Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
Anal Bioanal Chem ; 413(27): 6777-6791, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33961102

ABSTRACT

An SU-8 probe with an array of nine, individually addressable gold microband electrodes (100 µm long, 4 µm wide, separated by 4-µm gaps) was photolithographically fabricated and characterized for detection of low concentrations of chemicals in confined spaces and in vivo studies of biological tissues. The probe's shank (6 mm long, 100 µm wide, 100 µm thick) is flexible, but exhibits sufficient sharpness and rigidity to be inserted into soft tissue. Laser micromachining was used to define probe geometry by spatially revealing the underlying sacrificial aluminum layer, which was then etched to free the probes from a silicon wafer. Perfusion with fluorescent nanobeads showed that, like a carbon fiber electrode, the probe produced no noticeable damage when inserted into rat brain, in contrast to damage from an inserted microdialysis probe. The individual addressability of the electrodes allows single and multiple electrode activation. Redox cycling is possible, where adjacent electrodes serve as generators (that oxidize or reduce molecules) and collectors (that do the opposite) to amplify signals of small concentrations without background subtraction. Information about electrochemical mechanisms and kinetics may also be obtained. Detection limits for potassium ferricyanide in potassium chloride electrolyte of 2.19, 1.25, and 2.08 µM and for dopamine in artificial cerebral spinal fluid of 1.94, 1.08, and 5.66 µM for generators alone and for generators and collectors during redox cycling, respectively, were obtained.


Subject(s)
Dopamine/cerebrospinal fluid , Electrochemical Techniques/instrumentation , Microelectrodes , Animals , Calibration , Corpus Striatum/surgery , Electrochemical Techniques/methods , Electrolytes/chemistry , Ferricyanides/analysis , Ferricyanides/chemistry , Gold , Lasers , Male , Microelectrodes/adverse effects , Microtechnology , Oxidation-Reduction , Polymers/chemistry , Potassium Chloride/chemistry , Rats, Sprague-Dawley
2.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Article in English | MEDLINE | ID: mdl-33846241

ABSTRACT

Microscale needle-electrode devices offer neuronal signal recording capability in brain tissue; however, using needles of smaller geometry to minimize tissue damage causes degradation of electrical properties, including high electrical impedance and low signal-to-noise ratio (SNR) recording. We overcome these limitations using a device assembly technique that uses a single needle-topped amplifier package, called STACK, within a device of ∼1 × 1 mm2 Based on silicon (Si) growth technology, a <3-µm-tip-diameter, 400-µm-length needle electrode was fabricated on a Si block as the module. The high electrical impedance characteristics of the needle electrode were improved by stacking it on the other module of the amplifier. The STACK device exhibited a voltage gain of >0.98 (-0.175 dB), enabling recording of the local field potential and action potentials from the mouse brain in vivo with an improved SNR of 6.2. Additionally, the device allowed us to use a Bluetooth module to demonstrate wireless recording of these neuronal signals; the chronic experiment was also conducted using STACK-implanted mice.


Subject(s)
Electroencephalography/instrumentation , Electrophysiology/instrumentation , Electrophysiology/methods , Action Potentials/physiology , Animals , Brain/physiology , Electric Impedance , Electrodes, Implanted/adverse effects , Electroencephalography/methods , Equipment Design , Male , Mice , Microelectrodes/adverse effects , Neurons/physiology , Signal-To-Noise Ratio
3.
Behav Neurol ; 2020: 4385706, 2020.
Article in English | MEDLINE | ID: mdl-32211080

ABSTRACT

Physiological behaviours such as the sleep-wake cycle and exploratory behaviours are important parameters in intact and sham-operated animals and are usually thought to be unaffected by experimental protocols in which neurosurgery is performed. However, there is insufficient evidence in the literature on the behavioural and cognitive effects observed after deep microelectrode implantation surgery in animal models of neurological diseases. Similarly, in studies that utilize animal models of neurological diseases, the impact of surgery on the pathological phenomena being studied is often minimized. Based on these considerations, we performed a temporal analysis of the effects of deep microelectrode implantation surgery in the hippocampus of rats on quiet wakefulness, sleep, and exploratory activity and the pathological behaviours such as convulsive seizures according to the Racine scale. Male Wistar rats (210-300 g) were used and grouped in sham and epileptic animals. Single doses of pilocarpine hydrochloride (2.4 mg/2 µl; i.c.v.) were administered to the animals to generate spontaneous and recurrent seizures. Deep microelectrode implantation surgeries in both groups and analysis of Fast ripples were performed. Physiological and pathological behaviours were recorded through direct video monitoring of animals (24/7). Our principal findings showed that in epileptic animals, one of the main behaviours affected by surgery is sleep; as a consequence of this behavioural change, a decrease in exploratory activity was also found as well as the mean time spent daily in seizures of scale 4 and the number of seizure events of scales 4 and 5 was increased after surgery. No significant correlations between the occurrence of FR and seizure events of scale 4 (rho 0.63, p value 0.25) or 5 (rho -0.7, p value 0.18) were observed. In conclusion, microelectrode implantation surgeries modified some physiological and pathological behaviours; therefore, it is important to consider this fact when it is working with animal models.


Subject(s)
Electrodes, Implanted/adverse effects , Electrodes, Implanted/psychology , Microelectrodes/adverse effects , Animals , Brain/physiology , Disease Models, Animal , Electrodes, Implanted/veterinary , Electroencephalography/methods , Epilepsy/pathology , Exploratory Behavior/physiology , Hippocampus/drug effects , Hippocampus/physiology , Male , Pilocarpine/pharmacology , Rats , Rats, Wistar , Seizures/physiopathology , Sleep/physiology , Wakefulness/physiology
4.
Acta Biomater ; 102: 205-219, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31733330

ABSTRACT

Higher order tasks in development for brain-computer interfacing applications require the invasiveness of intracortical microelectrodes. Unfortunately, the resulting inflammatory response contributes to the decline of detectable neural signal. The major components of the neuroinflammatory response to microelectrodes have been well-documented with histological imaging, leading to the identification of broad pathways of interest for its inhibition such as oxidative stress and innate immunity. To understand how to mitigate the neuroinflammatory response, a more precise understanding is required. Advancements in genotyping have led the development of new tools for developing temporal gene expression profiles. Therefore, we have meticulously characterized the gene expression profiles of the neuroinflammatory response to mice implanted with non-functional intracortical probes. A time course of differential acute expression of genes of the innate immune response were compared to naïve sham mice, identifying significant changes following implantation. Differential gene expression analysis revealed 22 genes that could inform future therapeutic targets. Particular emphasis is placed on the largest changes in gene expression occurring 24 h post-implantation, and in genes that are involved in multiple innate immune sets including Itgam, Cd14, and Irak4. STATEMENT OF SIGNIFICANCE: Current understanding of the cellular response contributing to the failure of intracortical microelectrodes has been limited to the evaluation of cellular presence around the electrode. Minimal research investigating gene expression profiles of these cells has left a knowledge gap identifying their phenotype. This manuscript represents the first robust investigation of the changes in gene expression levels specific to the innate immune response following intracortical microelectrode implantation. To understand the role of the complement system in response to implanted probes, we performed gene expression profiling over acute time points from implanted subjects and compared them to no-surgery controls. This manuscript provides valuable insights into inflammatory mechanisms at the tissue-probe interface, thus having a high impact on those using intracortical microelectrodes to study and treat neurological diseases and injuries.


Subject(s)
Brain Injuries/physiopathology , Cerebral Cortex/physiopathology , Electrodes, Implanted/adverse effects , Immunity, Innate/genetics , Inflammation/physiopathology , Animals , Brain Injuries/genetics , Cerebral Cortex/surgery , Inflammation/genetics , Male , Mice, Inbred C57BL , Microelectrodes/adverse effects , Transcriptome
5.
Nano Lett ; 19(11): 8032-8039, 2019 11 13.
Article in English | MEDLINE | ID: mdl-31580687

ABSTRACT

Implantable microelectrodes that can be remotely actuated via external fields are promising tools to interface with biological systems at a high degree of precision. Here, we report the development of flexible magnetic microelectrodes (FMµEs) that can be remotely actuated by magnetic fields. The FMµEs consist of flexible microelectrodes integrated with dielectrically encapsulated FeNi (iron-nickel) alloy microactuators. Both magnetic torque- and force-driven actuation of the FMµEs have been demonstrated. Nanoplatinum-coated FMµEs have been applied for in vivo recordings of neural activities from peripheral nerves and cerebral cortex of mice. Moreover, owing to their ultrasmall sizes and mechanical compliance with neural tissues, chronically implanted FMµEs elicited greatly reduced neuronal cell loss in mouse brain compared to conventional stiff probes. The FMµEs open up a variety of new opportunities for electrically interfacing with biological systems in a controlled and minimally invasive manner.


Subject(s)
Cerebral Cortex/physiology , Electrodes, Implanted , Peripheral Nerves/physiology , Alloys/chemistry , Animals , Cerebral Cortex/cytology , Elasticity , Electric Stimulation , Electrodes, Implanted/adverse effects , Equipment Design , Iron/chemistry , Magnetic Fields , Mice , Microelectrodes/adverse effects , Nanostructures/chemistry , Neurons/cytology , Neurons/metabolism , Nickel/chemistry , Peripheral Nerves/cytology , Platinum/chemistry
6.
Clin Neurophysiol ; 130(10): 1781-1788, 2019 10.
Article in English | MEDLINE | ID: mdl-31401486

ABSTRACT

Microneurography, a technique used to detect postganglionic sympathetic nerve traffic in humans, is increasingly used to further the understanding of autonomic regulation in health and disease. The technique involves the transcutaneous insertion of a microelectrode into a peripheral nerve, following which, a variety of adverse acute responses; after-effect and chronic complications have been documented. Here, we comprehensively review the potential adverse outcomes of microneurography and provide updated quantifiable incidence rates of their occurrence within a general population. We also present recommendations for risk assessment and management of such outcomes, as well as recommendations to improve future reporting. This review aims to use objective evidence to improve the understanding of the rare, but present, adverse outcomes of microneurography.


Subject(s)
Electromyography/adverse effects , Microelectrodes/adverse effects , Peripheral Nerves/physiology , Animals , Electromyography/trends , Humans , Microelectrodes/trends , Muscle Weakness/diagnosis , Muscle Weakness/etiology , Muscle Weakness/physiopathology , Pain/diagnosis , Pain/etiology , Pain/physiopathology , Peripheral Nerves/pathology , Peripheral Nervous System Diseases/diagnosis , Peripheral Nervous System Diseases/physiopathology , Treatment Outcome
7.
Sci Rep ; 9(1): 111, 2019 01 14.
Article in English | MEDLINE | ID: mdl-30643182

ABSTRACT

Neural probes designed for extracellular recording of brain electrical activity are traditionally implanted with an insertion speed between 1 µm/s and 1 mm/s into the brain tissue. Although the physical effects of insertion speed on the tissue are well studied, there is a lack of research investigating how the quality of the acquired electrophysiological signal depends on the speed of probe insertion. In this study, we used four different insertion speeds (0.002 mm/s, 0.02 mm/s, 0.1 mm/s, 1 mm/s) to implant high-density silicon probes into deep layers of the somatosensory cortex of ketamine/xylazine anesthetized rats. After implantation, various qualitative and quantitative properties of the recorded cortical activity were compared across different speeds in an acute manner. Our results demonstrate that after the slowest insertion both the signal-to-noise ratio and the number of separable single units were significantly higher compared with those measured after inserting probes at faster speeds. Furthermore, the amplitude of recorded spikes as well as the quality of single unit clusters showed similar speed-dependent differences. Post hoc quantification of the neuronal density around the probe track showed a significantly higher number of NeuN-labelled cells after the slowest insertion compared with the fastest insertion. Our findings suggest that advancing rigid probes slowly (~1 µm/s) into the brain tissue might result in less tissue damage, and thus in neuronal recordings of improved quality compared with measurements obtained after inserting probes with higher speeds.


Subject(s)
Electrodes, Implanted , Microelectrodes/adverse effects , Neurons/physiology , Somatosensory Cortex/physiology , Animals , Rats , Rats, Wistar , Silicon
8.
Turk Neurosurg ; 29(3): 430-433, 2019.
Article in English | MEDLINE | ID: mdl-30649828

ABSTRACT

AIM: To investigate microelectrode recording (MER)-induced microlesion effect (MLE) on the motor symptoms of 30 patients with Parkinson’s disease (PD) who underwent deep brain stimulation of the subthalamic nucleus. MATERIAL AND METHODS: MER-induced MLE was evaluated based on the difference between tremor, rigidity, and bradykinesia scores in the preoperative off-state and intraoperative state following MER and before test stimulation. RESULTS: MLE scores improved by 21.7% [left (L) side] and by 13.6% [right (R) side] from baseline (p < 0.05). Tremor scores improved by 31.5% (L) and by 14.2% (R) (p < 0.05), rigidity scores improved by 17.3% (L) and by 14.2% (R) (p < 0.05) and bradykinesia scores improved by 20.6% (L) and by 11.5% (R) (p < 0.05) from baseline. There was no significant difference between MLE and the number of microelectrodes used (p > 0.05). CONCLUSION: MER-induced MLE improved motor symptoms and was not correlated with the number of microelectrodes used during the procedure.


Subject(s)
Deep Brain Stimulation/instrumentation , Intraoperative Neurophysiological Monitoring/instrumentation , Motor Skills Disorders/surgery , Parkinson Disease/surgery , Subthalamic Nucleus/surgery , Adult , Aged , Deep Brain Stimulation/adverse effects , Deep Brain Stimulation/methods , Female , Humans , Intraoperative Neurophysiological Monitoring/adverse effects , Intraoperative Neurophysiological Monitoring/methods , Male , Microelectrodes/adverse effects , Middle Aged , Motor Skills Disorders/diagnostic imaging , Parkinson Disease/diagnostic imaging , Subthalamic Nucleus/diagnostic imaging
9.
J Neural Eng ; 15(3): 036002, 2018 06.
Article in English | MEDLINE | ID: mdl-29485103

ABSTRACT

OBJECTIVE: Despite the feasibility of short-term neural recordings using implantable microelectrodes, attaining reliable, chronic recordings remains a challenge. Most neural recording devices suffer from a long-term tissue response, including gliosis, at the device-tissue interface. It was hypothesized that smaller, more flexible intracortical probes would limit gliosis by providing a better mechanical match with surrounding tissue. APPROACH: This paper describes the in vivo evaluation of flexible parylene microprobes designed to improve the interface with the adjacent neural tissue to limit gliosis and thereby allow for improved recording longevity. The probes were coated with an ultrafast degrading tyrosine-derived polycarbonate (E5005(2K)) polymer that provides temporary mechanical support for device implantation, yet degrades within 2 h post-implantation. A parametric study of probes of varying dimensions and polymer coating thicknesses were implanted in rat brains. The glial tissue response and neuronal loss were assessed from 72 h to 24 weeks post-implantation via immunohistochemistry. MAIN RESULTS: Experimental results suggest that both probe and polymer coating sizes affect the extent of gliosis. When an appropriate sized coating dimension (100 µm × 100 µm) and small probe (30 µm × 5 µm) was implanted, a minimal post-implantation glial response was observed. No discernible gliosis was detected when compared to tissue where a sham control consisting of a solid degradable polymer shuttle of the same dimensions was inserted. A larger polymer coating (200 µm × 200 µm) device induced a more severe glial response at later time points, suggesting that the initial insertion trauma can affect gliosis even when the polymer shuttle degrades rapidly. A larger degree of gliosis was also observed when comparing a larger sized probe (80 µm × 5 µm) to a smaller probe (30 µm × 5 µm) using the same polymer coating size (100 µm × 100 µm). There was no significant neuronal loss around the implantation sites for most device candidates except the group with largest polymer coating and probe sizes. SIGNIFICANCE: These results suggest that: (1) the degree of mechanical trauma at device implantation and mechanical mismatches at the probe-tissue interface affect long term gliosis; (2) smaller, more flexible probes may minimize the glial response to provide improved tissue biocompatibility when used for chronic neural signal recording; and (3) some degree of glial scarring did not significantly affect neuronal distribution around the probe.


Subject(s)
Absorbable Implants/trends , Cerebral Cortex/metabolism , Electrodes, Implanted/trends , Neuroglia/metabolism , Polymers/metabolism , Xylenes/metabolism , Absorbable Implants/adverse effects , Animals , Cerebral Cortex/surgery , Electrodes, Implanted/adverse effects , Electrodes, Implanted/standards , Male , Microelectrodes/adverse effects , Microelectrodes/standards , Microelectrodes/trends , Polymers/chemical synthesis , Rats , Rats, Sprague-Dawley , Time Factors , Xylenes/chemical synthesis
10.
J Neural Eng ; 15(3): 031001, 2018 06.
Article in English | MEDLINE | ID: mdl-28885187

ABSTRACT

This review intends to present a comprehensive analysis of the mechanical considerations for chronically-implanted neural probes. Failure of neural electrical recordings or stimulation over time has shown to arise from foreign body reaction and device material stability. It seems that devices that match most closely with the mechanical properties of the brain would be more likely to reduce the mechanical stress at the probe/tissue interface, thus improving body acceptance. The use of low Young's modulus polymers instead of hard substrates is one way to enhance this mechanical mimetism, though compliance can be achieved through a variety of means. The reduction of probe width and thickness in comparison to a designated length, the use of soft hydrogel coatings and the release in device tethering to the skull, can also improve device compliance. Paradoxically, the more compliant the device, the more likely it will fail during the insertion process in the brain. Strategies have multiplied this past decade to offer partial or temporary stiffness to the device to overcome this buckling effect. A detailed description of the probe insertion mechanisms is provided to analyze potential sources of implantation failure and the need for a mechanically-enhancing structure. This leads us to present an overview of the strategies that have been put in place over the last ten years to overcome buckling issues. Particularly, great emphasis is put on bioresorbable polymers and their assessment for neural applications. Finally, a discussion is provided on some of the key features for the design of mechanically-reliable, polymer-based next generation of chronic neuroprosthetic devices.


Subject(s)
Brain/surgery , Electrodes, Implanted/standards , Equipment Design/standards , Foreign-Body Reaction/prevention & control , Materials Testing/standards , Animals , Brain/pathology , Electrodes, Implanted/adverse effects , Equipment Design/instrumentation , Equipment Design/methods , Foreign-Body Reaction/etiology , Humans , Hydrogels/adverse effects , Hydrogels/standards , Materials Testing/instrumentation , Materials Testing/methods , Microelectrodes/adverse effects , Microelectrodes/standards , Polymers/adverse effects , Polymers/standards , Stress, Mechanical , Time Factors
11.
Crit Rev Biomed Eng ; 46(4): 341-367, 2018.
Article in English | MEDLINE | ID: mdl-30806249

ABSTRACT

Intracortical microelectrodes exhibit enormous potential for researching the nervous system, steering assistive devices and functional electrode stimulation systems for severely paralyzed individuals, and augmenting the brain with computing power. Unfortunately, intracortical microelectrodes often fail to consistently record signals over clinically useful periods. Biological mechanisms, such as the foreign body response to intracortical microelectrodes and self-perpetuating neuroinflammatory cascades, contribute to the inconsistencies and decline in recording performance. Unfortunately, few studies have directly correlated microelectrode performance with the neuroinflammatory response to the implanted devices. However, of those select studies that have, the role of the innate immune system remains among the most likely links capable of corroborating the results of different studies, across laboratories. Therefore, the overall goal of this review is to highlight the role of innate immunity signaling in the foreign body response to intracortical microelectrodes and hypothesize as to appropriate strategies that may become the most relevant in enabling brain-dwelling electrodes of any geometry, or location, for a range of clinical applications.


Subject(s)
Electrodes, Implanted/adverse effects , Foreign Bodies/immunology , Immunity, Innate , Microelectrodes/adverse effects , Neuroimmunomodulation , Animals , Brain-Computer Interfaces/adverse effects , Cytokines/immunology , Cytokines/physiology , Drosophila , Encephalitis , Humans , Immunity, Innate/immunology , Immunity, Innate/physiology , Neuroimmunomodulation/immunology , Neuroimmunomodulation/physiology
12.
J Neural Eng ; 15(3): 034001, 2018 06.
Article in English | MEDLINE | ID: mdl-29205169

ABSTRACT

OBJECTIVE: Our objective was to determine how readily disruption of the blood-brain barrier (BBB) occurred as a result of bone drilling during a craniotomy to implant microelectrodes in rat cortex. While the phenomenon of heat production during bone drilling is well known, practices to evade damage to the underlying brain tissue are inconsistently practiced and reported in the literature. APPROACH: We conducted a review of the intracortical microelectrode literature to summarize typical approaches to mitigate drill heating during rodent craniotomies. Post mortem skull-surface and transient brain-surface temperatures were experimentally recorded using an infrared camera and thermocouple, respectively. A number of drilling conditions were tested, including varying drill speed and continuous versus intermittent contact. In vivo BBB permeability was assayed 1 h after the craniotomy procedure using Evans blue dye. MAIN RESULTS: Of the reviewed papers that mentioned methods to mitigate thermal damage during craniotomy, saline irrigation was the most frequently cited (in six of seven papers). In post mortem tissues, we observed increases in skull-surface temperature ranging from +3 °C to +21 °C, dependent on drill speed. In vivo, pulsed-drilling (2 s-on/2 s-off) and slow-drilling speeds (1000 r.p.m.) were the most effective methods we studied to mitigate heating effects from drilling, while inconclusive results were obtained with saline irrigation. SIGNIFICANCE: Neuroinflammation, initiated by damage to the BBB and perpetuated by the foreign body response, is thought to play a key role in premature failure of intracortical recording microelectrodes. This study demonstrates the extreme sensitivity of the BBB to overheating caused by bone drilling. To avoid damage to the BBB, the authors recommend that craniotomies be drilled with slow speeds and/or with intermittent drilling with complete removal of the drill from the skull during 'off' periods. While saline alone was ineffective at preventing overheating, its use is still recommended to remove bone dust from the surgical site and to augment other cooling methods.


Subject(s)
Blood-Brain Barrier/metabolism , Cerebral Cortex/metabolism , Craniotomy/adverse effects , Electrodes, Implanted/adverse effects , Hot Temperature/adverse effects , Animals , Blood-Brain Barrier/pathology , Cerebral Cortex/pathology , Craniotomy/trends , Electrodes, Implanted/trends , Humans , Mice , Microelectrodes/adverse effects , Microelectrodes/trends , Rats , Thermography/methods , Thermography/trends
13.
Sci Rep ; 7(1): 15642, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-29142267

ABSTRACT

Silicon neuroprobes hold great potential for studies of large-scale neural activity and brain computer interfaces, but data on brain response in chronic implants is limited. Here we explored with in vivo cellular imaging the response to multisite silicon probes for neural recordings. We tested a chronic implant for mice consisting of a CMOS-compatible silicon probe rigidly implanted in the cortex under a cranial imaging window. Multiunit recordings of cortical neurons with the implant showed no degradation of electrophysiological signals weeks after implantation (mean spike and noise amplitudes of 186 ± 42 µVpp and 16 ± 3.2 µVrms, respectively, n = 5 mice). Two-photon imaging through the cranial window allowed longitudinal monitoring of fluorescently-labeled astrocytes from the second week post implantation for 8 weeks (n = 3 mice). The imaging showed a local increase in astrocyte-related fluorescence that remained stable from the second to the tenth week post implantation. These results demonstrate that, in a standard electrophysiology protocol in mice, rigidly implanted silicon probes can provide good short to medium term chronic recording performance with a limited astrocyte inflammatory response. The precise factors influencing the response to silicon probe implants remain to be elucidated.


Subject(s)
Astrocytes/drug effects , Neocortex/drug effects , Neurons/drug effects , Silicon/toxicity , Animals , Brain-Computer Interfaces , Electrodes, Implanted , Electrophysiology , Mice , Microelectrodes/adverse effects , Neocortex/physiopathology , Neurons/physiology
14.
Sci Rep ; 7(1): 15254, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29127346

ABSTRACT

Intracortical microelectrodes have shown great success in enabling locked-in patients to interact with computers, robotic limbs, and their own electrically driven limbs. The recent advances have inspired world-wide enthusiasm resulting in billions of dollars invested in federal and industrial sponsorships to understanding the brain for rehabilitative applications. Additionally, private philanthropists have also demonstrated excitement in the field by investing in the use of brain interfacing technologies as a means to human augmentation. While the promise of incredible technologies is real, caution must be taken as implications regarding optimal performance and unforeseen side effects following device implantation into the brain are not fully characterized. The current study is aimed to quantify any motor deficit caused by microelectrode implantation in the motor cortex of healthy rats compared to non-implanted controls. Following electrode insertion, rats were tested on an open-field grid test to study gross motor function and a ladder test to study fine motor function. It was discovered that rats with chronically indwelling intracortical microelectrodes exhibited up to an incredible 527% increase in time to complete the fine motor task. This initial study defines the need for further and more robust behavioral testing of potential unintentional harm caused by microelectrode implantation.


Subject(s)
Brain-Computer Interfaces/adverse effects , Electrodes, Implanted/adverse effects , Motor Activity , Motor Cortex/physiopathology , Animals , Humans , Microelectrodes/adverse effects , Rats , Rats, Sprague-Dawley
15.
J Neurosci Methods ; 290: 39-51, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28735876

ABSTRACT

BACKGROUND: Extracellular microelectrode recording (MER) is a prominent technique for studies of extracellular single-unit neuronal activity. In order to achieve robust results in more complex analysis pipelines, it is necessary to have high quality input data with a low amount of artifacts. We show that noise (mainly electromagnetic interference and motion artifacts) may affect more than 25% of the recording length in a clinical MER database. NEW METHOD: We present several methods for automatic detection of noise in MER signals, based on (i) unsupervised detection of stationary segments, (ii) large peaks in the power spectral density, and (iii) a classifier based on multiple time- and frequency-domain features. We evaluate the proposed methods on a manually annotated database of 5735 ten-second MER signals from 58 Parkinson's disease patients. COMPARISON WITH EXISTING METHODS: The existing methods for artifact detection in single-channel MER that have been rigorously tested, are based on unsupervised change-point detection. We show on an extensive real MER database that the presented techniques are better suited for the task of artifact identification and achieve much better results. RESULTS: The best-performing classifiers (bagging and decision tree) achieved artifact classification accuracy of up to 89% on an unseen test set and outperformed the unsupervised techniques by 5-10%. This was close to the level of agreement among raters using manual annotation (93.5%). CONCLUSION: We conclude that the proposed methods are suitable for automatic MER denoising and may help in the efficient elimination of undesirable signal artifacts.


Subject(s)
Artifacts , Brain/cytology , Microelectrodes/adverse effects , Neurons/physiology , Signal Processing, Computer-Assisted , Evoked Potentials/physiology , Fourier Analysis , Humans , Noise , Support Vector Machine
16.
J Neural Eng ; 14(3): 036026, 2017 06.
Article in English | MEDLINE | ID: mdl-28470152

ABSTRACT

OBJECTIVE: Flexible neural probes are hypothesized to reduce the chronic foreign body response (FBR) mainly by reducing the strain-stress caused by an interplay between the tethered probe and the brain's micromotion. However, a large discrepancy of Young's modulus still exists (3-6 orders of magnitude) between the flexible probes and the brain tissue. This raises the question of whether we need to bridge this gap; would increasing the probe flexibility proportionally reduce the FBR? APPROACH: Using novel off-stoichiometry thiol-enes-epoxy (OSTE+) polymer probes developed in our previous work, we quantitatively evaluated the FBR to four types of probes with different softness: silicon (~150 GPa), polyimide (1.5 GPa), OSTE+Hard (300 MPa), and OSTE+Soft (6 MPa). MAIN RESULTS: We observed a significant reduction in the fluorescence intensity of biomarkers for activated microglia/macrophages and blood-brain barrier (BBB) leakiness around the three soft polymer probes compared to the silicon probe, both at 4 weeks and 8 weeks post-implantation. However, we did not observe any consistent differences in the biomarkers among the polymer probes. SIGNIFICANCE: The results suggest that the mechanical compliance of neural probes can mediate the degree of FBR, but its impact diminishes after a hypothetical threshold level. This infers that resolving the mechanical mismatch alone has a limited effect on improving the lifetime of neural implants.


Subject(s)
Brain Injuries/etiology , Brain Injuries/pathology , Electrodes, Implanted/adverse effects , Foreign-Body Reaction/etiology , Foreign-Body Reaction/pathology , Microelectrodes/adverse effects , Neural Prostheses/adverse effects , Animals , Brain Injuries/prevention & control , Elastic Modulus , Electrodes, Implanted/classification , Equipment Design , Equipment Failure Analysis , Foreign-Body Reaction/prevention & control , Mice , Microelectrodes/classification , Neural Prostheses/classification , Reproducibility of Results , Sensitivity and Specificity , Stress, Mechanical
17.
Lancet ; 389(10081): 1821-1830, 2017 05 06.
Article in English | MEDLINE | ID: mdl-28363483

ABSTRACT

BACKGROUND: People with chronic tetraplegia, due to high-cervical spinal cord injury, can regain limb movements through coordinated electrical stimulation of peripheral muscles and nerves, known as functional electrical stimulation (FES). Users typically command FES systems through other preserved, but unrelated and limited in number, volitional movements (eg, facial muscle activity, head movements, shoulder shrugs). We report the findings of an individual with traumatic high-cervical spinal cord injury who coordinated reaching and grasping movements using his own paralysed arm and hand, reanimated through implanted FES, and commanded using his own cortical signals through an intracortical brain-computer interface (iBCI). METHODS: We recruited a participant into the BrainGate2 clinical trial, an ongoing study that obtains safety information regarding an intracortical neural interface device, and investigates the feasibility of people with tetraplegia controlling assistive devices using their cortical signals. Surgical procedures were performed at University Hospitals Cleveland Medical Center (Cleveland, OH, USA). Study procedures and data analyses were performed at Case Western Reserve University (Cleveland, OH, USA) and the US Department of Veterans Affairs, Louis Stokes Cleveland Veterans Affairs Medical Center (Cleveland, OH, USA). The study participant was a 53-year-old man with a spinal cord injury (cervical level 4, American Spinal Injury Association Impairment Scale category A). He received two intracortical microelectrode arrays in the hand area of his motor cortex, and 4 months and 9 months later received a total of 36 implanted percutaneous electrodes in his right upper and lower arm to electrically stimulate his hand, elbow, and shoulder muscles. The participant used a motorised mobile arm support for gravitational assistance and to provide humeral abduction and adduction under cortical control. We assessed the participant's ability to cortically command his paralysed arm to perform simple single-joint arm and hand movements and functionally meaningful multi-joint movements. We compared iBCI control of his paralysed arm with that of a virtual three-dimensional arm. This study is registered with ClinicalTrials.gov, number NCT00912041. FINDINGS: The intracortical implant occurred on Dec 1, 2014, and we are continuing to study the participant. The last session included in this report was Nov 7, 2016. The point-to-point target acquisition sessions began on Oct 8, 2015 (311 days after implant). The participant successfully cortically commanded single-joint and coordinated multi-joint arm movements for point-to-point target acquisitions (80-100% accuracy), using first a virtual arm and second his own arm animated by FES. Using his paralysed arm, the participant volitionally performed self-paced reaches to drink a mug of coffee (successfully completing 11 of 12 attempts within a single session 463 days after implant) and feed himself (717 days after implant). INTERPRETATION: To our knowledge, this is the first report of a combined implanted FES+iBCI neuroprosthesis for restoring both reaching and grasping movements to people with chronic tetraplegia due to spinal cord injury, and represents a major advance, with a clear translational path, for clinically viable neuroprostheses for restoration of reaching and grasping after paralysis. FUNDING: National Institutes of Health, Department of Veterans Affairs.


Subject(s)
Brain-Computer Interfaces/statistics & numerical data , Brain/physiopathology , Hand Strength/physiology , Muscle, Skeletal/physiopathology , Quadriplegia/diagnosis , Spinal Cord Injuries/physiopathology , Brain/surgery , Electric Stimulation Therapy/methods , Electrodes, Implanted/standards , Feasibility Studies , Hand/physiology , Humans , Male , Microelectrodes/adverse effects , Middle Aged , Motor Cortex/physiopathology , Movement/physiology , Quadriplegia/physiopathology , Quadriplegia/surgery , Self-Help Devices/statistics & numerical data , Spinal Cord Injuries/therapy , United States , United States Department of Veterans Affairs , User-Computer Interface
18.
Neuromodulation ; 20(5): 424-428, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28393415

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate the degree of brain tissue injury that could be potentially induced by the introduction of a) microrecording electrodes, b) macrostimulation electrodes, or c) chronic stimulation electrodes. We aimed to evaluate whether the use of five simultaneous microrecording tracks is associated with any brain injury not detectable by conventional imaging such as CT or MRI. MATERIALS AND METHODS: The study included 61 patients who underwent surgery for implantation of 121 DBS leads. In all cases, five simultaneous tracts were utilized for microelectrode recordings. All patients underwent measurements of serum S-100b at specific time points as follows: a) prior to the operation, and b) intraoperatively at specific stages of the procedure: 1) after opening the burr hole, 2) after the insertion of microrecording electrodes, 3) during macrostimulation, 4) at the end of the operation, and 5) on the first postoperative day. RESULTS: The levels of serum S-100B protein remained within the normal range during the entire period of investigation in all patients with the exception of two cases. In both patients, the procedure was complicated by intraparenchymal hemorrhage visible in neuro-imaging. The first patient developed a small intraparenchymal hemorrhage, visible on the postoperative MRI, with no neurological deficit. The second patient experienced a focal epileptic seizure after the insertion of the right DBS chronic lead and the postoperative CT scan revealed a right frontal lobe hemorrhage. CONCLUSION: These results strongly indicate that the insertion of either multiple recording electrodes or the implantation of chronic electrodes in DBS does not increase the risk of brain hemorrhage or of other intracranial complications, and furthermore it does not cause any biochemically detectable brain tissue damage.


Subject(s)
Deep Brain Stimulation/trends , Electrodes, Implanted/trends , Parkinson Disease/blood , Parkinson Disease/diagnostic imaging , S100 Calcium Binding Protein beta Subunit/blood , Subthalamic Nucleus/diagnostic imaging , Adult , Aged , Deep Brain Stimulation/adverse effects , Deep Brain Stimulation/instrumentation , Electrodes, Implanted/adverse effects , Female , Humans , Magnetic Resonance Imaging/methods , Male , Microelectrodes/adverse effects , Microelectrodes/trends , Middle Aged , Parkinson Disease/surgery , Subthalamic Nucleus/chemistry , Tomography, X-Ray Computed/methods
19.
Acta Biomater ; 53: 46-58, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28185910

ABSTRACT

Chronically implanted neural multi-electrode arrays (MEA) are an essential technology for recording electrical signals from neurons and/or modulating neural activity through stimulation. However, current MEAs, regardless of the type, elicit an inflammatory response that ultimately leads to device failure. Traditionally, rigid materials like tungsten and silicon have been employed to interface with the relatively soft neural tissue. The large stiffness mismatch is thought to exacerbate the inflammatory response. In order to minimize the disparity between the device and the brain, we fabricated novel ultrasoft electrodes consisting of elastomers and conducting polymers with mechanical properties much more similar to those of brain tissue than previous neural implants. In this study, these ultrasoft microelectrodes were inserted and released using a stainless steel shuttle with polyethyleneglycol (PEG) glue. The implanted microwires showed functionality in acute neural stimulation. When implanted for 1 or 8weeks, the novel soft implants demonstrated significantly reduced inflammatory tissue response at week 8 compared to tungsten wires of similar dimension and surface chemistry. Furthermore, a higher degree of cell body distortion was found next to the tungsten implants compared to the polymer implants. Our results support the use of these novel ultrasoft electrodes for long term neural implants. STATEMENT OF SIGNIFICANCE: One critical challenge to the translation of neural recording/stimulation electrode technology to clinically viable devices for brain computer interface (BCI) or deep brain stimulation (DBS) applications is the chronic degradation of device performance due to the inflammatory tissue reaction. While many hypothesize that soft and flexible devices elicit reduced inflammatory tissue responses, there has yet to be a rigorous comparison between soft and stiff implants. We have developed an ultra-soft microelectrode with Young's modulus lower than 1MPa, closely mimicking the brain tissue modulus. Here, we present a rigorous histological comparison of this novel ultrasoft electrode and conventional stiff electrode with the same size, shape and surface chemistry, implanted in rat brains for 1-week and 8-weeks. Significant improvement was observed for ultrasoft electrodes, including inflammatory tissue reaction, electrode-tissue integration as well as mechanical disturbance to nearby neurons. A full spectrum of new techniques were developed in this study, from insertion shuttle to in situ sectioning of the microelectrode to automated cell shape analysis, all of which should contribute new methods to the field. Finally, we showed the electrical functionality of the ultrasoft electrode, demonstrating the potential of flexible neural implant devices for future research and clinical use.


Subject(s)
Biocompatible Materials , Electrodes, Implanted , Microelectrodes , Neurons/physiology , Animals , Biocompatible Materials/adverse effects , Biocompatible Materials/chemistry , Blood-Brain Barrier , Electric Conductivity , Electric Stimulation , Electrodes, Implanted/adverse effects , Foreign-Body Reaction/prevention & control , Inflammation/prevention & control , Male , Materials Testing , Microelectrodes/adverse effects , Polymers , Rats , Rats, Sprague-Dawley , Silicone Elastomers , Subthalamic Nucleus/physiology , Subthalamic Nucleus/surgery , Tungsten/adverse effects
20.
JACC Clin Electrophysiol ; 3(3): 220-231, 2017 03.
Article in English | MEDLINE | ID: mdl-29759516

ABSTRACT

OBJECTIVES: This study sought to evaluate an investigational catheter that incorporates 3 microelectrodes embedded along the circumference of a standard 3.5-mm open-irrigated catheter. BACKGROUND: Mapping resolution is influenced by both electrode size and interelectrode spacing. Multielectrode mapping catheters enhance mapping resolution within scar compared with standard ablation catheters; however, this requires the use of 2 separate catheters for mapping and ablation. METHODS: Six swine with healed infarction and 2 healthy controls underwent mapping of the left ventricle using a THERMOCOOL SMARTTOUCH SF catheter with 3 additional microelectrodes (0.167 mm2) along its circumference (Qdot, Biosense Webster, Diamond Bar, California). Mapping resolution in healthy and scarred tissue was compared between the standard electrodes and microelectrodes using electrogram characteristics, cardiac magnetic resonance, and histology. RESULTS: In healthy myocardium, bipolar voltage amplitude was similar between the standard electrodes and microelectrodes, with a fifth percentile of 1.19 and 1.30 mV, respectively. In healed infarction, the area of low bipolar voltage (defined as <1.5 mV) was smaller with microelectrodes (16.8 cm2 vs. 25.3 cm2; p = 0.033). Specifically, the microelectrodes detected zones of increased bipolar voltage amplitude, with normal electrogram characteristics occurring at the end of or after the QRS, consistent with channels of preserved subendocardium. Identification of surviving subendocardium by the microelectrodes was consistent with cardiac magnetic resonance and histology. The microelectrodes also improved distinction between near-field and far-field electrograms, with more precise identification of scar border zones. CONCLUSIONS: This novel catheter combines high-resolution mapping and radiofrequency ablation with an open-irrigated, tissue contact-sensing technology. It improves scar mapping resolution while limiting the need for and cost associated with the use of a separate mapping catheter.


Subject(s)
Body Surface Potential Mapping/instrumentation , Catheter Ablation/instrumentation , Cicatrix/physiopathology , Electrophysiologic Techniques, Cardiac/instrumentation , Heart Ventricles/physiopathology , Microelectrodes/adverse effects , Animals , Cicatrix/pathology , Electrocardiography , Electrophysiologic Techniques, Cardiac/methods , Endocardium/pathology , Endocardium/physiopathology , Heart Ventricles/diagnostic imaging , Heart Ventricles/pathology , Magnetic Resonance Spectroscopy/methods , Male , Microelectrodes/standards , Myocardial Infarction/physiopathology , Myocardium/pathology , Prospective Studies , Swine , Tachycardia, Ventricular/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...