ArticlePDF Available

Electrode Failure Tissue, Electrical, and Material Responses

Authors:

Abstract and Figures

The development of invasive, rehabilitative neuroprosthetics for humans requires reliable neural probes that are capable of recording large ensembles of neurons for a long period of time. Recent advances in the development of neuroprosthetics in animals and humans have shown that communication and control can be directly derived from the central nervous system (CNS) for restoring lost motor ability. This proof of concept has opened the possibility of new therapies for the millions of individuals suffering from neurological disorders of the nervous system. The success of these therapies hinges on the ability to reliably access the relevant signals from the brain with high quality for the lifetime of the patient. As a result, research has focused on the cascade of events that follow chronic implantation of microelectrodes and temporal degradation in the signal and electrode quality: signal-to-noise ratio, noise floor, peak amplitude, and neuronal yield. Implanted microelectrodes have been reported to suffer from time-dependent degradation in signal quality due to unknown issues related to tissue interfaces.
Content may be subject to copyright.
30 IEEE PUL SE
JANUARY/FEB RUARY 2012
By Wolfgang J. Streit,
Qing-Shan Xue,
Abhishek Prasad,
Viswanath Sankar,
Eric Knott, Aubrey Dyer,
John R. Reynolds,
Toshikazu Nishida,
Gerald P. Shaw,
and Justin C. Sanchez
2154-2 287/12/$31. 00©2012 I EEE
Electrode Failure
Date of publi cation: 6 Februar y 2012
The development of invasive, rehabilitative neuropros-
thetics for humans requires reliable neural probes that
are capable of recording large ensembles of neurons for
a long period of time.
Recent advances in the development of neuropros-
thetics i n animals and humans have shown that com-
munication and control can be directly derived from the cen-
tral nervous system (CNS) for restoring lost motor ability [1].
This proof of concept has opened the possibility of new thera-
pies for the millions of individuals suffering from neurological
disorders of the nervous system. The success of these therapies
hinges on the ability to reliably access the relevant signals from
the brain with high quality for the lifetime of the patient. As a
result, research has focused on the cascade of events that follow
chron ic implantation of microelec trodes a nd temporal degrada-
tion in the signal and electrode quality: signal-to-noise ratio,
noise floor, peak amplitude, and neuronal yield. Implanted mi-
cr oe lectrodes have b een rep or ted to suffer f rom ti me -dependent
degradation in signal quality due to unknown issues related to
tissue interfaces.
The long-term reliability of microelectrode recording im-
plants has been correlated with two broad categories of factors:
biotic (issues related to the electrode itself) and abiotic (issues
related to the neural tissue). To conduct a comprehensive evalu-
ation of both abiotic and biotic aspects of electrode performance,
we have assembled a team of investigators composed of neu-
robiologists, biomedical and electrical engineers, and electro-
chemists working toward elucidating key mechanisms involved
in chronic microelectrode implantation. Previous studies in this
field have stressed, on the abiotic side, physical changes such as
damaged insulation, change in surface area, and oxidation and
Tissue, Electrical, and
Material Responses
Digital Obj ect Identifie r 10.1109/MPUL. 2011.2175632
JANUARY/FEB RUARY 2012
IEEE PUL SE 31
corrosion of electrodes; and on the biotic side, neuroinflamma-
tion, blood brain barrier disruption, tissue encapsulation, and
astroglial scarring [2]. We will build on and ex-
pand these prior studies by performing coupled
biotic–abiotic investigations. These include scan-
ning electron microscopy of electrodes both pre-
implantation and postimplantation, monitoring
changes in electrode impedance pre, post, and
during implantation, and measuring a number of
biological markers of injury in the cerebral spinal
fluid (CSF) and serum. These combined metrics
support our idea that degeneration of microg-
lial cells is a cellular event that occurs following
prolonged implantation and can be critically tied
to the degradation of neuronal signals. Because
microglial degeneration represents a relatively
recent concept in neuropathology [3] and still
has not been studied in the current context,
we briefly outline our rationale for postulating that microglial
degeneration is a pivotal cellular event in electrode failure.
Microglial Activation Versus
Degeneration for Neural Electrodes
In flammation i s defi ned as the cel lular response to tissue injury,
and the key cellular element involved in neuroinflammation
following CNS injury is the microglial cell. In short, microglia
constitute a population of endogenous CNS glial cells that are
neuroprotective and can perform immunological functions, i.e.,
they comprise the brain’s immune system [4]. Because microglia
are ubiquitous throughout the CNS, any acute injury quickly
triggers the activation of local microglia whose main task is to
restore tissue homeostasis and orchestrate the wound healing
process. Clearly, the implantation of electrodes into the living
brain constitutes an injury, and thus previous studies have re-
ported microglial activation as a conspicuous cellular event [2],
[5]. The problem that arises with long-term implantation of
electrodes is that the injurious stimulus does not go away, and
therefore activation of microglial cells endures and leads to a
chronic neuroinflammatory response. Although little is known
about the fate of microglia persisting in such a chronic state of
activation, there is reason to believe that long-term activation
eventually takes its toll and causes m icroglial cells to degenerate
[6]. This idea primarily stems from prior work done in neurode-
generative diseases such as Alzheimer’s disease and amyotrophic
lateral sclerosis, which have shown a direct correlation between
microglial and neuronal degeneration [3]. Thus, we believe that
degeneration and loss of microglia results in loss of neuroprotec-
tion that contributes to the onset of neurodegenerative changes,
which in the case of chronically implanted recording electrodes
becomes manifest in a loss of neuronal signal strength and qual-
ity. Based on this hypothesis, the main (biotic) goals of this proj-
ect are to 1) ascertain the time line of microglial degeneration, 2)
define molecular mechanisms that cause microg-
lial degeneration, and 3) find ways of interfering
with these mechanisms, thereby preventing both
microglial and neuronal damages and thus ex-
tending the working life of implanted electrodes.
Experimental Test Beds
In addition to the biotic goals, the observed
abiotic effec ts, which include the electrode’s
physical changes (damaged insulation, change
in sur face area, oxidation/reduction, and cor-
rosion) [7], will also impact the electrical
recordi ng properties of the neural probe fol-
lowing prolonged ex posure to the brain mi-
croenvironment in vivo. Although many of
these effec ts are typically studied individual-
ly, a deeper understanding of the true nature of the cause(s)
of electrode failure likely requires the coupling of both biotic
and abiotic perspectives. This combined scientific perspec-
tive upon which we have based our work may enable us to
explain the underlying mechanisms governing the effects
of chronic electrode implantation as well as derive from the
quantification of the various parameters new predictions
that would improve electrode performance in the future.
© BRAND X PICTURES
Recent advances in
the development of
neuroprosthetics in
animals and humans
have shown that
communication and
control can be derived
directly from the
CNS for restoring lost
motor ability.
32 IEEE PUL SE
JANUARY/FEB RUARY 2012
Thus, results from a multifaceted approach can provide a
more complete understanding of the effects of biotic–abiotic
interactions [8]. To better understand the spatiotemporal
responses that occur at the most fundamental levels, it is
necessar y to develop data acquisition methods that best cap-
ture the biochemical, structural, and electrophysiological
responses in real time. A key problem i n investigating the
failure of chronic microelectrodes is the temporal resolution
FIGURE 1 Coupling of biotic and abiotic metrics throughout the lifetime of electrodes.
Quantitative Time Line for In Vivo Studies
Acute Recovery Chronic Failure
Reported Electrophysiological Signal Quality
Simultaneous Electrophysiology, Histopathology, and Microdialysis
(Dots Indicate Sacrifice Points)
Surgery
Impact
Inflammation
Impact
Chronic Impact Between Neural Matrix
and Electrodes
Aggregation of Metrics for
Modeling and Statistical
Analysis to Rank Importance
of the Following:
Abiotic Abiotic Abiotic Biotic
Biotic
Biotic
• Geometry
• Impedance
• Signal-to-
Noise (SNR)
• Impedance
• SNR
• Insulation Failure
• Recording Site
Failure
• Impedance
• SNR
• Microglia Numbers
• Neuron Numbers
• Morphometry
• Biomarkers of
Injury
• Neuronal, Axonal
Integrity
• Region of Interest
• Effective Rate of Failure
• Variance in Failure
• Linear–Nonlinear
Relationship Among
Factors
• Cells of Interest
• Signaling Molecules of
Interest
• Electrode Integrity
• BBB
• Biomarkers
of Injury
• Impedance
• BBB
• Biomarkers
of Injury
• Synaptic
Changes
Hours 1–14 Days Time
1, 6, 12, and 18 Months
FIGURE 2 Scanning electron microscopy of electrode recording sites, (a) preimplant and (b) postimplant. Note that the recording tip
has undergone a transformation in its morphology. In addition, the amount of tungsten relative to the polyimide insulation has
been reduced. Implant duration here is seven days.
(a) (b)
JANUARY/FEB RUARY 2012
IEEE PUL SE 33
required to assess chronic performance. In an
effort to solve this problem, we have devel-
op ed t est b eds t hat e n a ble the rea l-ti me c olle c-
tion of abiotic/biotic metr ics and bring to bear
new probing capabilities. Included in these
metrics is the essential evaluation of neuronal
function, which is characterized with respect
to the electrode–tissue interaction.
Figure 1
provides a summary of metrics and shows a
time line of key phases known to be associ-
ated with electrode failure: acute, recovery,
chronic, and failure. Figure 1 is an average
representation of identifiable phases useful for guiding our
investigation.
This test bed that we have developed has the capability
to support large parallel studies of electrode failure mecha-
ni sms with pr ec ise ti me re so lution. I n add it io n to the tiss ue
reactions, there is a strong possibility that this test bed can
yield insight on microstructural features of electrodes that
evoke tissue reactions leading to failure. I n these studies,
we are investigating arrays of metal microwires. As shown
in
Figure 2
, surface modification of polyim ide-insulated
tungsten microwires (50 μm diameter) not only alters the
electrical properties, but also changes the surface area and
adhesive properties. The changes shown in Figure 2 are
representative of short implant durations (seven days) and
likely a contributing factor of failure for long-term implan-
tation (years). As electrode surface chemistry changes, it’s
affinity for adhesive molecules in the extracellular fluid
also changes. Electrode corrosion (physical and chemical)
and surface adhesiveness (biochemical and biological) can
be effectively evaluated. Every electrode that enters into
this study undergoes a quantitative preimplantation i n-
vestigation to show how certain electrode characteristics
correlate with the failure rate in vivo. By comparing these
rates, as in the pre- and postimages of Figure 2, we can
determine the corrosive feature responsible (surface charge
or porosity), demonstrate the adhesive molecules that bind
the corroded electrode, and then show which cell type pref-
erential ly attaches to the fai lure electrode to result in func-
tional changes. These features become predictive of failure
and can be correlated with the in vivo analyses of perielec-
trode cellular reactions.
Models to Predict Electrode Health
One of the hallmarks of this work is that a variety of new failure
markers and measurement test beds are being developed. With
these data, it is possible to systematically aggregate and evalu-
ate these metr ics in terms of their abil ity to pred ict electrode
health. These models will accept as inputs real-time biomark-
ers, impedance, and corrosion measurements, and these met-
rics will be used to predict thresholds for signal-to-noise-ratio,
electrode yield, and action potential peak-to-peak amplitude.
With these models, it will be possible to estimate the transi-
tion probabilities between specific neuronal marker states and
among the markers themselves to indicate the mechan isms of
action of electrode–tissue interactions. The knowledge gained
from these studies will help to elucidate the de-
sign of future neural electrodes that are capable
of high-performance function for long duration.
Acknowledgment
This work was sponsored by the Defense Ad-
vanced Research Projects Agency Microsystems
Technology Office under the auspices of Dr. Jack
Judy (jack.judy@darpa.mil) through the Space
and Naval Warfare Systems Center, Pacific Grant
N66001-11-1- 4 0 0 9
.
The views, opinions, and/or findings con-
tained in this article are those of the author and should not be
interpreted as representing the official views or policies, either
expressed or implied, of the Defense Advanced Research Projects
Agency or the Department of Defense.
Wolfgang J. Streit (pschorr@ufl.edu), Qing-Shan Xue (qsxue@
ufl.edu), and Gerald P. Shaw (shaw@mbi.uf l.edu) are with
the Department of Neuroscience, University of Florida, Gainesville,
Florida. Toshikazu Nishida (nishida@ufl.edu) and Viswanath
Sankar (vsanka1@ufl.edu) are with the Department of Electri-
cal and Computer Engineering, University of Florida, Gainesville,
Florida. Eric Knott (eknott@chem.ufl.edu), Aubrey Dyer (au-
breydyer@gmail.com), and John R. Reynolds (reynolds@chem.
ufl.edu), are with the Department of Chemistry, University of Flor-
ida, Gainesville, Florida. Abhishek Prasad (apr009@gmail.com)
is with the Neuroprosthetics Research Laboratory at the University
of Miami, Florida. Justin C. Sanchez (jcsanchez@ miami.edu)
is with the Departments of Biomedical Engineering, Neuroscience,
and the Miami Project to Cure Paralysis, University of Miami, Coral
Gables, Florida.
References
[ 1] R. A. Normann, “Technology Insight: Future neuroprosthetic therapies
for disorders of the nervous system,” Nature Clin. Pract. Neurol., vol. 3,
no. 8 , pp. 444–452, 2007.
[ 2] V. S. Polikov, P. A. Tresco, a nd W. M. Reichert, “Response of brain
tissue to c hron ical ly impla nted neural elect rodes,” J. Neurosci.
Methods, vol. 148, pp. 1–18, Oct. 2005.
[ 3] W. J. Streit and Q. S. Xue, “ Life and death of m icrog lia,” J Neuroim-
mune Pharmacol., vol. 4, pp. 371–379, Dec. 2009.
[ 4] W. J. Streit and C. A. Kincaid-Colton, “The brain’s immune sys-
tem,” Sci. Amer., vol. 273, no. 5, pp. 54– 61, 1995.
[ 5] B. D. Winslow and P. A. Tresco, “Quantitative analysis of the tis-
sue response to chronically implanted microwire electrodes in rat
cortex,” Biomaterials, vol. 31, no. 7, pp. 1558–1567, Mar. 2010.
[ 6] M. B. Graeber and W. J. Streit, “Microglia: Biology and pathol-
ogy,” Acta Neuropathol., vol. 119, no. 1, pp. 89–105, Jan. 2010.
[ 7] L. A. Geddes and R. Roeder, “Criteria for the select ion of materi-
als for implanted elec trodes,” Ann. Biomed. Eng., vol. 31, no. 7, pp.
879– 890, June 2003.
[ 8] T. W. Berger, J. K. Chapin, G. A. Gerhardt, D. J. McFarland, J. C.
Pri ncipe, W. V. Soussou, D. M. Taylor, a nd P. A. Tresco, “Interna-
tional assessment of research and development in brain –computer
interfaces,” Baltimore, M D, WT EC Panel Rep., 2007.
Degeneration and
loss of microglia
results in a loss of
neuroprotection
that contributes
to the onset of
neurodegenerative
changes.
... It has been shown that the mean failure time of implanted silicon microelectrode arrays (MEAs) in non-human primate is 334 days [2]. Implant failure, especially under chronic conditions, is likely due to multiple abiotic (i.e., probe corrosion and insulation failure) and biotic factors (i.e., loss of neurons at the interface, gliosis and oxidative stress due to neuroinflammation, etc.) [3][4][5][6][7][8][9][10][11][12]. Initial penetration of the probes into brain tissue induces tissue shearing that propagates inflammation and oxidative stress, which are hypothesized to result in a source of implant failure [13]. ...
Article
Full-text available
Several recent studies have reported improved histological and electrophysiological outcomes with soft neural interfaces that have elastic moduli ranging from 10 s of kPa to hundreds of MPa. However, many of these soft interfaces use custom fabrication processes. We test the hypothesis that a readily adoptable fabrication process for only coating the tips of microelectrodes with soft brain-like (elastic modulus of ~5 kPa) material improves the long-term electrical performance of neural interfaces. Conventional tungsten microelectrodes (n = 9 with soft coatings and n = 6 uncoated controls) and Pt/Ir microelectrodes (n = 16 with soft coatings) were implanted in six animals for durations ranging from 5 weeks to over 1 year in a subset of rats. Electrochemical impedance spectroscopy was used to assess the quality of the brain tissue–electrode interface under chronic conditions. Neural recordings were assessed for unit activity and signal quality. Electrodes with soft, silicone coatings showed relatively stable electrical impedance characteristics over 6 weeks to >1 year compared to the uncoated control electrodes. Single unit activity recorded by coated electrodes showed larger peak-to-peak amplitudes and increased number of detectable neurons compared to uncoated controls over 6–7 weeks. We demonstrate the feasibility of using a readily translatable process to create brain-like soft interfaces that can potentially overcome variable performance associated with chronic rigid neural interfaces.
... Neural probes that fail for abiotic reasons after a long period of implantation go through three phases of mostly biotic influences and finally an abiotic failure phase. These four time windows are termed "acute", "recovery", "chronic", and "failure" [103,108]. The acute time window spans several hours after the implantation. ...
Thesis
Diese Arbeit befasst sich mit der Entwicklung, Charakterisierung und der In- Vivo-Validierung eines neuen, zylindrischen hochauflösenden intrakaniellen Implantats zur Aufzeichnung und Stimulation neuronaler Signale. Fokale Epilepsie wurde als klinisches Applikationsszenario herangezogen um grundlegende Anforderungen, wie Implantatgeometry, Implantationsdauer und aufzuzeichnende Signale, zu definieren. Von diesen Anforderungen ausgehend, werden dem Implantat weitere Funktionalitäten hinzugefügt, um die Spanne der möglichen Anwendungen zu erweitern. Die Spanne potentieller Anwendungen umfasst fundamentale neuronale Forschung und die Behandlung der Parkinson-Krankheit, Dystonie und Depression. Die hier gezeigte neuronale Sonde hat eine zehnfach höhere Anzahl an Elektroden pro Implantat, d.h. 128 Elektroden, als klinisch zugelassene Sonden. Dies ermöglicht das Aufzeichnen neuronaler Signale mit erhöhter Auflösung verglichen zum Stand der Technik. Des Weiteren ermöglicht die Sonde das Aufzeichnen von Aktionspotentialen einzelner Neurone. Dies ermöglicht bisher nur äußerst selten angewandte diagnostische Methoden für Epilepsie bei unerreichter Auflösung. Der in dieser Arbeit genutzte Herstellungsprozess verwendet fotolithografische Methoden in Kombination mit Polyimidsubstraten, um Platin-Leiterbahnen und -Elektroden mit Strukturgrößen ab 3 μm mikromechanisch zu strukturieren. Anschließend werden die Substrate durch Rollen innerhalb einer Metallform bei 300°C ausgehärtet und so in ihre zylindrische Sondenform gebracht. Eine zusätzliche Cytop-Schicht wird als Trockenkleber verwendet, um die Sondenform zu fixieren und den Umfang der Sonde zu verschließen. Das hohle innere Volumen der Sonde wird dazu genutzt, um LEDs für optogenetische Forschung als auch CMOS-Chips zum Zwecke des Multiplexings der gewonnenen Signale zu integrieren. Um die mechanische Stabilität der Sonde zu gewährleisten, wird anschließend der Rest der Sonde mit einem Epoxidharz gefüllt. Die Sonden werden über die Dauer von 100 Tagen mittels eines speziell angefertigten Impedanzspektrometers elektrisch charakterisiert. Während dieser Periode werden die Sonden 37°C warmer Ringerlösung ausgesetzt, um so die biologische Umgebung nachzuahmen. Dabei werden zunächst biologische Zersetzungsfaktoren außer Acht gelassen. Die Hälfte der Elektroden einer jeden Sonde werden mit 1000 Stimulationspulsen von 1 V Amplitude und 1 ms Dauer pro Stunde beaufschlagt. Dies dient als erste Evaluation für klinische Zwecke, bei denen elektrische Stimulation benötigt wird. In-vivo-Implantationen in Ratten und einem Affen zeigen, dass es möglich ist, über die Dauer von 10 Tagen stabil Aktionspotentiale aufzuzeichnen, welches die ungefähre Imlpantationsdauer für die Diagnose fokaler Epilepsie ist. Die längste nachgewiesene Dauer, nach der eine Sonde es noch erlaubt Aktionspotentiale auf zu zeichnen, ist 13 Wochen. Dies ist in Übereinstimmung mit den Ergebnissen der elektrischen Charakterisierungsmessungen in Ringerlösung und damit ein Hinweis, dass elektrochemische Prozesse die Zersetzung der Sonde beherrschen. Da die Elektrodenschicht nur 300 nm dünn ist, ist eine Hypothese, dass Sonden mit stärkerer Elektrodenschicht der Zersetzung länger Stand halten. Um diese Hypothese zu testen, wurden die Elektroden einer Sonde nach dem Rollprozess galvanisch mit Gold beschichtet und 37°C warmer Ringerlösung ausgesetzt. Die Impedanz der Elektroden hat sich über die gesamte Dauer von 30 Tagen nicht geändert. Daraus lässt sich schließen, dass Sonden mit zusätzlicher Elektrodenschicht, oder unterschiedlichem Material, dem Zersetzungsprozess standhalten können. Dies ebnet den Weg zu chronischen Implantationen mit Anwendungen, welche die Dauer von 10 Tagen überschreiten. Des Weiteren wird in dieser Arbeit ein thermomechanisches Model des verwendeten Polyimidsubstrats etabliert. Dieses Model erlaubt es, mechanische Restspannungen und Dehnungen des Polyimids zu berechnen, nachdem dieses machanischen Belastungen und Temperaturen zwischen 22°C und 300°C für Zeitdauern bis zu 24 Stunden ausgesetzt wurde.
... Recording yield, which is a measure of device recording efficacy, was significantly impaired at the onset of implantation in cuprizonetreated mice (Fig. 3a). Given that other established modes of recording performance failure, such as glial scar formation and neurodegeneration, are identified as late-stage events that occur following chronic device implantation (> 2-4 weeks post-implantation), this decrease in yield can likely be attributed to the absence of cortical oligodendrocytes and myelin membranes following the cuprizone pre-treatment [94]. Reduced single-unit yields can suggest a failure in signal transduction of demyelinated axons, resulting in decreased signal amplitudes of more distal neurons, which would not be threshold detected by the recording device. ...
Article
Biological inflammation induced during penetrating cortical injury can disrupt functional neuronal and glial activity within the cortex, resulting in potential recording failure of chronically implanted neural interfaces. Oligodendrocytes provide critical support for neuronal health and function through direct contact with neuronal soma and axons within the cortex. Given their fundamental role to regulate neuronal activity via myelin, coupled with their heightened vulnerability to metabolic brain injury due to high energetic demands, oligodendrocytes are hypothesized as a possible source of biological failure in declining recording performances of intracortical microelectrode devices. To determine the extent of their contribution to neuronal activity and function, a cuprizone-inducible model of oligodendrocyte depletion and demyelination in mice was performed prior to microelectrode implantation. At 5 weeks of cuprizone exposure, mice demonstrated significantly reduced cortical oligodendrocyte density and myelin expression. Mice were then implanted with functional recording microelectrodes in the visual cortex and neuronal activity was evaluated up to 7 weeks alongside continued cuprizone administration. Cuprizone-induced oligodendrocyte loss and demyelination was associated with significantly reduced recording performances at the onset of implantation, which remained relatively stable over time. In contast, recording performances for mice on a normal diet were intially elevated before decreasing over time to the recording level of tcuprizone-treated mice. Further electrophysiological analysis revealed deficits in multi-unit firing rates, frequency-dependent disruptions in neuronal oscillations, and altered laminar communication within the cortex of cuprizone-treated mice. Post-mortem immunohistochemistry revealed robust depletion of oligodendrocytes around implanted microelectrode arrays alongside comparable neuronal densities to control mice, suggesting that oligodendrocyte loss was a possible contributor to chronically impaired device performances. This study highlights potentially significant contributions from the oligodendrocyte lineage population concerning the biological integration and long-term functional performance of neural interfacing technology.
... The lobar regions of the brain, by contrast, bear the brunt of load forces during rapid head movements, as evidenced by the patterns of diffuse axonal injury (Davceva, Basheska et al. 2015, Jang andLee 2017). Even in the absence of overt pathology, micro-motion at the tissueelectrode interface is considered a predominant factor in the functional degradation of 15 chronic electrodes implanted in the cortex (Streit, Xue et al. 2012). This degradation is a critical barrier to clinical implementation of brain-machine interfaces. ...
Article
Innovations in neuroprosthetics have restored sensorimotor function to paralysis patients and amputees. However, to date there is a lack of solutions available to adequately address the needs of spinal cord injury patients (SCI). In this dissertation we develop a novel sensor-brain interface (SBI) that delivers electric microstimulation to the cuneate nucleus (CN) to restore somatosensory feedback in patients with intact limbs. In Chapter II, we develop a fully passive liquid metal antenna using gallium-indium (GaIn) alloy injected in polydimethylsiloxane (PDM) channels to measure forces within the physiological sensitivity of a human fingertip. In Chapter III, we present the first chronic neural interface with the CN in primates to provide access to long-term unit recordings and stimulation. In Chapter IV, we demonstrate that microstimulation to the CN is detectable in a Three Alternative Force Choice Oddity task in awake behaving primates. In Chapter V, we explore the downstream effects of CN stimulation on primary somatosensory cortex, in the context of spontaneous and evoked spindles under sedation. In summary, these findings constitute a proof-of-concept for the sensory half of a bidirectional sensorimotor prosthesis in the CN.
... Though the overall stability of an implanted device is governed by many factors [10][11][12][13] the above mentioned electrophysiological changes have been associated with the underlying changes in biology surrounding an implanted device [5,[14][15][16]. Specifically, the decline in recording quality is attributed to a local loss of neuronal population [16], whereas an increase in impedance is associated with the formation of the glial sheath, which is primarily composed of activated microglia, macrophages, and reactive astrocytes surrounding an implanted device [14][15][16]. ...
Article
Full-text available
Objective: Neuroinflammation has long been associated with the performance decline of intracortical microelectrodes (IMEs). Consequently, several strategies, including the use of anti-inflammatories, have been employed to mitigate the inflammation surrounding IMEs. However, these strategies have had limited success towards achieving a chronically viable cortical neural interface, questioning the efficacy of anti-inflammatory approach. Approach: Herein, we conducted a systematic study in rats implanted with functional devices by modulating inflammation via systemic injection of lipopolysaccharide (LPS), dexamethasone (DEX), a combination of both, or none to assess the degree of inflammation on device functionality. We hypothesized that implanted rats treated with LPS will have a negative impact, and rats treated with DEX will have a positive impact on functionality IMEs and histological outcome. Main results: Contrary to our hypothesis, we did not observe adverse effects in recording metrics among different groups with LPS and/or DEX treatment despite alterations in initial pro-inflammatory markers. We also did not observe any functional benefit of anti-inflammatory treatment. Regardless of the treatment conditions, the recording quality degraded at chronic time points. In end-point histology, implanted rats that received LPS had significantly lower NeuN density and higher levels of CD68 surrounding the implant site, indicative of the pro-inflammatory effect of LPS, which, however, contradicted with the recorded results. Significance: Collectively, our results suggest that acute inflammatory events may not be the key driver for functional degradation of IMEs. Future intervention strategies geared towards improving the functional longevity of intracortical devices may benefit using multi-modal approaches rather than a single approach, such as controlling the initial inflammatory response.
Article
In order to develop long-lifetime neural electrodes, the insertion tissue injury caused by two optimized neural electrode (convex streamline electrode and vibration attenuation electrode) models were evaluated compared with a reference electrode. Based on the experimental evaluation system for testing tissue injury, the effects of insertion speeds on tissue injury of the two optimized electrodes with different insertion depths were studied. The maximum tissue strain caused by the two optimized neural electrodes firstly increased and then decreased with the increase of insertion speed at the depths of 3 mm and 4.5 mm. The insertion forces caused by vibration attenuation electrode are steady with the change of insertion speed. The convex streamline neural electrode caused less tissue injury compared with the other two electrodes. The higher or lower insertion speed causes smaller tissue strain for the two optimized electrodes, which is conductive to set implantation parameters to minimize tissue injury.
Article
In order to develop long-lifetime neural electrodes, the insertion tissue injury caused by two optimized neural electrode (convex streamline electrode and vibration attenuation electrode) models were evaluated compared with a reference electrode. Based on the experimental evaluation system for testing tissue injury, the effects of insertion speeds on tissue injury of the two optimized electrodes with different insertion depths were studied. The maximum tissue strain caused by the two optimized neural electrodes firstly increased and then decreased with the increase of insertion speed at the depths of 3 mm and 4.5 mm. The insertion forces caused by vibration attenuation electrode are steady with the change of insertion speed. The convex streamline neural electrode caused less tissue injury compared with the other two electrodes. The higher or lower insertion speed causes smaller tissue strain for the two optimized electrodes, which is conductive to set implantation parameters to minimize tissue injury.
Article
Objective: Neural interfacing technologies could significantly improve quality of life for people living with the loss of a limb. Both motor commands and sensory feedback must be considered; these complementary systems are segregated from one another in the spinal nerve. Approach: The dorsal root ganglion-ventral root (DRG-VR) complex was targeted chronically with floating microelectrode arrays (FMAs) designed to record from motor neuron axons in the VR or stimulate sensory neurons in the DRG. Hematoxylin and eosin and Nissl/Luxol fast blue staining were performed. Characterization of the tissue response in regions of interest and pixel-based image analyses were used to quantify MAC387 (monocytes/macrophages), NF200 (axons), S100 (Schwann cells), vimentin (fibroblasts, endothelial cells, astrocytes), and GLUT1 (glucose transport proteins) reactivity. Implanted roots were compared to non-implanted roots and differences between the VR and DRG examined. Main results: The tissue response associated with chronic array implantation in this peripheral location is similar to that observed in central nervous system locations. Markers of inflammation were increased in implanted roots relative to control roots with MAC387 positive cells distributed throughout the region corresponding to the device footprint. Significant decreases in neuronal density and myelination were observed in both the VR, which contains only neuronal axons, and the DRG, which contains both neuronal axons and cell bodies. Notably, decreases in NF200 in the VR were observed only at implant times less than ten weeks. Observations related to the blood-nerve barrier and tissue integrity suggest that tissue remodeling occurs, particularly in the VR. Significance: This study was designed to assess the viability of the DRG-VR complex as a site for neural interfacing applications and suggests that continued efforts to mitigate the tissue response will be critical to achieve the overall goal of a long-term, reliable neural interface.
Article
Neuroprosthetics and neuromodulation represent a promising field for several related applications in the central and peripheral nervous system, such as the treatment of neurological disorders, the control of external robotic devices, and the restoration of lost tactile functions. These actions are allowed by the neural interface, a miniaturized implantable device that most commonly exploits electrical energy to fulfill these operations. A neural interface must be biocompatible, stable over time, low invasive, and highly selective; the challenge is to develop a safe, compact, and reliable tool for clinical applications. In case of anatomical impairments, neuroprosthetics is bound to the need of exploring the surrounding environment by fast‐responsive and highly sensitive artificial tactile sensors that mimic the natural sense of touch. Tactile sensors and neural interfaces are closely interconnected since the readouts from the first are required to convey information to the neural implantable apparatus. The role of these devices is pivotal hence technical improvements are essential to ensure a secure system to be eventually adopted in daily life. This review highlights the fundamental criteria for the design and microfabrication of neural interfaces and artificial tactile sensors, their use in clinical applications, and future enhancements for the release of a second generation of devices. A comprehensive review on neural interfaces and artificial tactile sensors is presented. A special highlight on the fundamental criteria for the design, working principles, materials, and technical considerations for the development of these implantable devices is discussed, with a special focus on their clinical application in neuroprosthetics and their future progress toward optimized solutions.
Article
In order to reveal the effects of insertion speed with hydrogel coatings on tissue injury, comparative experiments were carried out with the naked neural probes (as the reference) and probes with PVA-H coatings based on tissue injury evaluation system. It was found that the coatings of the neural probes would cause more serious tissue injury during the insertion process. And the insertion speed could alleviate the negative effects on the tissue injury caused by implantation of neural probes.
Article
Full-text available
Brain-computer interface (BCI) research deals with establishing communication pathways between the brain and external devices. BCI systems can be broadly classified depending on the placement of the electrodes used to detect and measure neurons firing in the brain: in invasive systems, electrodes are inserted directly into the cortex; in noninvasive systems, they are placed on the scalp and use electroencephalography or electrocorticography to detect neuron activity. This WTEC study was designed to gather information on worldwide status and trends in BCI research and to disseminate it to government decision makers and the research community. The study reviewed and assessed the state of the art in sensor technology, the biotic/abiotic interface and biocompatibility, data analysis and modeling, hardware implementation, systems engineering, functional electrical stimulation, noninvasive communication systems, and cognitive and emotional neuroprostheses in academic research and industry.
Book
Brain-computer interface (BCI) research deals with establishing communication pathways between the brain and external devices where such pathways do not otherwise exist. Throughout the world, such research is surprisingly extensive and expanding. BCI research is rapidly approaching a level of first-generation medical practice for use by individuals whose neural pathways are damaged, and use of BCI technologies is accelerating rapidly in nonmedical arenas of commerce as well, particularly in the gaming, automotive, and robotics industries. The technologies used for BCI purposes are cutting-edge, enabling, and synergistic in many interrelated arenas, including signal processing, neural tissue engineering, multiscale modeling, systems integration, and robotics. This WTEC study gathered information on worldwide status and trends in BCI research to disseminate to government decisionmakers and the research community. The study reviewed and assessed the state of the art in sensor technology, the biotic-abiotic interface and biocompatibility, data analysis and modeling, hardware implementation, systems engineering, functional electrical stimulation, noninvasive communication systems, and cognitive and emotional neuroprostheses in academic research and industry. The study also compared the distinctly different foci, range, and investment levels of BCI research programs in the United States, Canada, China, Europe, and Japan.
Article
The past 20 years have seen a gain in knowledge on microglia biology and microglia functions in disease that exceeds the expectations formulated when the microglia "immune network" was introduced. More than 10,000 articles have been published during this time. Important new research avenues of clinical importance have opened up such as the role of microglia in pain and in brain tumors. New controversies have also emerged such as the question of whether microglia are active or reactive players in neurodegenerative disease conditions, or whether they may be victims themselves. Premature commercial interests may be responsible for some of the confusion that currently surrounds microglia in both the Alzheimer and Parkinson's disease research fields. A critical review of the literature shows that the concept of "(micro)glial inflammation" is still open to interpretation, despite a prevailing slant towards a negative meaning. Perhaps the most exciting foreseeable development concerns research on the role of microglia in synaptic plasticity, which is expected to yield an answer to the question whether microglia are the brain's electricians. This review provides an analysis of the latest developments in the microglia field.
Article
Several hypotheses have been proposed to explain how the brain tissue reaction to single unit recording electrodes influences biocompatibility including progressive changes in the spatial distribution of reactive astrocytes, and the loss of neurons over the indwelling period. To examine these hypotheses, the spatial distribution of biomarkers associated with the foreign body response to insulated microwires placed in rat cerebral cortex was analyzed 2, 4, and 12 weeks after implantation using quantitative methods. We observed a stereotypical tissue response that was similar in some aspects to that previously reported for penetrating planar silicon microelectrode arrays with some specific differences including an overall lower degree of cortical tissue reactivity. While we found no evidence that reactive gliosis increases over time or that neuronal loss is progressive, we did find evidence of persistent inflammation and enhanced BBB permeability at the electrode brain tissue interface that extended over the 3 month indwelling period and that exhibited more animal to animal variability at 3 months than at 2 and 4 weeks.
Article
The importance of microglial cells in the maintenance of a well-functioning central nervous system (CNS) cannot be overstated. As descendants of the myelomonocytic lineage they are industrious housekeepers and watchful sentries that safeguard a homeostatic environment through a number of mechanisms designed to provide protection of fastidious neurons at all times. Microglia become particularly active after homeostasis has been perturbed by physical injury or other insults and they enter into a state of activation which is determined largely by the nature and severity of the lesion. Microglial activation is the main cellular event in acute neuroinflammation and essential for wound healing in the CNS. Recent studies from this laboratory have been focused on microglia in the aging brain and identified structural abnormalities, termed microglial dystrophy, that are consistent with cell senescence and progress to a form of accidental cell death that is marked by cytoplasmic degeneration and has been termed cytorrhexis. Cytorrhexis of microglia is infrequent in the normally aged human brain and non-detectable in aged rodents, but its occurrence increases dramatically during neurodegenerative conditions, including Alzheimer's disease (AD) in humans and motoneuron disease in transgenic rats. The identification of degenerating microglia has given rise to a novel theory of AD pathogenesis, the microglial dysfunction hypothesis, which views the loss of microglial neuroprotection as a central event in neurodegenerative disease development.
Article
There are four criteria that must be considered when choosing material for an implanted electrode: (1) tissue response, (2) allergic response, (3) electrode-tissue impedance, and (4) radiographic visibility. This paper discusses these four criteria and identifies the materials that are the best candidates for such electrodes. For electrodes that make ohmic contact with tissues: gold, platinum, platinum–iridium, tungsten, and tantalum are good candidates. The preferred insulating materials are polyimide and glass. The characteristics of stimulator output circuits and the importance of the bidirectional wave- form in relation to electrode decomposition are discussed. The paper concludes with an analysis, the design criteria, and the special properties and materials for capacitive recording and stimulating electrodes. © 2003 Biomedical Engineering Society. PAC2003: 8754Dt, 8780Fe, 8768+z, 8719Nn
Article
Chronically implanted recording electrode arrays linked to prosthetics have the potential to make positive impacts on patients suffering from full or partial paralysis. Such arrays are implanted into the patient's cortical tissue and record extracellular potentials from nearby neurons, allowing the information encoded by the neuronal discharges to control external devices. While such systems perform well during acute recordings, they often fail to function reliably in clinically relevant chronic settings. Available evidence suggests that a major failure mode of electrode arrays is the brain tissue reaction against these implants, making the biocompatibility of implanted electrodes a primary concern in device design. This review presents the biological components and time course of the acute and chronic tissue reaction in brain tissue, analyses the brain tissue response of current electrode systems, and comments on the various material science and bioactive strategies undertaken by electrode designers to enhance electrode performance.
Article
Most disorders of the nervous system result from localized sensory or motor pathologies attributable to disease or trauma. The emerging field of neuroprosthetics is focused on the development of therapeutic interventions that will be able to restore some of this lost neural function by selective electrical stimulation of sensory or motor pathways, or by harnessing activity recorded from remnant neural pathways. A key element in this restoration of function has been the development of a new generation of penetrating microelectrode arrays that provide unprecedented selective access to the neurons of the CNS and PNS. The active tips of these microelectrode arrays penetrate the nervous tissues and abut against small populations of neurons or nerve fibers, thereby providing selective access to these cells. These electrode arrays are not only beginning to provide researchers with the ability to better study the spatiotemporal information processing performed by the nervous system, they can also form the basis for new therapies for disorders of the nervous system. In this Review, three examples of this new generation of microelectrode arrays are described, as are potential therapeutic applications in blindness and spinal cord injury, and for the control of prosthetic limbs.