology , Electroencephalography , Humans
20.
J Neural Eng ; 4(3): 213-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17873423

ABSTRACT

Recent advances in functional electrical stimulation (FES) show significant promise for restoring voluntary movement in patients with paralysis or other severe motor impairments. Current approaches for implantable FES systems involve multisite stimulation, posing research issues related to their physical size, power and signal delivery, surgical and safety challenges. To explore a different means for delivering the stimulus to a distant muscle nerve site, we have elicited in vitro FES response using a high efficiency microcrystal photovoltaic device as a neurostimulator, integrated with a biocompatible glass optical fiber which forms a lossless, interference-free lightwave conduit for signal and energy transport. As a proof of concept demonstration, a sciatic nerve of a frog is stimulated by the microcrystal device connected to a multimode optical fiber (core diameter of 62.5 microm), which converts optical activation pulses ( approximately 100 micros) from an infrared semiconductor laser source (at 852 nm wavelength) into an FES signal.


Subject(s)
Action Potentials/physiology , Electric Stimulation/instrumentation , Fiber Optic Technology/instrumentation , Lasers , Prostheses and Implants , Sciatic Nerve/physiology , Animals , Electric Stimulation/methods , Equipment Design , Equipment Failure Analysis , Miniaturization , Optical Fibers , Rana catesbeiana
SELECTION OF CITATIONS
SEARCH DETAIL
...