Sie sind auf Seite 1von 46

Home Search Collections Journals About Contact us My IOPscience

Progress towards biocompatible intracortical microelectrodes for neural interfacing

applications

This content has been downloaded from IOPscience. Please scroll down to see the full text.

2015 J. Neural Eng. 12 011001

(http://iopscience.iop.org/1741-2552/12/1/011001)

View the table of contents for this issue, or go to the journal homepage for more

Download details:

IP Address: 134.225.1.226
This content was downloaded on 01/01/2015 at 05:32

Please note that terms and conditions apply.


Journal of Neural Engineering

J. Neural Eng. 12 (2015) 011001 (45pp) doi:10.1088/1741-2560/12/1/011001

Topical Review

Progress towards biocompatible


intracortical microelectrodes for neural
interfacing applications
Mehdi Jor1,5, John L Skousen2,3,5, Christoph Weder1 and
Jeffrey R Capadona2,4
1
Adolphe Merkle Institute, University of Fribourg, Rte de lAncienne Papeterie, CH-1723 Marly,
Switzerland
2
Advanced Platform Technology Center, Rehabilitation Research and Development, Louis Stokes
Cleveland VA Medical Center, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH 44106-
1702, USA
3
Department of Biomedical Engineering, University of Utah, James LeVoy Sorenson Molecular
Biotechnology Building, 36 S. Wasatch Drive, Rm. 3100, Salt Lake City, UT 84121, USA
4
Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr.
Drive, Wickenden Bldg, Cleveland, OH 44106, USA

E-mail: jeffrey.capadona@case.edu

Received 12 May 2014, revised 30 September 2014


Accepted for publication 23 October 2014
Published 2 December 2014

Abstract
To ensure long-term consistent neural recordings, next-generation intracortical microelectrodes
are being developed with an increased emphasis on reducing the neuro-inammatory response.
The increased emphasis stems from the improved understanding of the multifaceted role that
inammation may play in disrupting both biologic and abiologic components of the overall
neural interface circuit. To combat neuro-inammation and improve recording quality, the eld
is actively progressing from traditional inorganic materials towards approaches that either
minimizes the microelectrode footprint or that incorporate compliant materials, bioactive
molecules, conducting polymers or nanomaterials. However, the immune-privileged cortical
tissue introduces an added complexity compared to other biomedical applications that remains to
be fully understood. This review provides a comprehensive reection on the current
understanding of the key failure modes that may impact intracortical microelectrode
performance. In addition, a detailed overview of the current status of various materials-based
approaches that have gained interest for neural interfacing applications is presented, and key
challenges that remain to be overcome are discussed. Finally, we present our vision on the future
directions of materials-based treatments to improve intracortical microelectrodes for neural
interfacing.
Keywords: intracortical microelectrodes, brain machine interfaces, neuro-inammatory response,
neurodegeneration, biocompatibility
(Some gures may appear in colour only in the online journal)

5
Denotes an equal contribution of co-rst authorship.

1741-2560/15/011001+45$33.00 1 2015 IOP Publishing Ltd Printed in the UK


J. Neural Eng. 12 (2015) 011001 M Jorfi et al

In particular, the evolution of traditional intracortical


microelectrode systems is discussed from a materials science
perspective (section 2). Emphasis is given to key develop-
ments that have facilitated the longest and highest quality
in vivo recordings. In addition, a number of primary failure
modes are discussed that must be overcome to achieve the full
potential of intracortical microelectrodes for in vivo recording
applications (section 3). Lastly, the impressive progress that
has been made in recent years to develop the next generation
of intracortical microelectrodes is reviewed (section 4). By
framing recent advancements within the context of current
successes, the most promising strategies are highlighted and
the most critical challenges for improving intracortical elec-
trode-based neural interfaces are discussed.

Figure 1. Examples of recording neural electrodes for brain machine


interface devices. (A) EEG activity is recorded non-invasively with
electrodes placed on the scalp. (B) ECoG electrodes are placed either 2. Traditional intracortical microelectrodes for brain
outside the dura mater (epidural ECoG) or under the dura mater machine interfacing
(subdural ECoG) and can record neural activity on the cortical
surface. (C) Intracortical microelectrodes penetrate the cortex and
can record action potentials from individual or small populations of A number of intracortical microelectrodes have been designed
neurons within the cortex. to interface with cortical neurons, including insulated metal
microwires and semiconductor-based devices such as the
Michigan and Utah electrode arrays (UEAs). Regardless of
1. Introduction
the specic design or manufacturer, a similar compound cir-
Neural interfaces bridge the central nervous system to the cuit can be used to describe how microelectrodes extract
outside world. Originally, neural interfaces were developed as electrical signals generated from single target neurons
a basic science tool, and as such, have been used extensively (gure 2). Extensive descriptions of each of the primary
to develop our understanding of how the nervous system portions of the compound circuit are available elsewhere
works [14]. Additionally, neural interfaces hold great [18, 19], and therefore only a brief description will be
potential for functional restoration in persons with paralysis, included here.
other forms of motor dysfunction, or limb loss. Such reha- The rst portion of the circuit involves the cortical col-
bilitative applications are commonly referred to as brain umn and the complex set of presynaptic inputs that innervate
machine (or brain computer) interfaces [5]. In brain machine the target neurons being recorded from. These inputs can be
interface (BMI) applications, a recording device is used to both excitatory and inhibitory. If a sufcient excitatory
extract volitional intent in the form of consciously modulated postsynaptic potential is created then a compound action
neuronal signals. Using a variety of signal transducing sys- potential is generated through depolarization of the axon
tems and processing algorithms, extracted neural signals can hillock. The ion-based signal then travels through the extra-
then be used to drive external devices such as limb prostheses cellular space to the electrode-recording site. As transport is
or computers [612]. primarily diffusion based, the distance traveled and the
A number of types of recording electrode devices have impedance of the extracellular space governs the strength of
been developed to access different forms of neural informa- the signal reaching the recording site. It has been suggested
tion through varying levels of invasiveness (gure 1). For that the maximum effective recording range for classic
example, non-penetrating recording electrodes placed exter-
microelectrode designs is roughly 50150 m [20].
nally on the scalp or on the brain surface can gain functional
At the recording site, the electric potential produced by
information [11, 12]. However, many researchers believe that
the ion-based signal is recorded as a voltage change. Signals
recording devices that penetrate into specic regions of the
can then be amplied and analyzed using various acquisition
brain will provide the most useful control signals for complex
BMI applications [13]. Despite the potential that penetrating and processing techniques [21]. Once analyzed, algorithms
intracortical microelectrodes have shown, widespread imple- are applied to translate the signal into device commands/
mentation is impeded by the inability to consistently record orders that carry out the users volitional intent [2123].
high quality neural signals over clinically relevant time Output devices can vary from application to application and
frames [1417]. As such, this review focuses on intracortical have ranged from moving a cursor on a computer screen, to
microelectrodes implanted within the cerebral cortex, which facilitating a robot to walk on a treadmill, driving a wheel-
record from single or small populations of nearby neurons. chair, or controlling a robotic arm [24].

2
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

Figure 2. A commonly used equivalent circuit model (Robinson Model) of metal microelectrode recoding in the brain. Signals at the tip of
the microelectrode (Vsig) generate currents (I) that ow to ground through the microelectrode and effective amplier circuit, creating the
potential (Vin) at the input of the amplier before being recorded (Vrec); Rs is the resistance of the electrolyte; Re is the leakage resistance
which models the ow of the charge carriers crossing the electric double layer; Ce is the capacitance of the microelectrode-electrolyte
interface; Rm is the resistance of the microelectrode; Cs is all the shunt capacitance to ground and Za is the input impedance of the amplier.
Thus, the effective impedance of the microelectrode is comprised of the resistance of the electrolyte (Rs), the resistance and capacitance of the
double layer interface of the electrolyte (Re and Ce) and the (negligible) resistance of the microelectrode (Rm). The impedance of the
microelectrode is frequency dependent. At low frequencies, the impedance is dominated by the series combination of Rs and Re, whereas at
high frequencies Ce bypasses the effect of Re so that the impedance is now close to Rs. Thus, by measuring the impedance of an electrode at
high and low frequencies, it is possible to determine the component values for the equivalent circuit.

2.1. Microwires: from an acute electrophysiology tool to a Several new electrode designs and materials were intro-
useful interface between man and machines duced in the 1970s that permitted recordings to be performed
for longer durations. A collaboration between Salcman, Bak
Metal wire electrodes have an extensive history as the go to
and Schmidt at the National Institutes of Health (NIH) led to
tool of neuroscientists for acute electrophysiology experi-
the development of microwire electrodes from iridium (Ir),
ments. Therefore, it is not surprising that metal wire electro-
platinum (Pt) and platinum-iridium- alloys (Pt/Ir-) [28, 29].
des have been further developed to extend their use to long-
Either a glass [28] or poly (monochloro-p-xylene) (Parylene-
term BMI applications. Prominent developments include C) coating [29] was incorporated to insulate the microwire
reducing wire size, enhancing electrode geometry as well as electrodes. Iridium electrodes in particular demonstrated that
optimizing both the underlying conducting and insulating microwire-based electrodes are capable of chronic recording
materials. A schematic example of a microwire electrodes by detecting single unit activity from primate cortex up to 223
described in the literature can be found in gure 3(A). days after implantation [29, 30]. However, in addition to the
Many of the earliest descriptions of metal wire electrodes
promise shown by the NIH studies, the authors also observed
used to record from single or small populations of neurons
that average recording performance was inconsistent and
date back to the 1940s. Renshaw performed one of the earliest
decreased over time [30]. Salcman, Bak and Schimdt were
studies utilizing metal wire electrodes to record electrical among the rst to propose that inammatory-mediated device
signals from single neurons, using Ag/AgCl based electrodes encapsulation reduced recording performance over time by
[3]. Other metals that would prove more effective and safer in increasing impedance and electrically isolating the device
chronic recording applications, such as stainless steel, tung- from the surrounding tissue [30, 31]. Section 3.4 provides
sten and platinum, also had roots during this early period further information on inammatory and encapsulation-
[1, 25, 26]. For example, in 1942, Grundfest and Campbell mediated electrode failure.
conducted one of the rst studies utilizing stainless steel Over the last few decades, a number of groups have
electrodes to record electrical impulses from neurons within shown that a variety of microwire devices are capable of
the feline spinal cord [1]. Improving upon their original recording the signals needed for BMI applications over
design, Grundfest next began utilizing electrolytic pointing to extended periods [16, 30, 32, 33]. Despite these successes, a
create sharpened stainless steel microwires [2]. Electrolytic major hurdle for microwire-based microelectrodes is still the
pointing reduced the variability between individual electrodes challenge of consistently recording high quality units over
and improved insertion into cortical tissue. time [34, 35].
Despite the limited recording duration (hours to days) of
early devices, metal recording electrodes facilitated a rapid
increase in knowledge concerning neural pathways and 2.2. Silicon-based microelectrodes
volitional movement. For example, in 1966, Evarts described
that specic patterns of neuronal activity correlate with set 2.2.1. Michigan-style microelectrodes. Beginning in the
motor responses [27]. Specically, Evarts found that the 1960s, advancements in semiconducting materials and
electrical activity of pyramidal neurons in the precentral improvements in micromachining capabilities drove the
cortex of primates correlated with specic behavior patterns. development of silicon-based microelectrodes. Based on
The results of Evarts study contributed greatly to the idea of foundational work with silicon etching for beam-lead
using volitionally controlled neural signals to manipulate integrated circuits at Bell Telephone Laboratories, Angell,
external devices. Starr and Wise developed the rst intracortical microelectrode

3
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

Figure 3. Schematic representation of the generation design of leading microelectrode array technologies, including (A) microwires, (B)
Michigan-style microelectrodes, (C) Utah electrode arrays (EUA) and (D) cone (glass pipette) electrodes.

with a silicon substrate in 1966 [36]. Similar to existing [39]. First a diffused boron etch stop is created using a
microwire devices, early silicon-based devices consisted of a thermal oxide mask to dene the substrates dimensions. Etch
penetrating tine with an exposed conducting tip capable of stops allows for all processes to be performed on the topside
electrically interacting with nearby cells [36, 37]. of the silicon wafer rather than having to pattern both sides of
By the 1980s, further work by Wise and colleagues at the wafer, as was done when creating the original MI-style
the University of Michigan led to the development of what is devices. Following denition of the probe dimensions, a
commonly referred to as the Michigan (MI)-style microelec- dielectric layer is added to insulate the backside of the device.
trode [38]. Applying newly developed microfabrication The dielectric usually consists of a silicon oxide/silicon
processes such as diffusion-based etch stops, silicon micro- nitride stack or an alternative passivation layer. Building on
electrodes were fabricated with multiple recording sites the dielectrics, a series of conducting traces are applied to the
placed along a single or multiple planar shanks [39]. The length of the probe to link the recording sites to the bond
advantage of MI-style electrodes over traditional metal pads. The recording sites and bond pads are then created from
microwire devices is their ability to record from numerous conducting metals such as gold or iridium. A second stack of
sites at well-controlled tissue depths. stress-compensated silicon oxide/silicon nitride is then
The basic structure of a single-shank MI-style micro- deposited through chemical vapor deposition to insulate the
electrode is shown in gure 3(B). Several microfabrication conducting traces. To further shield the device and protect the
processes are used in the creation of MI-style microelectrodes dielectrics from dissolution under in vivo conditions

4
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

(section 3.3), insulating polymer coatings such as Parylene-C manipulated to create devices that better conform to complex
or Epoxylite have been adopted over time. Additionally, anatomical geometries using variable depth dicing and wet
improved hermetic protection through anodic silicon-glass isotropic etching [49]. Ultra-high aspect ratio devices made
bonding for on-chip processors has also been developed. from highly conductive bulk silicon have been created using
A number of groups have shown that MI-style micro- microwire electrical discharge machining [50]. In addition,
electrodes are capable of chronic recording in a variety of high density arrays have also recently been created [51].
species [4042]. As with microwires, despite a number of Similar to the MI-style devices, signicant recent efforts
studies showing that chronic recording is feasible, the major have focused on creating wireless versions of the UEA, where
hurdle for MI-style microelectrodes has been the challenge of power and telemetry systems are incorporated on the base of
consistently recording high quality units over time [43]. the UEA via ip-chip bonding [52, 53]. Other developments
Today MI-style microelectrodes are being further devel- that are being pursued include the addition of optical
oped at a number of universities and laboratories. MI-style waveguides for optogentic research [54].
microelectrodes are also commercially available for neu- The ability of the UEA to record the neural signals
roscience and preclinical applications from NeuroNexus, a needed for chronic rehabilitative applications has been
subsidiary of GreatBatch Inc. Advanced MI-style microelec- demonstrated by a number of groups. For example, the
trodes have been developed with on-chip processing as well UEA has been utilized for recording in the visual and auditory
as wireless telemetry systems. Additionally, microuidics and cortex [55, 56]. Additionally a number of primate studies
optical waveguides have been incorporated to expand the have shown the usefulness of the UEA in BMI applications,
number of ways in which MI-style microelectrodes can such as cursor and prostheses control [57, 58].
interact with the surrounding tissue. However, as discussed in The commercialization and clinical translation of the
section 3, several factors still limit the clinical success of MI- UEA began by spinning-off Bionic Technologies from the
style microelectrode technology. For further details on the University of Utah in 1997. Cyberkinetics Neurotechnology
development and successes of MI-style microelectrodes, Systems, Inc. later acquired this spin-off. In 2004, the FDA
readers are referred to the excellent review by Wise [39]. granted Cyberkinetics the rst of two Investigational Device
Exemptions (IDEs) to begin human clinical trials with a
UEA-based system (BrainGate).
2.2.2. The UEA. Normann and colleagues developed an After achieving major milestones in the clinical devel-
alternative, silicon-based microelectrode, which due to its opment of the UEA, portions of Cyberkinetics were acquired
origin at the University of Utah is referred to as the UEA [44]. in 2008 by two distinct, yet collaborative, entities. Blackrock
Instead of the thin lm design of the MI-style arrays, the UEA Microsystems, LLC, under Dr Florian Solzbacher, acquired
uses glass reow, dicing and etching to create an array of rights to many of the underlying hardware components.
well-dened penetrating electrode tines. Figure 3(C) shows a Today, Blackrock Microsystems, LLC operates as the original
basic schematic of the UEA along with the slanted UEA equipment manufacturer for the UEA. The BrainGate, co-
design created using slight modications of the original founded by Jeffery Stibel, acquired rights to the BrainGate
processing steps (described below). While originally designed Neural Interface System and many of Cyberkinetics clinical
for stimulation applications, the UEA has been widely used as applications for the technology. Further information can be
a recording tool. In fact, the UEA is the only high-density, found on Blackrock Microsystems and the BrainGate
penetrating recording electrode approved by the US Food and websites.
Drug Administration and that has received the CE mark for As part of the BrainGate clinical trials, Hochberg et al
use in Europe. have shown that UEAs implanted into the primary motor
Since the rst generation, signicant development efforts cortex can be used to restore volitional control of external
have been devoted to improve the performance of the UEA. devices, including a computer cursor and a simple robotic
For example, to improve charge transfer, Pt/Ti/W/Pt, and then hand, to patients with tetraplegia [7]. Building on Hochbergs
subsequently sputtered iridium oxide (SIROF), have been seminal work, recently another publication from the Brain-
used instead of the original gold or platinum contacts on the Gate group has described the success achieved in an
terminal recording sites [45]. Furthermore, conformal Par- additional pair of patients. In the second study, the authors
ylene-C coatings have been applied through chemical vapor demonstrated that patients with long-standing tetraplegia were
deposition to provide additional insulation to the electrode able to produce useful movements of a prosthetic arm with the
tines and protect the underlying dielectrics from dissolution UEA based system [6]. For example, one patient was able to
[46]. Electrical isolation of individual channels has been use her thoughts to control a robotic arm, reach and grasp a
further enhanced by incorporating a glass dielectric between bottle of coffee, bring it towards her mouth to drink, and then
individual bond pads on the backside of the wafer [47]. return the bottle to the table for the rst time in 14 years [6].
Beyond the initial design of the UEA with a 10 10 array Excitingly, results were achievable ve years after implanta-
of 1.5 mm tines, developers have also shown that a number of tion, and the systems were still functional at the time of
alternative structures can be created. For example, the Utah publication. While the successes of animal studies and the
slanted electrode array was developed to facilitate stimulation BrainGate project in particular are quite promising, improving
and recording at various tissue depths [48]. Recent studies recording consistency is still a primary focus for the
have also shown that the UEAs structure can be further UEA [59].

5
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

Figure 4. (A) Cellular level failure modes. Cellular level failure modes act (1) disrupting the neuronal signal source, (2) impeding charge
transport through the extracellular space or (3) disrupting charge transfer at the electrode recording site. (B) Higher, tissue level failure modes
that have not been as widely described or considered in the literature. Asterisks denote failure modes with an underlying inammatory
component. As nearly all failure modes have an underlying inammatory component, strategies that reduce neuro-inammation will be
critical to improving the biocompatibility and function of intracortical microelectrodes.

2.3. Neurotrophic cone electrode scheme of the cone electrode. However, Kennedys approach
does highlight the potential of bioactive strategies for
Diverging from many in the eld who believed that single
improving neural interfacing (Discussed further in
units were the key to BMI success, Kennedy et al took a new section 4.5).
multi-unit approach towards BMIs and developed the neu-
rotrophic cone electrode (gure 3(D)) [60]. Kennedys elec-
trode was built around a glass cone with a Teon-insulated
gold wire. This design is very similar to pipette electrodes 3. Challenges to obtaining consistent, high-quality
used in acute electrophysiology experiments prior to the neural recordings
development of microwire devices. Building on the work of
David and Aguayo, who had demonstrated endogenous Despite the substantial success that has been demonstrated
innervation of peripheral nerve grafts, Kennedy placed a using intracortical microelectrodes in neural interface appli-
segment of sciatic nerve into the glass cone [61]. Similar to cations, many studies have shown chronic cortical recording
the innervation of the nerve graft, implantation of the cone to be inconsistent in a variety of species and with multiple
electrode into rat cortex elicited the ingrowth of neuronal electrode types. As early as 1974, Burns et al showed a
processes into the glass cone. Using his novel approach, progressive decline in unit recordings in cat cerebral cortex
Kennedy was able to record neural signals for up to 11 after implantation, with only 8% of the electrodes functioning
months following implantation in the rat cortex and up to 15 after ve months [31]. Forty years later, recording instability
months in the monkey cortex [60, 62]. is still a commonly documented problem. For example, Liu
Beyond successful animal experiments, Kennedy imple- et al reported that implanted electrodes are unstable during
mented the neurotrophic cone electrodes as the earliest plat- the acute phases of tissue remodeling, and thereafter experi-
form for successful human clinical studies of BMIs [810]. In ence a continual decrease in recording ability over the ensuing
their seminal study, Kennedy and colleagues implanted neu- months [16, 17]. Additionally, recently Ludwig et al and
rotrophic cone electrodes into the cortex of three patients and Freire et al have both described uctuations in recording
consciously modulated neural signals were used to drive the stability that agree well with previous ndings [43, 63].
movement of a computer cursor. A number of failure modes likely inuence chronic
While the data obtained using neurotrophic cone elec- recording stability and quality including: (1) direct mechan-
trodes were quite promising, widespread adoption has been ical damage of the electrode; (2) corrosion of electrical con-
limited, possibly due to the fragility and boutique fabrication tacts; (3) degradation of passivation layers and insulating

6
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

Figure 5. Pre-implant (left) and post-explant (right) SEM images from acute (top, 1 day), recovery (middle, 16 days) and chronic (bottom,
187 days). The images show the progression of corrosion of the tungsten metal over the increased implantation time.

coatings; and (4) the neuro-inammatory response that the study were made from materials that are commonly con-
brain mounts against chronically implanted devices [14, 34]. sidered brittle, such as silicon or ceramics, only one failure of
Figure 4 illustrates how each failure modes could impact the a penetrating shank was described [14]. Similar occurrences
compound circuit describing how microelectrodes extract of mechanical failures away from the penetrating wires,
electrical signals from neural tissue. Traditionally, micro- shanks and tines of traditional microelectrode recording sys-
electrode failure modes have largely been studied indepen- tems have independently been described in recent reports
dently from one another. However, there is likely [34, 35, 64]. Thus, improvements in the mechanical stability
considerable interplay among the various modes making it of the entire recording system, and not just the intracortical
difcult to attribute failure to a single mechanism. microelectrode should be further pursued.

3.1. Direct mechanical damage 3.2. Corrosion of electrical contacts

Several studies have indicated that mechanical damage during While descriptions concerning electrode corrosion have been
or following insertion can lead to microelectrode failure. For reported for stimulating electrodes, relatively few have been
example, Ward et al experienced mechanical failure in seven provided for recording microelectrodes. However, even under
of nineteen devices, regardless of the type of electrode [14]. non-stimulating conditions (i.e. under conditions in which no
Interestingly, while a number of the electrodes used in Wards electrochemical reactions should occur via an externally

7
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

applied electric eld) some materials used in recording 3.3. Degradation of passivation layers and insulating coatings
microelectrodes likely experience faradaic charge transfer and
Similar to electrode recording sites, the passivation layers and
corrosion over time [65]. In fact, structural changes at the
insulating coatings commonly incorporated into microelec-
electrode-recording sites have been observed to progress with
trodes may degrade over time. Signicant degradation of an
time after electrode insertion for tungsten microwires
electrodes insulating or passivation layers could reduce an
(gure 5) [34], while little corrosion was reported for Pt/Ir
electrodes ability to detect local ionic signals (gure 4) [32].
electrodes [35]. The susceptibility for silicon or glass passivation layers
The rate of corrosion is likely environment and material to degrade in vivo has been shown in a number of studies. For
specic. For example, Patrick et al have shown that bare example, Wang et al observed that the corrosion of silicon
tungsten and gold-plated tungsten wires corrode readily in begins in as little as ten days from implantation into the rat
phosphate-buffered saline even under non-stimulating con- brain [72]. Furthermore, Hmmerle et al have shown that
ditions [66]. Patrick and colleagues also found that tungsten while silicon oxide is stable for over 21 months in saline
corrosion was increased in the presence of oxidative species solution, signicant degradation occurs after implantation in a
in vitro. The critical role of oxidative species in electrode subretinal model [73]. Beyond the removal of the as-fabri-
corrosion provides an important link to the brains inam- cated silicon oxide surface layer within 12 months of
matory response, since reactive oxygen species are actively implantation, Hmmerle and colleagues also observed pro-
produced surrounding implanted microelectrodes gressive corrosion of their underlying silicon substrate [73].
(section 3.4.4). In vivo corrosion rates were reported to be as Degradation is not exclusive to silicon and silicon oxide
high as 100 m/year, indicating that corrosion is a likely layers, as Maloney and colleagues have shown similar
contributor to at least tungsten-based electrode failure. degradation rates (1 m/year) in triple layered silicon oxide/
By contrast, titanium forms a natural passivation layer, nitride stacks [74].
and is more resistant to oxidative corrosion. McCarthy et al The degradation of traditional passivation layers is not
have begun to develop titanium-based MI-style microelec- surprising as they were originally designed to serve as
trodes that may perform better in the oxidative environment dielectrics in dry, non-corrosive environments that are shiel-
that develops surrounding implanted microelectrodes [67]. ded from mechanical stresses. A number of mechanisms may
Furthermore, platinum wires are not only stable in saline/ inuence the degradation of traditional passivation materials.
H2O2 environments, but actively convert hydrogen peroxide These mechanisms include mechanical stress, lm defects, as
species to water [66], mirroring the catalytic activity of nat- well as chemical or electrochemical reactions. Further infor-
ural anti-oxidative enzymes [68]. Potter et al have previously mation regarding these mechanisms is presented by Scmitt
demonstrated that reactive oxygen species accumulation may et al [75].
facilitate neurodegeneration at the microelectrode surface To overcome the limitations of traditional passivation
[69, 70]. Therefore, the ability for platinum materials to materials, Cogan et al developed an amorphous silicon car-
reduce the concentration of oxidative species could explain bide (a-SiC) dielectric lm for microelectrodes [76]. Degra-
the improved performance of platinum-based microelectrodes dation testing showed that the a-SiC had a dissolution rate of
0.1 nm h1 at 90 C (1/20th that of silicon nitride) and no
in neural interface applications.
Beyond impacting recording site stability, electrode measurable dissolution at 37 C.
Due to the chemical vulnerability of common dielectric
corrosion can also generate toxic species. For example, the
passivation layers, further encapsulation of microelectrodes
generation of toxic species from Ag/AgCl electrodes has been
with insulating polymers has become common practice
well documented [71]. In addition, Patrick et al reported that
[32, 46, 77]. While no direct comparison has been made,
the primary species generated by tungsten corrosion were
historically there has been a signicant trend towards
tungstic ions, which are known to be moderately toxic [66].
improved recording longevity when silicon microelectrodes
Production of toxic species could be another important con- were coated with polymeric insulators. However, in vivo
nection linking biotic and abiotic failure modes. rodent studies have shown no difference in the neuro-
Information regarding the corrosion rates of many com- inammatory response of Parylene-coated Michigan-style
mon microelectrode materials under non-stimulating condi- microelectrodes compared to uncoated devices [78]. There-
tions is not readily available. In view of the above-mentioned fore, it is likely that any increased recording longevity is not
ndings further analysis of the corrosion of common elec- due to a signicant reduction in the neuro-inammatory
trode materials would be valuable. Furthermore, the impact of response on account of reduced degradation of the Si-based
corrosive species generated from the breakdown of many devices. Nevertheless, as many descriptions of the loss of
electrode materials is not well understood and deserves recording quality come from electrodes with polymer-based
additional study. When conducting corrosion analysis it is insulating coatings, it is clear that improving insulation alone
important to mimic the in vivo environment, including the is not a silver bullet and that other sources of instability, such
presence of oxidative species and acidic pH. Additionally, the as the neuro-inammatory response, are still at play.
impact of corrosion products should be considered when While insulating polymer coatings have signicantly
examining and comparing the biocompatibility of chronically improved recording systems, a limited number of studies have
implanted microelectrodes made from different materials. described degradation of common insulators used on

8
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

Figure 6. Insulation deterioration post-explant SEM. Post-explant SEM of individual microwire to indicate deterioration in electrode
insulation for parylene-C coated Pt/Ir microwires. The deterioration occurs in the form of delamination and cracks. While the insulation
deterioration varies among microwires even with the same array, Prasad et al observed it to be present in all the wires across animals for all
implant durations (7 days6 months).

microelectrodes. For example, Prasad et al showed evidence neuro-inammatory response that develops following micro-
that polyimide insulation on tungsten microwires was peeled electrode implantation in the brain, and developing strategies
away from the recording site, and had signs of cracking as to reduce its impact are critical to achieving the promise of
early as 42 days after implantation (gure 6) [34]. Insulation BMIs and to enable longer recording durations for basic
damage was particularly common in chronic implants, where science experiments.
seven of twelve electrodes implanted showed damage. Over 100 studies have described stereotypic features of
However, it is unclear if changes to the insulation were a the brains response to microelectrodes that occur irrespective
result of direct damage of the polyimide or a result of cor- of the type of implant, method of sterilization, species studied,
rosion of the underlying tungsten. or implantation method. From this rich body of literature, it
As with recording site corrosion, further analysis of the has become increasing clear that the brains response consists
degradation of common passivation layers and insulting
of an interconnected web of molecular and cellular compo-
coatings is needed. While in vitro experiments can facilitate
nents. The ultimate result of which is the continuous perpe-
higher throughput analysis, it is critical to also investigate
tuation of the response, and the prevention of microelectrode
degradation using in vivo models to more accurately under-
stand the contribution of the neuro-inammatory response integration into the surrounding tissue.
(section 3.4). With respect to the molecular and cellular components,
several theories have been presented to explain how indivi-
dual components of the response might adversely impact
3.4. The neuro-inflammatory response recording quality. However, it is highly likely that multiple
There is increasing consensus that the neuro-inammatory aspects of the response are at play simultaneously. Thus,
response to intracortical microelectrodes is a primary hurdle further study into the details of the neuro-inammatory
preventing microelectrode-driven BMIs from reaching their response and the development of more comprehensive miti-
full potential. Therefore, improving the understanding of the gation strategies are indicated.

9
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

Figure 7. Electrode implantation results in localized pro-inammatory cellular and biochemical events. Early after implantation, activated
microglia begin to attach to the surface of the electrode and locally release pro-inammatory factors. Glia cell adhesion is followed by
astrocytic encapsulation along the entire shaft of the electrode (formation of the glial scar). These events, as well as localized hemorrhaging,
have been shown to be correlated with neurodegeneration at the interface. Representative IHC images of the dominant cell types are shown
left. Scale = 100 mm.

3.4.1. Initial injury and early wound healing events. Due to propagation of the neuro-inammatory response may be due
the dense and, in many cases highly vascularized nature of to perpetual motion-induced damage at the interface of
nervous tissue, microelectrode implantation inevitably causes traditional microelectrodes. The base materials used in
vascular and cellular injury [79, 80]. Following the initial traditional microelectrodes are signicantly stiffer than
iatrogenic injury, several acute cascades and processes are cortical tissue. Therefore, starting with Goldstein and
initiated to induce wound closure and promote tissue Salcmans work in 1973, a number of groups have
remodeling. Directly after injury, the coagulation cascade is suggested that motion of the brain with respect to the
initiated and forms a provisional matrix to restore vascular microelectrode may induce damage to the surrounding tissue
integrity [81, 82]. Simultaneously the complement system is [8895].
also initiated. The complement cascade may directly induce In silico studies support the hypothesis that even
apoptosis in nearby cells or invading pathogens through the micromotion of the brain relative to a stiff microelectrode
membrane attack complex. Additionally, complement assists
could induce strain on the surrounding tissue [89]. However,
in recruiting inammatory cells to the site of injury through
to date, limited work has been performed to quantify
the alternative arm of the cascade [83].
microelectrode-induced strain on the surrounding tissue.
Much is known about early wound healing events and
their roles in injury and other device implantation models Recently, the Muthsuwamy lab developed a method to
[84, 85]. However, comparably few studies have explored the measure the mechanical properties of the biotic component
early wound healing events after implantation of intracortical of the brain-electrode interface, surrounding non-compliant
microelectrodes [79, 86]. The majority of what is known stainless steel microelectrode implants [96]. Specically, they
about the brains response to implanted microelectrodes have found that the estimated shear and elastic modulus in the
comes from end-point histological studies focused on later surrounding brain tissue uctuates and evolves over time.
time points that range from 124 weeks post-implantation. Ongoing studies are investigating the effects of implant
Figure 7 provides images and an illustration of the stereotypic stiffness on the strain placed on the cortical tissue adjacent the
response of the brain to chronically implanted microelec- implant. Determining whether implanted microelectrodes
trodes [87]. induce sufcient strain to affect neural and inammatory
cells is a critical gap in the eld. Additionally, quantifying
3.4.2. Motion induced injury at later time points. in vivo or ex vivo strain data would be extremely useful in the
Microelectrode-induced injury events are likely not limited creation of improved predictive models for driving future
to the initial iatrogenic trauma. It is widely accepted that microelectrode designs (discussed further in section 4.1).

10
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

Figure 8. Self-perpetuating neuroinammatory pathways. After intracortical microelectrode implantation, the damage of localized
vasculature can result in two mechanistic paradigms at the interface of the implanted device. The order of events in either cycle is unknown,
as any one event can perpetuate the subsequent step. Left: extravasated blood-derived proteins from damaged vasculature become adsorbed
onto the surface of the implanted device and dispersed throughout the local tissue environment. Blood-derived proteins then activate
inammatory cells and stimulate the release of pro-inammatory and cytotoxic soluble factors. Release of pro-inammatory molecules
facilitates self-perpetuation of both blood-brain barrier (BBB) breakdown and persistent neuro-inammation around the implant. Right:
release of pro-inammatory and cytotoxic soluble factors can directly and indirectly lead to neuronal apoptosis. Cellular debris from
apoptotic cells can further stimulate microglia activation and initiate further BBB instability. Therefore, the neuro-inammatory response to
intracortical microelectrodes will last as long as the implant is implanted in the tissue, and interacting with cells or proteins.

Despite the infancy of strain quantication, recent in vivo serum/plasma proteins, phagocytizing damaged or dead cells
studies have shown that microelectrodes made from materials [110], and in clearing residual cell debris [111]. Following
that more closely match the brains mechanical properties phagocytosis, microglia and macrophages are known to enter
may elicit a reduced neuro-inammatory response [94, 97 the lymphatic system and act as antigen presenting cells in a
99]. However, the precise mechanism underlying how variety of diseases and pathological states [112116]. As
mechanical mismatch facilitates the neuro-inammatory suggested by Skousen et al cell trafcking to and from the
response is still being debated. Nevertheless, the hypothesis implant interface provides a potentially persistent stimulus for
that mechanical mismatch between the microelectrode and the neuro-inammatory response via extravasated brinogen,
brain tissue contributes to the neuro-inammatory response bronectin, complement factors and other blood products
has resulted in the development and use of compliant [117]. Following extravasation, blood products adsorb to the
materials (section 4.1) to replace the stiffer silicon, ceramic microelectrode surface and perpetuate inammatory cell
and metal substrates used in traditional microelectrodes activation through receptor-mediated pathways such as toll-
[70, 99105]. It is, however, important to note that the like receptor (TLR)-mediated pathways (gure 8)
inuence, which the compliant behavior exerts on the quality [69, 118, 119].
of neural recordings of microelectrodes fabricated from such
materials, has yet to be described.
3.4.4. The critical role of pro-inflammatory and cytotoxic
soluble factors. Multiple studies have shown that activated
3.4.3. Microglia/macrophage response to intracortical microglia and macrophages release a plethora of pro-
microelectrodes. Similar to the response in the rest of the inammatory/cytotoxic soluble factors that can damage
body [84, 85], a key feature of the brains response to healthy bystander cells and the surrounding tissue [120
chronically implanted devices is persistent inammation at 124]. Furthermore, as described in sections 3.2 and 3.3, a
the biotic-abiotic interface [99, 106]. Persistent inammation number of soluble factors may also be involved in recording
involves activation of both resident microglia and the site corrosion and degradation of insulating coatings.
perpetual recruitment of blood-born macrophages It should be noted that astrocytes and other cells are also
[15, 106109]. known to secrete pro-inammatory and cytotoxic soluble
Both microglia and macrophages play a primary role in factors. However in general these cells are believed to
responding to invading pathogens, recognizing extravasated produce signicantly less pro-inammatory and cytotoxic

11
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

soluble factors than activated macrophages and microglia Following injury, astrocytes increase the number and size
[120]. Furthermore, comparative studies have indicated that of their cellular processes, and are primarily identied by
macrophages/microglia, and not astrocytes, are the key source increased staining for glia brillary acid protein (GFAP), an
of pro-inammatory and cytotoxic soluble factors that astrocyte-specic intermediate lament. Hypertrophic astro-
mediate neurodegeneration in a number of disease states cytes are believed to play a similar role to that of reactive
[125127]. broblasts in the foreign body response in other tissue
Of the plethora of soluble factors within a macrophages compartments [151]. Specically, astrocytes create a dense
available palette, previous work from Biran et al has shown scar-like layer that limits volume transmission [152].
that adherent cells retrieved from explanted devices secrete Many neuro-inammatory studies have hypothesized that
both tumor necrosis factor-alpha (TNF-) and monocyte the astrocytic diffusion barrier may play a benecial role in
chemotactic protein-1 (MCP-1) [107]. TNF- can have direct restricting the impact of macrophage-secreted factors on the
toxic effects on neurons and oligodendrocytes, while MCP-1 surrounding tissue, as well as mechanically shielding the
is a chemokine involved in opening the blood-brain barrier surrounding tissue from micromotion induced strains sur-
(BBB) and recruiting new macrophages to sites of injury and rounding the historically stiff microelectrodes [94, 98, 153
inammation [120, 121, 124, 128134]. 157]. However, astrogliosis or other forms of brotic
Several recent studies have also provided further support encapsulation, may also increase the tissues impedance to
for the predominant role of macrophage-released soluble small ion transport, potentially limiting recording function as
factors on recording function and the neuro-inammatory suggested by Porter et al and later by Schmidt and colleagues
response. For example, Karumbaiah et al have shown that [30, 31, 158].
gene expression for various pro-inammatory soluble factors,
specically IL-1,6 and 17 as well as TNF-, is up-regulated in
tissue surrounding poorly performing microelectrodes 3.4.6. Neuronal loss at the electrode-tissue interface.
[135, 136]. In addition, Potter et al have shown that Associated with the regions of inammation and reactive
accumulation of reactive oxygen species surrounding gliosis, studies have described a decrease in the local nerve
implanted microelectrodes may impact neuronal viabi-
ber and neuronal cell body densities surrounding implanted
lity [69].
devices [107, 109, 143, 145, 159, 160]. While a signicant
Skousen et al have suggested that macrophage-secreted
number of neurons remain within the recording range, the
soluble factors may be critical in both propagating as well as
overall decrease in neuronal density (approximately 4060%
shaping the response to traditional microelectrode designs
in most studies) indicates that the environment may no longer
[117, 137]. Specically, it was shown that predicted
be ideal for promoting neuronal health and function. Clearly
distributions for macrophage-released soluble factors corre-
any compromise of the target neuronal population may
late well with the shape and structure of the neuro-
inuence device function.
inammatory response to traditional microelectrode designs
It has become well established that chronic inammation
regardless of device compliance. These observations reveal
and neuronal loss are associated with the persistent presence
that presented architecture is a major contributing factor to the
overall neuro-inammatory impact on surrounding neural of the implant, and are not solely the result of iatrogenic
tissue. Taken together, the studies referred to above indicate injury. In their seminal paper, Biran et al compared various
the utility of strategies that reduce the concentration of pro- markers of neuro-inammation in chronically implanted
inammatory and cytotoxic soluble factors to improve animals to animals that received only a stab wound injury
recording function. To have maximal impact, as suggested [107]. The authors found that chronic neuro-inammation and
by Skousen et al strategies should focus on (1) limiting the neuronal loss does not accompany stab wound injuries made
local number of activated macrophages at the device inter- with microelectrodes identical to those left in place.
face, (2) reducing the degree of inammatory cellular Birans ndings have been conrmed and expanded upon
activation, and (3) directly antagonizing the accumulation of by several groups, including McConnell et al and Potter et al
pro-inammatory and cytotoxic soluble factors themselves [109, 161]. Both of these studies also observed that
[117, 137]. microelectrode implantation within the cerebral cortex may
trigger a multiphasic neuro-inammatory and neurodegen-
erative response. However, it should be noted that the time-
3.4.5. Astrogliosis and fibrotic encapsulation. Surrounding course of the neuro-inammatory response is still being
the inammatory core, a region consisting of hypertrophic debated due to discrepancies between, and even within,
astrocytes as well as inltrating broblasts and meningeal different laboratories [78, 87, 109, 161, 162]. Furthermore,
cells has also been observed [69, 70, 107, 109, 117, 138 several studies have failed to establish a direct correlation
146]. In healthy brain tissue, astrocytes regulate the local between neuro-inammation and recording quality. This
microenvironment. Astrocytes sequester a number of disconnect may be due to the complex interconnectedness
neurotransmitters and ions, while also maintaining the BBB of microelectrode failure modes or, as discussed in section
that isolates the cellular and ionic milieu of the brain from that 3.4.10, nonlinear relationships between electrode function and
of the supporting vasculature [147150]. the neuro-inammatory response [160].

12
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

3.4.7. Local ECM changes. Associated with the region of dysfunction should be of key focus in the eld to improve
astrocyte hypertrophy and reduced neuronal density, a microelectrode biocompatibility.
number of studies have described changes in local ECM.
Injury-induced changes in ECM have been widely reported
following traumatic brain injuries and in many neurological 3.4.9. Connecting the neuro-inflammatory response and
diseases [163165]. Following microelectrode implantation, recording quality. While a number of potential mechanisms
Zhong et al have described an up-regulation of chondroitin have been presented to describe how the brains response may
sulfate proteoglycans (CSPGs) at the biotic/abiotic interface impact recording function, the direct connection remains
[166]. CSPGs are generally considered neuro-inhibitory unclear. However, there is increasing evidence indicating that
[167173]. Therefore, as with successful repair and the neuro-inammatory response may be a primary hurdle to
regeneration following spinal cord injury, it is likely that consistently obtaining high quality recordings. For example,
the altered ECM impedes successful neuronal regeneration in in 2007, Rennekar et al examined whether systemic anti-
tissue adjacent to the implanted microelectrode. Furthermore, inammatory administration could improve recording
changes in the ECM density could also further limit volume performance [179]. The drug used in Rennekars study,
transmission surrounding implanted microelectrodes. Minocycline, is a tetracycline antibiotic known to shift
macrophages and microglia away from a pro-inammatory
(M1) phenotype [180, 181]. Electrodes in rats that received
3.4.8. Blood-brain barrier dysfunction. New observations oral minocycline treatment showed a signicant improvement
from Trescos group have opened other potential in both signal to noise ratio (SNR), and the number of
explanations as to how the neuro-inammatory response to channels that recorded stimulus-driven neural activity.
an implanted microelectrode could inuence recording. As Unfortunately, while likely implicating inammation, little
observed in many neurodegenerative disorders, it was found histological examination was performed to link particular
that local blood BBB integrity is compromised in the tissue cells types (such as macrophages) or reactive species to
immediately surrounding implanted microwires and recording function.
Michigan-style microelectrodes [78, 117]. These ndings Delivery of another anti-inammatory drug, dexametha-
suggest that an altered local ionic milieu could inuence sone, has been shown to reduce the inammatory response to
recording instability [174176]. Recently Potter et al studied inserted microelectrodes [182185]. However, studies
the progression of BBB integrity over time and found that describing the impact of dexamethasone administration on
similar to neuro-inammatory diseases such as multiple recording performance have, to our knowledge, not been
sclerosis, BBB dysfunction is highly dynamic performed. Interestingly, studies that delivered dexametha-
[87, 109, 177, 178]. sone locally around an implanted microelectrode showed no
Additional recent data further highlights the potential role signicant impact on the reactivity at later time points. This
of BBB dysfunction in connection with poor recording apparent discrepancy is likely due to exhaustion of the drug
performance. Findings from the Bellamkonda group with source. Therefore a chronic anti-inammatory regimen or
Michigan-style and microwire electrodes [162] have shown more permanent solution will be needed to regulate the neuro-
that recording performance correlates with markers of BBB inammatory response through the lifetime of the implanted
dysfunction such as extravasated immunoglobulin G (IgG) or microelectrode.
labeled albumin. Unfortunately, even if it was possible to continually
Beyond directly impacting neurons and recording func- deliver dexamethasone or minocycline, this would not be an
tion, it is important to emphasize that inltrating blood adequate long-term solution, as chronic use of either drug can
products also serve as persistent stimuli for perpetuating result in immune system impairment, decreased renal
neuro-inammation and vice-versa. For example, extrava- function, vertigo, bone discoloration/loss, fatal colitis, and
sated brinogen, plasma soluble bronectin, complement intracranial hypertension [186189]. Therefore, while the use
factors, and other blood products have been shown to be of Minocycline and dexamethasone provides a mechanistic
potent mediators of macrophage and microglial activation understanding regarding how inammation may impact
[118]. Following extravasation, blood-products are involved electrode performance, better-tolerated pharmaceutical and
in inammatory cell activation through TLR, CD14 (i.e. materials-based approaches need to be developed.
glycosylphosphatidylinositol-anchored membrane glycopro- To further elucidate the role of inammation on
tein), and other receptor-mediated pathways [119, 120]. A recording function, Tyler and colleagues examined whether
variety of blood components are likely present at the exacerbation of the inammatory response would reduce
microelectrode/tissue interface throughout the lifetime of the recording performance [190]. To answer this question,
implant as a combination of phenomena. This combination Tylers group compared the recording quality of Michigan-
includes (1) the initial damage of microelectrode implantation style devices in control animals to that from animals that were
into the cortex, (2) motion-induced damage at later time- administered the bacterial endotoxin lipopolysaccharide
points, (3) macrophage/microglia trafcking at both early and (LPS). LPS is a known stimulus for driving macrophages
chronic time points and (4) persistent pro-inammatory and microglia to a pro-inammatory state through TLR
signaling. Thus, developing a combination of methods to pathways [191193]. Microelectrodes in rats that received
break the self-perpetuating cycle of inammation and BBB LPS had signicantly lower SNRs and number of recorded

13
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

units compared to saline-only control animals [98]. Thus, approaches to mitigating microelectrode failure and/or poor
Tyler and colleagues results further implicate neuro-inam- tissue integration have received considerable attention.
mation as a primary biological mediator of recording
performance.
While Tylers work demonstrated that large-scale
exacerbation of neuroinammation impacts recording quality, 4. Material strategies for improving microelectrode
more recent evidence from Ravikumar et al suggests that even biocompatibility and recording performance
small-scale shifts in neuroinammation can dramatically
impact the local tissue [160]. Specically, Ravikumar et al In the last decade, various materials-based strategies have
examined the brain tissue response to sterilized silicon been investigated with the objective of minimizing the neuro-
microelectrodes with varied amounts of low-level endotoxin inammatory response and enabling high-delity neural
contamination. Histological evaluation at two weeks showed interfacing over clinically relevant timeframes. In all cases,
a direct correlation between microglia/macrophage activation developers have sought to address one or a set of the lim-
and residual endotoxin levels. By contrast, astrogliosis, itations discussed in sections 2 and 3. Throughout the
neuronal loss, and blood brain barrier dysfunction demon- remainder of the paper we will review the primary approaches
strated a threshold-dependent response to bacterial endotoxins to develop the next generation of intracortical microelectrodes
and macrophage/microglia activation. A threshold-dependent including:
response demonstrates that even subtle changes in the neuro-
inammatory environment over time could underlie observa- Minimizing motion-induced injury using compliant
tions of recording inconsistency as the environment shifts microelectrode substrates.
back and forth beyond a critical inammatory threshold. Limiting surgical trauma and/or inammatory cell
In their study, Ravikumar et al also indicated that in the accumulation by manipulating microelectrode
108 published microelectrode studies that they reviewed, a architecture.
wide range of sterilization methods were used. Quite Preventing protein and inammatory cell adhesion
strikingly, different distributions of sterilization methods through non-fouling surface coatings.
are seen in studies that utilize functional and non-functional Manipulating inammatory cell phenotype through use
electrodes, respectively. It appears to be rather concerning of surface topography.
that in as many as 20% of the studies, no details on how the Directing tissue integration at the microelectrode-tissue
implants were cleaned and sterilized were reported. This interface using bioactive materials.
raises the question, to what extent our understanding of Reducing the concentration or impact of inammatory
electrode performance is confounded by inammation soluble factors through the use of passive and active
triggered by incomplete sterilization of the implanted antagonists.
devices. Improving the electrical performance of intracortical
microelectrodes using conducting polymers and
3.5. Summary of the challenges to achieving consistent, high- nanomaterials.
quality neural recordings
Each subsection is concluded with our interpretation on
The above section summarizes the many mechanisms that can the strengths/limitations and questions that must be addressed
spatially and temporally mediate microelectrode failure. to enable consistent, high-quality long-term neural recordings.
These failure modes include, but are not limited to, (1) direct There are a number of important facts to consider when
mechanical damage; (2) corrosion of electrical contacts; (3) comparing and analyzing the impact of material-based
degradation of passivation layers and insulating coatings; and approaches for improving microelectrode function. First,
(4) the neuro-inammatory response that the brain mounts isolating the impact of a given strategy to one specic vari-
against chronically implanted devices. Figure 4 highlights able that could inuence the neuro-inammatory response is
how each of these various failure modes may impact the difcult at best. For example, as will be discussed in
overall neural interface circuit. section 4.1, a major strategy in the eld for reducing the
Due to the variety of failure modes and the high level of neuro-inammatory response is the creation of compliant,
interplay involved, it is increasingly evident that combina- polymer-based microelectrodes that better match the
torial strategies may be needed to obtain consistent, high mechanical properties of the surrounding tissue. However,
quality neural recordings. While a number of anti-inamma- many of the polymers used to create compliant microelec-
tory drugs have been investigated and have provided infor- trodes absorb a signicant degree of water and are likely
mation on whether/how neuro-inammation may impact permeable to small molecules. Thus the innate permeability of
electrode performance, better-tolerated, longer-lasting these complaint materials adds the possibility that ndings
approaches need to be further developed. from these studies have been inuenced by improved clear-
Furthermore, one could argue that all four of the ance of pro-inammatory and cytotoxic soluble factors
described failure modes could be mitigated through the (section 4.6.1). Therefore, to elucidate the overall design
appropriate choice and/or development of more appropriate space available for microelectrode designers, further studies
materials. Therefore, it is not surprising that material-based should be conducted to isolate the impact of individual design

14
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

variables as well as to identify possible interactions or


emergent phenomena.
Equally as important when analyzing ndings from stu-
dies that have examined new strategies for reducing the
neuro-inammatory response, one must critically assess the
role that tissue processing and other techniques may have on
reported results. For example, in almost all cases the
implanted microelectrodes are removed from tissue prior to
analysis. Microelectrode removal may disrupt the tissue
interface and inuence data interpretation, especially for
coatings that impact cell attachment [78, 107, 108, 194].
Different groups also use a variety of diverse markers to
describe related cellular and molecular features of the neuro-
inammatory response. An example of this is the use of pan-
Figure 9. Forces acting on the intracortical microelectrodes upon
macrophage markers such as OX-42 and IBA-1versus mar-
penetration.
kers for activated macrophages such as CD-68. There are also
large to subtle differences in the methods used to image,
quantify, and statistically compare histological results that can force, frictional forces excreted on any presented surface, and
lead to differences in interpretation. Common differences a compressive clamping force (gure 9). The summation of
include the use of confocal versus traditional microscopy, the these three forces is commonly referred to as the total
use of boutique quantication packages, as well as dis- insertion force (IF). The IF for traditional microelectrodes
crepancies in dening what makes for an independent mea- ranges from 5001000 N depending on the shape of
surement/observation. Therefore, efforts to improve the electrodes tip and dimensions of the electrodes shank
quality and consistency of methods across and even within [196, 197].
groups would be useful for improving intra-study To avoid buckling during insertion, the IF must be lower
comparisons. than the critical loading force (CLF) for a given design.
Therefore, both the IF and CLF should be considered when
4.1. Mechanically compliant intracortical microelectrodes creating any new microelectrode design. Given similar
dimensions to traditional microwire or planar silicon micro-
As discussed above, traditional microelectrodes have been
electrodes, compliant devices should have an IF > 1000 N to
composed of extremely stiff materials such as metals or sili-
avoid buckling [198201]. To satisfy this design criterion
con. The high stiffness facilitates microelectrode implantation
many compliant microelectrodes were designed with larger
into the cortical tissue [79]. Unfortunately, a number of
cross sectional areas than traditional microwires or silicon
groups have hypothesized that increased stiffness may
microelectrodes. As will be discussed in sections 4.1.3 and
adversely impact neuronal tissue through a number of
mechanisms [8895]. First, in vitro evidence indicates that 4.1.4, a number of groups have also moved to using insertion
substrate stiffness, even in a static culture environment, may aides or in situ softening materials in order to insert smaller or
adversely impact neuronal and glial cell types. However, a more compliant devices.
number of in vivo studies looking at either stiff materials or As stated, there is substantial belief in the eld that
those coated with compliant polymers have indicated that mechanical differences between the brain and traditional
haptic-mediated mechanotransduction may not play as sig- microelectrodes induce adverse strain in the surrounding
nicant a role as initially thought (see sections 4.1.3, 4.2.1, tissue during normal respiratory and circulatory pulsations.
4.3 and 4.6.1) [117, 195]. The second, and perhaps more Unfortunately, while in vivo studies have described insertion
predominant hypothesis, is that mechanical differences and extraction mechanics [96, 157], only one study has been
between the brain and microelectrodes induce adverse strains performed to directly quantify microelectrode-induced strain
in the surrounding tissue during regular brain micromotion over the indwelling period [96]. Determining whether
[95, 138, 196]. Therefore, compliant materials that have implanted microelectrodes induce sufcient strain to
mechanical properties closer to that of brain tissue have adversely impact neural and inammatory cells is a critical
received extensive attention towards improving microelec- gap in the eld.
trode integration within the surrounding tissue. Aside from the one in vivo study, several computational
models have been developed to estimate this elusive
4.1.1. Mechanical factors impacting intracortical parameter [89, 202204]. Such modeling studies support the
microelectrode biocompatibility. When manipulating hypothesis that mechanical mismatch between the implanted
microelectrode compliance it is important to further discuss microelectrode and surrounding brain tissue could lead to
a number of mechanical factors that may impact adverse strains and stresses being generated during normal
microelectrodes or the surrounding tissue during insertion brain micromotion [95]. Furthermore, while the majority of
and throughout the indwelling period. During insertion, three the eld has focused on electrode stiffness, the models
primary forces act on the microelectrode, namely: an axial tip suggest that tethering scheme and the degree of tissue

15
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

adherence are additional variables that can be manipulated to inammatory response or improve chronic recording
reduce microelectrode-induced strains. performance.
Of these additional variables, perhaps the best character- There are still a number of inherent limitations to these
ized in vivo is the impact of device tethering. Starting with off-the-shelf materials. For example, in nearly all cases their
Biran et al multiple studies have indicated that tethering stiffness is still at least three orders of magnitude higher than
devices to the skull exacerbates the neuroinammatory brain tissue. While modeling studies have indicated that
response [108, 145, 205]. While these ndings support the reducing microelectrode stiffness to the MPa range will limit
hypothesis that mechanical mismatch plays a role in tissue strain, it is unclear exactly how soft a material must be
propagating the neuroinammatory response, there are to achieve reductions in the neuroinammatory response or
alternative explanations and contradictory evidence in the improvements in recording function.
eld. For example, one alternative explanation is that As with many of the early metals used for microelec-
anchoring exacerbates inammation by facilitating meningeal trodes, toxicity has been a concern with a number of off-the-
broblast migration into the brain. This is supported by recent shelf compliant polymers. Toxicity may be caused from the
ndings indicating that sub-meningeal implantation reduces polymer itself or from leaching of residual solvents,
the neuro-inammatory response [206]. Furthermore, the only plasticizers, or degradation products. For example, Vernekar
in vivo tethering study driven directly by a computational showed that thick untreated SU-8 substrates are not
model failed to conrm the predicted impact of various compatible with primary neuronal culture as less than 10%
tethering schemes [207]. of primary neurons survived when cultured on SU-8
Therefore, while mechanical models have expanded our substrates [216]. The authors suggested that the poor
understanding of the impact of a number of biotic and abiotic cytocompatibility of SU-8 was due to leachables as neuronal
parameters, there is still further work that should be pursued. survival increased when substrates underwent heated vacuum
Direct validation of the in silico models with in vivo data is treatment and sonication in isopropanol. It is important to
necessary. Mechanical models also could be further improved note that leachable-mediated toxicity is not isolated to SU-8
by incorporating more accurate mechanical properties of the alone, as other groups have described toxicity with PDMS
surrounding glial scar. Work has only recently been
and Poly Vinyl Alcohol (PVA) as well. Therefore toxicity
completed by the Muthuswamy group to quantify microelec-
testing should be performed on any new polymer system
trode-induced changes in the mechanical properties of the
before time consuming and costly animal trials. However, as a
surrounding tissue [96].
word of encouragement, it is likely that more careful
preparation of these materials will overcome issues related
to toxicity, as many are routinely used in biomedical
4.1.2. Off-the-shelf compliant polymeric materials for
applications without incident.
intracortical microelectrodes. Several groups have
Moisture uptake is another factor that may impact the
developed compliant microelectrode substrates and coatings
performance of polyimide, as well as a number of the in situ
from off-the-shelf polymeric materials. These materials
softening materials that will be discussed in section 4.14.
include polyimide, benzocyclobutene (BCB),
polydimethylsiloxane (PDMS), parylene-C and SU-8 Polyimide, for example, swells by approximately 4-6% (w/w)
[201, 207213]. upon implantation [208, 217, 218]. Swelling of polyimide has
To date, only a limited examination of the tissue response been linked to a rapid decrease in electrode performance after
to microelectrodes made from off-the-shelf complaint implantation. However swelling may not be altogether
materials has been performed. In vitro culture has been the negative. As will be described in section 4.6.1, Tresco and
predominant characterization tool. The results of these studies colleagues have proposed that polymer swelling may also
have indicated that a number of traditional compliant provide an additional clearance mechanism for pro-inam-
polymers are non-toxic and support the attachment of matory soluble factors [137]. Therefore, swelling may
neuronal and glial cells. Interestingly, there is little evidence provide an alternative/complimentary explanation for
that any of these off-the-shelf materials signicantly reduce improvements in the neuroinammatory response to compli-
the in vivo neuro-inammatory response. ant polymer substrates, provided that the increased water
Characterization of recording performance from electro- uptake does not interfere with the electrical circuit or
des made from off-the-shelf compliant materials is also electrode insulation.
quite limited. Again the majority of microelectrodes made
from off-the-shelf compliant materials have proven success-
ful during in vitro recording studies. A limited number of 4.1.3. Strategies to prevent buckling during insertion of
materials have undergone acute in vivo testing. For example, compliant microelectrodes. While the stiffness of many
using BCB-based microelectrodes, Clement et al succeeded off-the-shelf compliant materials is still signicantly
in recording neural signals from rat cortex [214]. Addition- higher than brain tissue, it is low enough to cause buckling
ally, Altuna et al were able to record multi-unit activity as in devices made on the same scale as traditional microwires or
well as local eld potentials (LFPs) using an SU-8 based planar MI-style microelectrodes. To prevent buckling a
microelectrode [215]. Longer-term studies are now needed to number of larger device designs and insertion aides have
examine if these complaint microelectrodes reduce the neuro- been developed.

16
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

Perhaps the simplest method to prevent buckling of more polymers have many attractive properties including the
compliant microelectrodes is to increase the size of the device neutral pH of products created during hydrolysis of the
beyond the traditional architectures used in microwire or polymer. Other degradable shuttles that have also been
silicon-based implants. LaPlaca took this approach to investigated include poly(lactic acid), glucose and gelatin
facilitate insertion of Parylene-C based microelectrodes [212, 222, 224228].
[201]. Similar to MI-style microelectrodes LaPlacas probes In summary, a number of strategies have been developed
were designed to be 100 m wide; however, the thickness was to facilitate insertion of compliant microelectrodes including
roughly doubled to 25 m. LaPlacas ndings indicate that increasing device size and the use of permanent and
only slightly larger designs may be needed to prevent temporary reinforcement schemes. It is important to note that
buckling. Obviously increasing device size may have the use many of these methods will increase the initial
consequences. For example, increasing device size will iatrogenic injury. Furthermore increasing device size or the
reduce device compliance and increase strain induced on use of a permanent insertion aide may negate any mechanical
the surrounding tissue. In addition, increasing device size will benet achieved by using a compliant polymer, thus
also exacerbate the initial iatrogenic injury. Interestingly, providing no increase to the quality and stability of neural
Skousen et al has indicated that the neuroinammatory recordings.
response to single penetrating devices may not be greatly
impacted by increasing device size beyond that found in
traditional designs [117]. However, ndings from Skousen 4.1.4. In situ softening materials. An alternative approach to
and others also indicate that the neuroinammatory response the strategies described above has been the development of
can be greatly reduced by transitioning to architectures that in situ softening materials as substrates for intracortical
are smaller than traditional designs (see section 2.1) microelectrodes. Such smart materials are being considered
[117, 145, 159]. for a broad range of biomedical applications, including use as
Other efforts to prevent buckling of compliant micro- delivery vehicles for therapeutic molecules, use as mechanical
electrodes have focused on reinforcing compliant polymers actuators, and as scaffolds for regenerative medicine
with stiffer materials. For example, Lee et al reported on a applications [229234]. In situ softening materials have
new design for polyimide-based intracortical microelectrodes, received attention as microelectrode substrates as they are
which provides adequate stiffness for insertion into neural sufciently stiff to facilitate implantation into the brain, but
tissue [217, 218]. In Lees design, a 510 m thick silicon then soften in vivo to better match the mechanical properties
layer was applied to the polymer to prevent buckling during of cortical tissue.
insertion. Similar designs have been reported for BCB The rst realization of an in situ softening microelectrode
[219, 220]. Penetration tests into rat brains showed that was reported by Capadona et al in 2008 [100]. Taking a
reinforced polyimide based microelectrodes of similar size to biomimetic approach, the team utilized the microstructure of
a standard MI array could penetrate the rat pia without the sea cucumber dermis as the blueprint for a new class of
buckling. However it should be noted, that the composite stimuli-responsive, mechanically adaptive polymer nanocom-
Youngs modulus of these electrodes increased signicantly posites. Specically, the current generation of mechanically-
from 2.8 GPa (neat polyimide without silicon backbone) to adaptive nanocomposites is based on a poly(vinyl acetate)
31 GPa and 58 GPa with a 5 or 10 m thick silicon layer, (PVAc) matrix reinforced with rigid cellulose nanocrystals
respectively [217, 218]. Therefore, the use of permanent (CNCs) [103]. When dry, these nanocomposites are in a rigid
reinforcement may be counterproductive towards minimizing state (E' = 5.1 GPa), due to the glassy matrix and the rigid
chronic tissue strain as the overall compliance is reduced percolating network of the CNCs. Upon exposure to
little, if at all, compared to traditional devices. physiological conditions, the nanocomposite absorbs uid
As an alternative to the silicon-reinforced systems and swells considerably (3090% w/w depending on the type
discussed above, Takeuchi et al incorporated a microuidic of CNCs). Subsequently the nanocomposite undergoes phase
channel into a compliant parylene-C based microelectrode transition and softens (E' = 12 MPa) as water plasticizes the
[209]. Takeuchi lled the channel with a dissolvable poly matrix and disassembles the CNC network (gure 10). It was
(ethylene glycol) (PEG) reinforcement. Using this approach, shown that dry implants of this nanocomposite can readily be
the authors were able to successfully insert their compliant inserted through the pia mater into the cerebral cortex of a rat
device and record neural signals directly after implantation. without the need for assistive devices. The insertion of the
Despite the promise of these early reports, longer-term studies chronically compliant materials was a signicant feat as
have not been reported. reference implants consisting of the neat matrix polymer
More recently, and perhaps inspired by Takeuchis (PVAc) buckled under lower loads before they could be
dissolvable system, several groups have investigated the use inserted into the cortical tissue [98]. Ex vivo studies conrmed
of biodegradable polymers as shuttles for compliant devices that the initially stiff microscale nanocomposites rapidly
[243, 251, 252]. One of the most promising examples of a softened when implanted into the rodent brain. Figure 11
degradable shuttle was explored by Shain and Kohn. In a shows the stiffness of a 12.2% v/v PVAc/CNC nanocompo-
series of investigations, Shain and Kohn studied several site upon introduction into articial cerebrospinal uid
tyrosine-based polycarbonates as biodegradable carriers for (ACSF) at 22 C (Youngs modulus 3400 MPa), and reveals
intracortical microelectrodes [221223]. Tyrosine-based that the softening occurs over the period of 15 min to reach a

17
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

Figure 10. Top: pictures of a sea cucumber, in the threatened (stiff) and relaxed (soft) and state. Bottom: simplied schematic representation
of the switching mechanism found in the sea cucumber dermis and used in physiologically responsive mechanically-adaptive
nanocomposites. A soft matrix is reinforced with rigid particles, whose interactions in are moderated by a chemical agent.

evaluations of in situ softening materials in vivo demonstrated


that compliant implants more rapidly stabilized neural cell
populations than rigid microwires at four weeks post-
implantation [94]. However, no signicant difference was
observed at eight weeks post-implantation. Thus, the results
of Harriss initial studies could be interpreted that, despite
acute benets, the mechanical mismatch between microelec-
trodes and cortical tissue appears to have little effect on the
chronic neuro-inammatory response. However, this over-
simplied interpretation deserves further consideration. Pre-
viously Tresco and colleagues have observed that microwires,
on a similar size scale as those used by Harris et al have a
Figure 11. Plot showing the log of the Youngs modulus of reduced response compared to larger MI-style microelec-
mechanically adaptive PVAc/CNC nanocomposites as function of trodes [194]. Therefore the lack of signicant difference at
exposure time to ACSF or implantation time in the rat cortex. Data
were acquired by either a dynamic mechanical analysis (DMA data, eight weeks may actually quite promising.
open squares; bulk materials) using a submersion clamp and To verify this interpretation, Nguyen et al recently
exposing the sample to ACSF preheated to 37 C or mechanical tests completed a more comprehensive evaluation of the neuroin-
of microprobes that had been implanted into the rat cortex for the ammatory response to PVAc/CNC nanocomposite implants
time indicated and which were explanted for microtensile testing
(ex vivo data, open circles). The x-axis indicates the time of exposure through a 16 week implantation period. At this later time
to either ACSF or implanted in the rat cortex, respectively. point, they observed nearly complete attenuation of inam-
matory cell activation, and the absence of any appreciable
neuron loss surrounding PVAc/CNC nanocomposites com-
Youngs modulus of 33 MPa. A comparison between the pared to chemically-matched PVAc-coated MI-style micro-
Youngs modulus of microprobes that had either been electrodes (gure 12) [235]. Interestingly, unlike Harris
implanted in a living rat cortex showed no statistical initial study, few statistically signicant differences were
difference immersed for 15 min in ACSF (gure 11) [97, 98]. observed between compliant and stiff PVAc implants at early
In preliminary investigations, Hess et al [91, 92] have time points. This discrepancy could be due to differences in
shown that functional microelectrodes can be made using either the controls, surgical technique, or a number of other
laser micromachining followed by deposition of a Parylene-C factors, but it does emphasize the difculty of intra-study
insulating layer, sputtering of Ti/Au electrodes, and deposi- comparisons and the need for further standardization of
tion of an overlaying Parylene-C coating. Initial histological techniques throughout the eld.

18
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

sensitivity to a number of environmental factors necessary for


photolithographic processing, a transfer-by-polymerization
process was used instead. In vivo studies demonstrated that
both acrylate and thiolene/acrylate-based microelectrodes
were capable of recording neuronal signals in a rat cortex.
However, improved histological studies to further verify these
ndings, and a head-to-head comparison of recording
performance to that of traditional microelectrodes have not
been reported to date [104].
Recently Tien et al used silk broin to fabricate a third
type of mechanically adaptive microelectrode. [238] The
elastic modulus of silk is reduced upon hydration from
1.8 GPa in the dry state to 20 MPa in the wet state. Beyond
being a compliant, Tien et al have also engineered their
system to release scar-inhibiting agents such as the enzyme
chondroitinase ABC (chABC) as a potential avenue to
ameliorate axonal growth inhibition by CSs within the glial
scar. While promising, to date, the effect of silk broin
coatings has only been examined using in vitro models and no
recording studies have been performed.

4.1.5. Summary of important considerations regarding


compliant microelectrodes. Mechanical mismatch has been
hypothesized to promote and perpetuate the neuro-
inammatory response. If this hypothesis proves true, an
obvious solution is to increase the compliance of implanted
microelectrodes. However, microelectrode design also must
be concerned with the conicting mechanical requirements
necessary to prevent microelectrode buckling during
insertion. Fortunately, a number of strategies have been
developed to overcome this hurdle including insertion aides
and in situ softening materials.
Although, some of the strategies described above have
Figure 12. (A) Immunohistochemical analysis of CD68, a cellular facilitated insertion of single-shank electrodes, challenges still
marker for activated microglia/macrophages. Representative uor- exist if they are to be applied to multi-shank designs. Parallel
escence microscopy images of stained tissue show a distinct benet to the idea that it is improbable that one absolute mechanism
of mechanically compliant implants, compared to the chemically mediates microelectrode failure, it is also unlikely that one
matched non-compliant implants (16 weeks post-implantation;
p < 0.05). Scale bar = 100 m. Error bars represent standard error. (B)
device will serve the needs of the entire recording community.
Immunohistochemical analysis of neuronal nuclei (NeuN) around Therefore, even single shank implants may provide promise
the implant site. Representative uorescence microscopy images of to individual laboratories and in specic applications.
stained tissue show that neuronal dieback around the non-compliant As stated, perhaps the two largest gaps in the eld of
implant was signicantly higher than in case of the compliant compliant microelectrodes is that no studies have been
nanocomposite implant at 16 weeks post-implantation. The bar
graphs show quantication of neuron densities. * Denotes
performed to directly quantify: (1) the effects of device
signicance between non-compliant and compliant samples; # stiffness/compliance on electrode recording quality, and (2) if
denotes signicance between noted implant and age-matched sham microelectrode-induced strain over the indwelling period can
control (p < 0.05). The horizontal dashed line represents the 100% be reduced by material choice or electrode design. Therefore,
neuron level as determined by quantication of age-matched sham due to the well-established strategies for overcoming
animals. Error bars represent standard error.
mechanical mismatch, particular emphasis should be directed
at validating this widely pursued hypothesis. If in vivo studies
conrm that compliant materials are indeed better than
In situ softening intracortical implants have also been traditional rigid microelectrodes, one must ask how compliant
made from shape memory polymers (SMPs) [236, 237] do they need to be? While answering this question it will be
[104, 236]. For example, Sharp et al developed SMP-based critical to utilize properly designed experiments and appro-
microelectrodes from an epoxy-based polymer using a micro- priate controls to avoid the insertion of confounding
casting technique [237]. More recently, Ware et al developed explanations and interpretations.
SMP based-microelectrodes from acrylate and thiol-ene/ Finally, and most importantly, one of the larger problems
acrylate polymers [93, 236] [104, 236]. Due to acrylates with softening implants is the requirement for some degree of

19
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

water to switch the mechanical properties. In our unpub- while the extent of iatrogenic injury may play a role in the
lished experience, the accumulation of water into early severity of the neuro-inammatory response, Seymour and
devices created delamination between material components, Kipke clearly demonstrated that other architecture-governed
and created problems which limits evaluation of the probes to properties are at play as well. The authors concluded that
histological studies, preventing long-term electrophysiology mechanical differences between the thin adjoining lattice
experiments. Recent advances in fabrication and materials structures and the larger primary solid shanks were the
processing has overcome this limitation [91], and electro- underlying cause of their results. While studies have
physiology studies are underway. suggested that mechanics may play a role in the neuro-
inammatory response (sections 3.4.2 and 4.1), further
4.2. Manipulating microelectrode architecture to reduce neuro- mechanisms could also be at play.
inflammation For example, Skousen et al have suggested that
architecture (specically the local surface area) may be
A second major strategy to reduce the neuro-inammatory manipulated to control the number of inammatory cells and
response to intracortical microelectrodes has focused on the concentration of pro-inammatory and cytotoxic soluble
altering device geometry and architecture. A number of the- factors at the device interface [117]. To test their hypothesis,
ories have been proposed for how altering geometry and Skousen et al compared the tissue response of planar silicon
architecture could be used to reduce the neuro-inammatory microelectrodes with either a solid shank or thin lattice
response. Theories include (1) altering tip geometry or the architecture. Despite being less compliant than the polymer-
penetrating prole of the microelectrode to reduce surgical based lattice devices used by Seymor and Kipke or the
trauma, (2) reducing feature size to improve mechanical compliant materials discussed in section 4.1 [159], the
compliance, and (3) minimizing presented surface area to silicon-based lattice microelectrodes still signicantly reduced
reduce the local number of inammatory cells in a given the neuro-inammatory response after an eight-week indwel-
volume. ling period. Specically, Skousen et al observed a reduction
in inammatory cell activation, blood brain barrier dysfunc-
4.2.1. Theories for how microelectrode architecture impacts tion and neuronal loss surrounding the lattice microelectrodes.
neuro-inflammation. Szarwoski et al conducted the rst Skousens ndings conrm that other architecture-governed
study investigating the potential of changing device properties beyond iatrogenic injury or microelectrode com-
architecture to alter the neuro-inammatory response [239]. pliance inuence the severity of neuro-inammatory
The authors studied the neuro-inammatory response to a response. Current studies are investigating the use of novel
variety of devices with different cross sectional areas, tip architectures to improve microelectrode function and
geometries, and surface roughness and concluded that the biocompatibility.
tissue response was independent of these electrode properties.
However, it is possible that their results and conclusions stem
from the fact that the range of given parameters were too 4.2.2. Driving next-generation microelectrode designs using
narrow to induce signicant changes in the neuro- predictive modeling. To facilitate further investigation of the
inammatory response. Indeed, the broad conclusions of role that microelectrode architecture and other constitutive
Szarwoski et al have since been contradicted by several recent properties play in the neuro-inammatory response, several
studies [145, 159, 240]. computational models have been developed. The rst series
Of the contradicting studies, both Stice et al and Thelin of these models estimated the mechanical strains induced in
et al provided evidence that changing device geometry and the surrounding tissue due to microelectrode architecture and
reducing presented surface area impact the neuro-inamma- stiffness [89, 202204]. Such modeling studies support the
tory response [145, 240]. The two studies independently hypothesis that mechanical mismatch between the implanted
revealed signicant differences in classic hallmarks of the microelectrode and surrounding brain tissue could lead to
neuro-inammatory response between microwires of different adverse strains and stresses being generated [95].
diameters. Both groups hypothesized that reducing the initial While mechanical models have expanded our under-
iatrogenic injury by presenting a smaller cross-sectional area standing of the impact of a number of biotic and abiotic
drove their results. However, while plausible, this interpreta- parameters, there is still further work that should be pursued.
tion is confounded due to differences between the presented For instance, mechanical models could be further improved
surface area and curvature of the disparate sized microwires by incorporating more accurate mechanical properties of the
as well as possible differences in mechanical compliance. surrounding glial scar. Work has only recently been
Additionally, work presented by Seymour and Kipke completed by the Muthuswamy group to quantify microelec-
found signicant differences in both the neuronal and non- trode-induced strain on the surrounding tissue [96]. This
neuronal cell responses between a SU-8/parylene-C based newly described quantied strain data will be extremely
electrodes larger shank and an adjoining lateral platform useful to understand whether modeled in vivo strain elds
designed with a variety of different sized lattice architectures around microelectrodes compare to strain ranges shown to
[159]. Using devices with identical penetrating proles affect various neural and inammatory cells in vitro.
Seymour and Kipke removed the impact of the initial In addition to models describing the mechanical impact
iatrogenic injury from their ndings/interpretations. Thus, of microelectrode design on the surrounding tissue, Skousen

20
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

et al have introduced models to estimate soluble factor at least in the near future until such techniques become more
distribution surrounding various electrode designs [117]. commonplace.
Results indicate that the spatial distribution of pro-inamma- In addition, as microelectrode dimensions are becoming
tory factors surrounding an implanted device is governed by smaller the devices become increasingly fragile and prone to
the local number of adherent inammatory cells, and the mechanical failure. Use of structural elements such as lattice
spatial summation of their released soluble factor gradients. designs could be employed to improve the durability of small
Thus, reducing feature size and isolating architectural features. However, as noted by Seymor and Kipke as well as
components spatially from one another can be used to Skousen et al lattice structures permit increased tissue growth
minimize the concentration of negative soluble factors. This through the device [117, 159]. Such ingrowth could
specic architectural approach reduces soluble factor con- potentially exacerbate the amount of tissue removed upon
centration by limiting the quantity of source cells at the device extraction as a result of failure or infection. Therefore,
microelectrode/tissue interface. Other means of reducing the a higher-level view must be taken to ensure that any
concentration of pro-inammatory and cytotoxic molecules architectural changes used to reduce the neuro-inammatory
by incorporating passive permeability sinks or using active response do not limit microelectrode performance or
antagonists will be discussed further in section 4.6. biocompatibility by eliciting new complications.
Similar to the mechanical models described by above,
Skousen et als models to describe soluble factor distribution 4.3. Non-fouling surface modifications to prevent cell adhesion
are still relatively simple and should be further improved to
fully establish their validity while also increasing their An alternative strategy to reduce the neuro-inammatory
accuracy and usefulness. For example, to date soluble factor response to implanted microelectrodes is the use of non-
distribution models have focused primarily on estimating the adhesive coatings to prevent inammatory cell attachment
distribution of the potent cytokines TNF and MCP-1 [117]. and activation at the microelectrode surface. Several surface
However, as mentioned above in section 3.4.4, various coatings or materials have been described to reduce or prevent
interleukins, reactive oxygen species, and matrix metallo cell adhesion.
Silicon carbide (SiC) has been long studied as an alter-
proteinases (MMP) 2 and 9 are also believed to mediate the
native substrate for neural electrodes due to its documented
neuro-inammatory response [69, 107, 135]. Therefore
biocompatibility in other medical device applications. SiC has
expanding these models to include other soluble factors that
gained attention because it is chemically inert, and lends
have different effective concentration levels, half-lives,
itself readily to MEMs fabrication and chemical surface
clearance rates or diffusivity is needed. Additionally, the
modications to improve in vitro biocompatibility (or biolo-
models could be further improved by incorporating a graded
gically inert/non-fouling). SiC was recently reviewed by
diffusivity that better reects the impact of the glial scar on
Saddow [241].
volume transmission. To best understand the impact of the
The most broadly applied approach in biomaterial sci-
glial scar, as well as to ultimately validate such models,
ence for preventing cell adhesion is through the presentation
techniques need to be developed or adapted to accurately of biologically inert chemical moieties on the surface of the
measure soluble factor distribution in tissue surrounding implant. For the purposes of this section, the term inert
implanted microelectrodes. refers to the ability to resist protein adsorption and cell
Despite the considerable amount of progress based, at adhesion, but is not meant to reect the materials ability to
least in part, on modeling studies, predictive models are still resist corrosion. For review of a variety of surface treatment
under-utilized as a research and design tool. For example, to approaches to create non-fouling substrates, see Raynor
date mechanical models have only been used to predict the et al [242].
potential strains surrounding classic single tine microelec- Tresco and colleagues rst investigated whether reducing
trode designs. Expanding such models to understand how cell adhesion could alter the neuro-inammatory response to
microelectrode architecture can be manipulated to reduce microelectrode arrays. [194, 243] Specically, Leung et al
persistent mechanical damage would be extremely useful. characterized microglial adhesion to a variety of surfaces
Ultimately, once properly validated, both mechanical and in vitro [273]. Subsequently, Winslow et al compared the
soluble factor modeling will be pivotal tools in the toolboxes neuro-inammatory response of planar silicon microelec-
of microelectrode designers. trodes that had a uniform coating of the hydrophobic insulator
parylene-C to that of identical uncoated devices [191]. In
vitro, parylene-C reduced microglial adhesion by 95%. A
4.2.3. Important considerations when manipulating similar reduction of cell adhesion was observed following
microelectrode architecture. While considering architectural device removal after two, four or 12 weeks of implantation in
modications for improving microelectrode function and rat cortical tissue. Interestingly, no signicant difference was
biocompatibility, it is important to consider the limitations observed in the neuro-inammatory response or the level of
and hurdles inherent to this approach. For example, while neuronal loss surrounding the parylene-C coated devices
already in existence, newer methods of placing conducting compared to uncoated microelectrodes.
traces along the substrate may need to be employed. Use of Garcia and colleagues recently built upon the work by
such fabrication techniques could increase production costs, Winslow et al using conformal microgel coatings of poly(N-

21
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

isopropylacrylamide) (pNIPAm) cross-linked with poly interface. However, Moxon and coworkers only examined the
(ethylene glycol diacrylate) (PEG-DA) on silicon microelec- response out to one week post-implantation, and the lasting
trodes [195]. Long polyethylene glycol (PEG) chains take impact on the neuro-inammatory response or recording is
advantage of entropic and osmotic repulsion to prevent cell still unclear and deserves further study.
adhesion through the inhibition of protein adsorption. In vitro More recently, the VandeVord laboratory has also begun
analyses demonstrated signicantly reduced astrocyte and investigating the effects that nanopatterned substrates may
microglia adhesion to microgel-coated microelectrodes, have on astrocyte reactivity [251]. Utilizing nanofabrication
compared to uncoated controls. However, persistent inam- techniques, Ereifej et al created poly(methyl methacrylate)
mation was observed surrounding both uncoated and coated (PMMA) surfaces with various degrees of groove width (no
microelectrodes following one, two and 24 weeks of grooves, 555 nm, or 277 nm). Cultured astrocytes were less
implantation in rat cortex. Furthermore, neuronal density responsive to the narrower 277 nm grooves, compared to
around the implanted electrodes was also lower for both other surfaces. This preliminary study further demonstrates
implant groups compared to the uninjured controls. the role of surface topography in manipulating glia cell
As no cells were found adhered to either the Parylene-C reactivity. However, future studies will require characteriza-
or pNIPAm/PEG-DA coated microelectrodes upon removal, tion with microglia and macrophages, which are more likely
it would appear that both coatings were still functioning at the to experience topographical cues presented by implanted
end-points studied. Therefore, the combined ndings indicate microelectrodes.
that cell adhesion is not necessary to drive the neuro- While surface topography is very important for cell-
inammatory response. In that respect, Otto at colleagues material interactions, cells cannot adhere directly to synthetic
recently investigated the effects of PEG coatings on electrode materials. The natural ECM is composed of many bular
impedance in acute in vitro and in vivo models [244]. Otto proteins. Surface topography (or architecture) alone does not
demonstrated that exposure of the unmodied electrode to facilitate all cell function. Typically, the presentation of bio-
bovine serum albumin in vitro, as well as expose to in vivo logical motifs are equally as important [252], and should be
protein solutions (the brain), resulted in both resistive and considered in combination with the above approaches.
capacitive changes to the electrode impedance. Further, by Additionally, the above studies, which investigated sur-
applying a high molecular weight PEG to the electrode, the face topography, did not differentiate between the insulating
increase in impedance both in vitro and in vivo was reduced. substrate of the electrode and the conductive metal/polymer
Ottos study demonstrated that non-cellular components portions of functional microelectrodes. However, it appears to
likely inuence the performance of microelectrodes as well. be important to probe how changes to the conducting elec-
Unfortunately, to the best of our knowledge, no type of inert trical contact would affect the surface area of the contact, and
coating has been shown to effectively reduce chronic device- thus the electrical impedance and recording performance. For
associated inammation in any other tissue or implant model example, studies with increased surface area of the recording
over the extended periods of time that may be clinically site must critically evaluate if the source of improved impe-
relevant for BMI applications. dance measurements are a result of reduced neuroinamma-
tion or the increased surface area.
4.4. Topographical control of cell phenotype
4.5. Incorporating bioactive materials
In contrast to the general failure of non-adhesive surfaces to
improve the long-term biocompatibility of microelectrodes, Over the last decade, another promising approach to control
active approaches that permit adhesion while controlling cell cell phenotype at the biotic/abiotic interface has been devel-
phenotype have shown considerable promise. One active oped, which involves the decoration of microelectrodes with
approach that has received extensive study is the use of bioactive surface coatings [226, 253263]. A broad-spectrum
controlled surface topography. Specically, topographical of bioactive materials has been immobilized on the implant
cues have been used to control the adhesion, migration, surface to control the neuro-inammatory response (table 1).
orientation and gene expression of a variety of cell types Bioactive materials have been shown to be at least tempora-
[245, 246]. Despite studies showing that nanostructured sur- rily successful in attenuating the neuro-inammatory response
faces can have a positive inuence towards controlling cell to intracortical microelectrodes within the brain tissue.
functions, the underlying mechanism(s) are not well under- However, it is not clear if the temporary effect of most
stood [247]. bioactive strategies is a result of biomolecule consumption
Building in part upon this research, Moxon and co- (degradation or exhaustion of the coating), or an evolution of
workers conducted studies to investigate the potential of redundant biology overcoming the initial effect of the surface
controlled topography to improve microelectrode bio- modication.
compatibility [248250]. Specically, Moxon et al examined Perhaps one of the simplest and most common bioma-
the impact of presenting roughened, porous silicon or ceramic terials approaches is the passive adsorption or covalent
surfaces, which were designed to better mimic the nanos- immobilization of ECM components to promote directed
tructured and brous nature of the ECM. Implantation of cell attachment. As biomaterialists became interested in the
nano-porous surfaces was found to induce less glial activation device-mediated neuro-inammatory limitations to intracor-
and to improve neuronal density at the microelectrode/tissue tical microelectrodes, the attachment of ECM proteins and

22
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

Table 1. Non-exhaustive list of bioactive surface treatments for intracortical microelectrodes.

Bioactive agent Coating material Electrode type Reference


Dexamethasone (DEX) Poly(pyrrole) (PPy) Gold-coated coverslip [185]
PLGA nanoparticles embedded in alginate hydrogel Silicon [262]
Nitrocellulose Silicon [166]
PLGA nanobers Silicon [264]
Poly(propylene sulde) nanoparticles embedded in poly(ethy- Platinum [265]
lene oxide)
Laminin (LN) Dextran Silicon, gold and [266]
polyimide
Poly(ethyleneimine) (PEI) or chitosan (CH) Silicon [260]
Poly(pyrrole) (PPy) Gold-coated coverslip [267]
Poly(ethyleneimine) (PEI) Silicon [268]
L1 Poly(ethylene glycol) (PEG) Silicon [257]
Silicon dioxide modied using silane chemistry Silicon [256]
Parylene-C Tungsten [269]
-MSH Nitrocellulose Silicon [263]
Silicon modied using silane chemistry Silicon [255]
Nerve growth factor (NGF) Parylene-C Silicon [224]
Poly(ethylene dioxythiophene) (PEDOT) Platinum [270]
Poly(pyrrole) (PPy) Polystyrene [271]
Superoxide Dismutase (SOD) Surface functionalization Silicon [272]

peptides onto the microelectrode surface were among the rst implantation [268]. However, LN coating elicited a more
methods reported. ECM-based materials for neural interfacing robust pro-inammatory response at one day post-implanta-
applications have been recently reviewed by Chen and Allen tion than uncoated devices, as indicated by increased CD-68
[273]. Therefore, only select representative examples or new positive microglia, GFAP astrocytes, and pro-inammatory
considerations will be discussed here. cytokine expression. Interestingly, neuron densities were
Among the most important ECM proteins for neural statistically similar at all of the time points investigated,
applications is laminin (LN), an adhesive protein that plays suggesting no advantage to the LN coating for recording
crucial roles in cell migration, differentiation, and axonal applications.
pathnding [274]. The two main peptide sequences from LN In parallel to the LN work in the Bellamkonda lab,
that are often targeted for biomaterial applications include Ile- several laboratories have investigated the ability of doping
Lys-Val-Ala-Val (IKVAV) and Tyr-Ile-Gly-Ser-Arg conductive polymers (discussed further in section 4.7), with
(YIGSR). In 1993, Massia and Hubble were among the rst to LN-based peptides in order to increase neuronal attachment.
report on receptor-specic cell spreading on surfaces cova- For example, Stauffer and Cui investigated two different LN
lently immobilized with YIGSR [275]. Twenty years later, fragments, YIGSR and RNIAEIIKDI, as dopants in an elec-
Massia developed a surface grafting method that allows for tropolymerized poly(pyrrole) (PPy). The goal of the initial
the covalent immobilization of IKVAV on the surface of in vitro study was to combine the critical electrical properties
silicon, silicon oxide, gold and insulating polymers such as of conducting polymers with the ability to promote specic-
polyimide, all common components of intracortical micro- cell attachment (YIGSR) and neurite outgrowth
electrodes [266]. This work highlights the specicity of LN (RNIAEIIKDI) [267]. Stuaffer and Cuis results conrm the
peptides for supporting neuronal attachment, and reinvigo- cell-specic attachment and growth seen over the previous
rated the use of LN-derived strategies for intracortical decades in many laboratories. The novelty of their work was
microelectrode applications. that the combination of the two peptides on a conducting
Likely based on Massias work, the Bellamkona group polymer scaffolding synergistically increased both neuronal
reported a series of publications utilizing LN-based coatings attachment, and neurite outgrowth, while also demonstrating
as surface modications for intracortical microelectrodes low impedance and increased charge capacity. Despite the
applications [260, 268]. First, a layer-by-layer (LBL) assem- promising in vitro results suggesting that LN-containing
bly was used to build up a deposition of poly(ethyleneimine) coatings may enhance the long-term recording stability of
(PEI) and LN on silicon wafers with an oxide layer. It was neural interfaces, no studies to date have described the in vivo
found that the PEI-LN layer was stable for at least 7 days recording performance of these materials.
under simulated physiological conditions, and signicantly In a second example of LN-derived peptide incorporation
improved neuron adhesion and differentiation in vitro. The into conducting polymers, Green et al doped PEDOT with
subsequent in vivo study revealed that PEI-LN coatings are DEDEDYFQRYLI and DCDPGYIGSR [276]. Interestingly,
able to reduce the counts of reactive microglia and astrocytic Green et al demonstrated that large peptide dopants produced
tissue response to Si-based electrodes after four weeks post- softer PEDOT lms with a minimal decrease in

23
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

electrochemical stability. However, despite the retained single ECM components such as LN or L1 alone may not be
bioactivity of dopant peptides, the effects were largely sufcient to mitigate the neuro-inammatory response. For
dependent on initial cell attachment, and neither of the pep- example, Tanaka et. al showed that microglia cultured on
tides investigated provided the bioactivity of the native LN xed (dead) astrocyte monolayers, even in the presence of
protein. In a later study, Green et al also examined the effect serum, display a resting phenotype [280, 281]. Tanakas
of entrapping nerve growth factor (NGF) within the PEDOT results indicate that cues presented by the astrocyte ECM are
during electrodeposition [277]. The incorporation of NGF sufcient to regulate microglia activation. Interestingly, the
was shown to remain biologically active within the PEDOT. impact of xed astrocyte ECM was signicantly more
However, Green et al also found that the use of both a LN effective at reducing microglial activation than individual
peptide dopant and NGF in the PEDOT resulted in polymers ECM components such as LN or bronectin (FN).
with decreased mechanical and electrical properties compared Building, in part on the ndings of Tanaka et al Tresco
with controls containing only NGF [277]. and colleagues have developed approaches to harvest the
Despite retained biological activity of the incorporated ECM produced by CNS cells, including astrocytes, cultured
peptides, the combination of biological molecules within on sacricial open-celled foam substrates [282]. Immuno-
conducting polymers has thus far failed to provide the histochemical and proteomic analysis with tandem mass
synergistic benets that the eld has anticipated. In fact, the spectroscopy revealed that the harvested material consisted of
Poole-Warren group recently provided a report of the per- a complex network of ECM components including collagen,
formance of conducting polymer electrodes, without the bronectin, laminin and various glycosaminoglycans. In vitro
incorporation of biological molecules [278]. Since isolated cytocompatibility studies of the decellularized material have
peptide sequences typically demonstrate enhanced activity of shown the material to be non-toxic and adhesive to various
targeted functions compared to full protein controls [279], it is cell types. Tresco and colleagues are currently investigating
likely that the immobilization methods employed within the impact of astrocyte-derived ECM coatings on the neuro-
conducting polymer requires further optimization, perhaps inammatory response in rodent models.
insulating spacer groups to isolate the biomolecules from the Utilizing complete, tissue-specic ECM may provide
polymers.
additional benets to single protein approaches. Although
While the use of LN surface modication alone has not
ECM throughout the body shares common protein and gly-
successfully improved the biocompatibility or recording
cosoaminoglycan building blocks, subtle differences indicate
quality of microelectrodes, there is increasing evidence that
that the precise make-up of a tissues-specic ECM is vital in
the presentation of other specic ECM molecules or networks
regenerative applications [283]. Several studies have shown
of ECM components that mimic the complexity of natural
that culturing cells on tissue-specic ECM improves inl-
brain tissue may be useful. For example, the Cui group has
trating cell proliferation rates and increases the expression of
demonstrated encouraging work with the neural adhesion
desired phenotypic cell and tissue characteristics [284288].
molecule L1. Azemi et al demonstrated that neural electrode
In contrast, implantation of non-tissue specic ECM induces
arrays coated with immobilized L1 showed enhanced levels
of attachment of mouse cerebellum neurons in vitro [257]. the formation of undesired, phenotypically irregular tissue at
Azemi et al directly compared the efcacy of L1 with LN. the implantation site [289, 290].
The study showed that while the LN-functionalized surfaces While bioactive approaches, based primarily on ECM
greatly promoted the growth of astrocytes, the L1-functio- proteins and peptides, have shown promise in improving the
nalized surfaces showed signicantly reduced astrocyte neuro-inammatory response to intracortical microelectrodes,
attachment compared to both LN-coated and uncoated control one limitation of these strategies is their short-lived nature.
surfaces [257]. In a subsequent paper [256], Azemi et al For example, inammatory cells that unavoidably become
investigated the neuro-inammatory response to L1-functio- activated in response to the initial iatrogenic trauma are
nalized Michigan-type microelectrodes implanted in a rat known to phagocytize and remove adherent and even cova-
cortex for up to eight weeks. The study revealed that L1- lently immobilized proteins over time. Therefore, bioactive
functionalized microelectrodes show signicant reduction in coatings should primarily be thought of and used as one
reactive tissue gliosis when compared with uncoated elec- component of a combinatorial strategy for improving micro-
trodes. The most promising aspect of the L1 approach is the electrode function and biocompatibility. Specically, bioac-
ability to maintain normal neuronal populations while also tive materials may serve as a key component to direct initial
signicantly increasing the density of neuronal lament at the wound healing events and tissue integration following
interface (gure 13). The Cui laboratory has more recently implantation, but are not as likely to be used to improve the
begun to explore the utility of L1-functionalized electrodes long-term recording performance.
for peripheral nervous system applications. Unfortunately, to A further, and often overlooked, concern with protein-
date, no description of the impact of L1 immobilization on based coatings is their potential for immunogenicity. While
recording quality for intracortical microelectrode applications the majority of proteins found in the ECM are well conserved
has been reported, but deserves further attention due to the between animals and humans, interspecies differences do
success of foundational studies. exist. As a result the implantation of even decellularized,
While Azemi et al showed the supremacy of L1 over LN xenogenic ECM has been shown to elicit an adaptive immune
[256], other studies have also shown that presentation of response [291294]. Therefore the use of autologous or

24
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

Figure 13. (a), (b) Representative images of NeuN+ cells (green) around the NM (unmodied) and L1 (L1-peptide grafted) probes after eight
weeks of implantation in rat cortex. Below the set off images, the corresponding normalized cell count differences between L1 and NM
probes for the 0100 mm region away from the interface (*p < 0.05). (c), (d) Representative images of neuronal lament (green) stained
tissue after 8 weeks of implantation in rat cortex. Below the set off images, the corresponding normalized neuronal lament intensity level
differences between L1 and NM probes for the 0100 mm region away from the interface (*p < 0.05). Scale bar = 100 mm. Reprinted with
permission from [257], Copyright 2008 Elsevier.

allogenic materials may prove key to maximizing the clinical and encapsulation. Despite having a larger penetrating prole
success of ECM-based coatings. than any single tined microelectrode, HFMs elicit a very
minimal neuro-inammatory response [152, 205, 299]. While
the roughened surface of the HFM wall may play a role in
4.6. Antagonizing pro-inflammatory and cytotoxic soluble reducing the response [248, 249]. Tresco and colleagues also
factors hypothesize that HFMs act as a permeability sink for pro-
4.6.1. Passive permeability sinks. Several strategies have inammatory and cytotoxic soluble factors. Specically, the
been developed to antagonize the pro-inammatory and membranes semi-permeable wall structure and uid lled
cytotoxic effector molecules secreted by inammatory cells at lumen permit diffusion of reactive soluble factors into the
the microelectrode/tissue interface. The simplest form of device, and thus away from the surrounding brain tissue. Due
antagonism is the use of passive diffusion to reduce the to the short half-life of many pro-inammatory and cytotoxic
concentration of pro-inammatory and cytotoxic factors in molecules, a large portion of the molecules are believed to be
the adjacent tissue. For example, it has long been degraded within the HFM, without ever impacting the
hypothesized that permeability of the adjacent tissue may surrounding tissue.
inuence the overall integration of biomedical devices by To investigate whether a permeable sink strategy could
allowing better clearance of pro-inammatory and cytotoxic be used to improve microelectrode biocompatibility, Tresco
molecules away from the device/tissue interface [152]. and colleagues have investigated the use of thick hydrogel
However new evidence indicates that the permeability of coatings to reduce the neuro-inammatory response [137].
implants themselves may also be manipulated to reduce For this strategy to be effective the coating or sink volume
soluble factor concentration and facilitate improved wound must be thick enough to passively entrap the soluble factors
healing [295299]. until they breakdown by hydrolysis and other passive
While not a microelectrode, one of the earliest descrip- degradation mechanisms. Finite element modeling showed
tions using a passive permeability sink is work with semi- that it is possible to retain pro-inammatory cytokines in the
permeable hollow ber membranes (HFMs) for cell delivery diffusion sink to passively reduce their concentration and

25
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

their subsequent impact on the biology of the surrounding investigate the anti-inammatory properties of the
tissue as long as their residence time in the sink exceeded neuropeptide in the context of intracortical microelectrodes.
their biological half-life. Given the appropriate size of the They initially developed nitrocellulose-based coatings that
sink, cytokines diffusing into the semipermeable surface of were capable of locally delivering -MSH from Michigan-
the device would be unlikely to diffuse out of this region in type microelectrodes [263]. Zhong et al found that -MSH
their active state, and thus would be effectively silenced. released over 21 days remained bioactive and successfully
Subsequent in vivo studies showed that the incorporation inhibited NO production by LPS-stimulated microglia,
of the thick permeable hydrogel signicantly reduced a in vitro.
number of classic hallmarks of the neuro-inammatory Subsequent work by He and Bellamkonda further
response compared to uncoated microelectrodes or those that demonstrated that the immobilization of -MSH retained
received a thin hydrogel coating (surface chemistry control). anti-inammatory properties both in vitro and in vivo [255].
Reduced hallmarks included macrophage recruitment and Specically, when immobilized on the microelectrode sur-
activation, astrogliosis, blood brain barrier dysfunction and face, -MSH again successfully inhibited NO and pro-
neuronal cell loss. inammatory cytokine production by LPS-stimulated micro-
It should be noted that better mechanical matching glia in vitro. More importantly, immobilization of -MSH on
between the soft hydrogel and the surrounding brain tissue the surface of Michigan-type microelectrodes qualitatively
may contribute to the reduced neuro-inammatory response reduced the detection of TNF- mRNA one week post
seen around hydrogel coated microelectrodes described by implantation in rat cortex, and quantitatively reduced the
Tresco and colleagues. However, a reduced inammatory density of both CD-68 and GFAP positive microglia/
response to other stiff semipermeable devices suggests that macrophages and astrocytes, respectively. Unfortunately, He
mechanical matching is not the only factor involved. For et al did not report on the effects of -MSH functionalized
example, work from Desai and colleagues have shown a microelectrodes on the local neuron density or viability [255].
similar reduction in the inammatory response, as well as Furthermore, despite the promise of their short-term results,
complement activation, to stiff silicon and metal membranes He and Bellamkonda have not pursued longer histological or
with a semipermeable membrane/lumen structure [300, 301]. functional studies with -MSH.
Furthermore, the similarities in the inammatory response to As an alternative means of antagonizing the impact of
passive permeability sinks across a broad range of stiffness pro-inammatory soluble factors, Taub et al recently reported
combined with the high moisture uptake of many of the on the use of interleukin receptor antagonist (IL-1Ra)-coated
polymers described in section 4.1 could suggest a compli- microelectrodes [259]. While IL-1Ra-coated microelectrodes
mentary/alternative mode of action for many compliant demonstrated signicantly reduced astrogliosis compared to
materials. non-coated microelectrodes, no other histology was provided,
Further studies aimed at better characterizing the and it remains unclear how these materials affect neuronal
response to a variety of design variables are needed due to viability or inammatory cell activation.
the potential that well-established and controllable strategies Another active strategy for antagonizing pro-inamma-
for reducing the neuro-inammatory response could have on tory and cytotoxic soluble factors has been the use of the anti-
intracortical microelectrode technology. Specically, more in oxidant resveratrol [69]. While the use of resveratrol to date
depth studies to isolate the individual and composite roles of has been limited to systemic delivery, the overall approach
mechanical mismatch, microelectrode architecture and device and ndings t well with other active antagonist strategies
permeability are needed. To facilitate these types of advanced and show exciting promise for improving microelectrode
design studies, a number of new or improved methods are function and biocompatibility. Specically, at two weeks
needed to quantify response variables, including both in vivo post-implantation, Potter et al found that animals receiving
strain proles and cytokine distributions surrounding resveratrol therapy at the time of implantation demonstrated
implanted devices. In addition, novel test devices that better reduced blood-brain barrier instability, accompanied with
isolate or control individual design elements need to be increased neuronal density at the microelectrode-tissue inter-
created. face (gure 14). At four weeks post implantation, no
difference was observed in neuronal density between
resveratrol-receiving and control cohorts. The authors have
4.6.2. Active antagonism. In contrast to the passive strategies suggested that the loss of impact on neuronal density is likely
explored by Tresco and colleagues, a number of groups have due to clearance or inactivation of resveratrol over time.
investigated active antagonism of pro-inammatory and While the ndings from the resveratrol studies have not
cytotoxic soluble factors to improve microelectrode provided a long-term solution, they do support the use of
technology. For example, the Bellamkonda group active antagonism of pro-inammatory and cytotoxic factors
investigated alpha melanocyte-stimulating hormone (- to improve microelectrode technology. Therefore, ongoing
MSH) as an anti-inammatory target molecule. -MSH is studies are currently investigating the impact of repeated
an endogenous tridecapeptide with potent anti-inammatory dosing and local delivery of resveratrol, as well as other
properties. Specically, -MSH acts through the inhibition of natural and synthetic antioxidants, that may prove safer and
pro-inammatory cytokines and neurotoxic nitric oxide (NO) more effective than resveratrol itself. Additionally, in order to
production [302]. Zhong and Bellamkonda were the rst to develop a system to provide sustained neuroprotection, Potter

26
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

Blood-brain
Control
Resveratrol Neurons Neurodegeneration barrier stability

Figure 14. Rats treated with resveratrol showed increased neuronal densities at the electrode interface, no appreciable neurodegeneration, and
a signicant improvement in the stability of the blood-brain barrier two weeks post-implantation of Michigan-type silicon microelectrodes.
Scale = 100 mm.

et al also investigated modifying the microelectrode surface factors, singling out one or even a limited number of factors
with an anti-oxidative coating [272]. For initial proof of may not be effective.
concept, they chose the superoxide dismutase (SOD) mimetic Another limitation of many active approaches is their
Mn(III)tetrakis(4-benzoic acid)porphyrin (MnTBAP). Their limited effective duration. There are currently few if any
system utilizes a composite coating of adsorbed and viable options for locally delivering soluble antagonists for
immobilized MnTBAP designed to provide an initial burst- the extended periods of time that may be clinically relevant
release followed by sustained presentation of an immobilized for BMI applications. Systemic delivery over time may be
layer of the antioxidant. Potters results indicate that the effective at reducing neuro-inammation to a tolerable level.
hybrid modied surfaces provide sustained anti-oxidative However, the unwanted side effects associated with the drugs
activity, and reduced the accumulation of reactive oxygen and supplements studied to date make continual systemic
species both intra- and extracellularly.
dosing unattractive. Passive approaches involving the use of
permeability sinks, on the other hand, should persist for much
4.6.3. Further considerations concerning soluble factor greater periods of time, if not indenitely, depending how
antagonization. While comparing passive and active they are implemented.
approaches to antagonize pro-inammatory and cytotoxic Due to the benets and limitations of passive and active
soluble factors, it is important to consider the benets, approaches, we believe that ultimately a composite approach
limitations and hurdles inherent to the two approaches. For will be most effective at enabling effective recording over
example, passive approaches are inherently indiscriminant
clinically relevant time frames for BMI applications. Such an
and will reduce the concentration of likely any soluble factor.
approach will utilize active strategies over the initial acute
This indiscriminate nature could make passive approaches
time frames to promote effective wound healing, while
desirable as they can impact the entire range of pro-
inammatory and cytotoxic soluble factors. Unfortunately, longer-acting passive approaches such as increased substrate
the indiscriminant nature of passive approaches also compliance, reduced surface area or incorporation of a
necessitates that benecial, pro-healing factors will be permeability sink will be used to maintain a homeostasic
impacted along with any negative factors. In contrast, active environment throughout the implant duration. For other
approaches can be tailored to specic targets. However, due applications where microelectrodes will be implanted for
to the overlapping impact of many pro-inammatory soluble shorter durations (weeks-months), composite strategies may

27
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

not be needed and even systemic dosing of soluble


antagonists or anti-inammatory drugs may be sufcient.

4.7. Conducting polymers

The inherent conductive properties of intrinsically conductive


polymers make them a useful class of materials for a wide
range of biomedical applications, such as biosensors, tissue
engineering, neuroprosthetic electrodes, drug delivery, and
actuators [303, 304]. Conducting polymers are particularly
attractive for intracortical microelectrode applications because
they have mechanical properties that lie between those of
conventional metallic microelectrodes and the brain tissue,
can provide high surface area and therewith facilitate an Figure 15. Chemical structures of poly(pyrrole) (PPy) and poly-(3,4-
efcient ion exchange between recording sites and the brain ethylene dioxythiophene) (PEDOT), examples of conducting poly-
mers explored in neural interfaces.
tissue, and can at least in principle, be processed into a broad
range of geometries/structures/architectures. Charge transfer
is improved through reduced impedance and greater selec- PPy as a surface coating for neural electrodes [226]. In his
tivity for both recording and stimulating neural interfacing report, PPy was combined with a genetically engineered
applications; although their intrinsic conductivity is lower protein, designed to incorporate GAGAGS sequences of silk
than that of gold, platinum, or stainless steel electrodes. alternated with the cell-binding sequence RGD. The polymers
The key feature of conducting polymers is conjugated were deposited electrochemically onto the silicon microelec-
double bonds along the backbone with a high degree of - trodes. The study showed that the PPy-coated Michigan-style
orbital overlap, results in electrically conductive materials. microelectrodes had a higher surface area and charge density
Conducting polymers with various morphologies can be compared with uncoated electrodes, which facilitates charge
directly deposited onto intracortical microelectrode surfaces. transport, and more efcient neural communication [305]. In
As a result, the conducting polymer coatings lower the addition, it has been reported that a higher surface area sig-
impedance of the electrodes and can provide a mechanical nicantly lowers the overall impedance of the intracortical
buffer between the stiff intracortical microelectrode and the microelectrodes [310].
compliant brain tissue. Additionally, bioactive agents such as PSS has been used commonly as a dopant material for
anti-inammatory drugs and neurotrophic factors can be PPy due to its stability and in vitro compatibility with
incorporated and delivered from these conducting polymer mammalian neuronal cells [305, 311313]. Cui and Martin
coatings. Several studies have shown that intracortical electrochemically deposited PPy doped with PSS on the
microelectrode functionality can be improved to some extent
neural electrodes, and found that the coated electrodes had an
by coating the microelectrode surface with low-impedance
increased surfaces area, which resulted in a 30-fold decrease
conductive polymer with nanoscale roughness or porosity
in impedance [305]. In 2005, George et al reported on the
[43, 305], or through addition of cell adhesion peptides [305],
biocompatibility of PPy-based cortical implants that had been
proteins [260, 261, 306], or anti-inammatory drugs
doped with PSS or sodium dodecylbenzenesulfonate
[262, 263]. Overall, in vivo studies have shown that these
(NaDBS) [312]. Immunohistochemical studies showed that
conducting coatings may enhance the chronic recording per-
PPy-based intracortical implants-doped either with PSS or
formance of intracortical microelectrodes [43].
Among the currently available conducting polymers, NaDBS after three and six weeks implanted in a rat cerebral
poly(pyrrole) (PPy) and poly-(3,4-ethylene dioxythiophene) cortex had less gliosis than Teon-coated microwire controls.
(PEDOT) (gure 15) are the most studied conducting poly- However, the differences in gliosis at the six-week time point
mers for intracortical microelectrode applications. Such con- had lessened compared to three weeks. Georges study also
ducting polymers have been doped with various dopants such showed that incorporating neurotrophic molecules such as
as poly(styrene sulfonate) (PSS) [305], perchlorate (ClO4 ) nerve growth factor (NGF) and brain-derived neurotrophic
[307], para-toluene sulfonate (pTS) [278], or sulphate (SO4) factor (BDNF) into the PPy matrix promoted the ingrowth of
[308] to modify the surface of metallic intracortical micro- neural tissue into the lumen of the PPy-based implants,
electrodes [309]. In this section we discuss the development compared to implants without growth factors. As noted in
of conducting polymers used to modify the intracortical earlier sections, analysis at limited intermediate time points
microelectrode surfaces with particular attention to the use of can lead to incomplete conclusions. Therefore, further works
PPy and PEDOT. need to be done to demonstrate whether the bioactive mole-
Much of the initial research on conducting polymers for cules can enhance neuronal adhesion and interaction with
neural interfacing focused on PPy due to the ease of pre- conducting polymer-based intracortical implants.
paration, high conductivity, controllable surface properties, The incorporation of conducting polymers within
and the possibility to electropolymerize this polymer from hydrogels that are used to coat conventional microelectrodes
water. In 2001, Martin and co-workers investigated the use of is another intriguing approach to better integrate intracortical

28
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

This is likely due to the deposition of conducting polymer


(i.e. PEDOT), which decreases the impedance of the electrode
(increase sensitivity). Cui and Martins ndings are highly
desirable for potential use of PEDOT as alternative con-
ducting material for neural electrodes.
After Cui and Martin initial report, several studies fol-
lowed, which further explored the use of PEDOT as elec-
trically conductive material in intracortical microelectrode
applications. For example, Xiao et al modied the surface of
intracortical microelectrodes using PEDOT-MeOH that was
Figure 16. Chemical structures of monomers used to fabricate electrochemically doped with poly(styrene sulfonate) (PSS)
PEDOT and PEDOT-MeOH.
[316]. In this study, Xiao et al improved the limited proces-
sability (aqueous solubility) of PEDOT through the addition
microelectrodes with the neural tissue. Hydrogels are attrac- of an appropriate pendant side group onto the backbone. To
tive due to their use in many biomedical device applications, this end, polar derivatives of EDOT, specically EDOT-
their high water content which causes the mechanical prop- MeOH (the chemical structure of which has been shown in
erties to be similar to those of the brain tissue, and their gure 16) was used. Xiao et al found that the PEDOT-MeOH
porous network structure which can facilitate charge transport coating decreased the impedance by almost two orders of
especially if conducting polymers are insulated. In one magnitude in comparison to the uncoated Michigan-style
example, Michigan-style microelectrodes were rst coated microelectrode. Decrease in electrode impedance leads to
with cross-linked alginate and then PPy/PSS was subse- improved charge transfer from the surrounding brain tissue to
quently electrochemically polymerized on the device surface the intracortical microelectrode, which is argued to lead to
[90]. PPy was observed to grow vertically form the electrode more effective recording and stimulating. In another investi-
surface, and at the recording site recording site. It was found gation, Yang et al [317] electrochemically deposited surfac-
that the impedances of the porous hydrogel modied with tant-induced ordered PEDOT onto gold-coated Michigan-
conducting polymer lms are around three orders of magni- style microelectrodes. Although, this ordered PEDOT poly-
tude less than the impedance of the metal microelectrodes. mer coated-electrodes exhibited a lower impedance and a
The authors also found that the PPy/PSS-alginate-coated higher charge capacity than uncoated electrodes, it was found
recording sites were capable to transporting charges as ef- that the surfactant used in the preparation leached from the
device when placed in a cell culture medium and killed all
ciently as conventional electrodes. Despite the growing
nearby cells in the culture.
number of studies being conducted with PPy for intracortical
More recent studies by Martin and co-workers have
microelectrode applications, electrochemically made PPy has
shown that neural cells can be incorporated into PEDOT,
a poorly dened chemical structure in which there are a sig-
while still maintaining cell viability and signal transduction
nicant amount of - couplings. The presence of defective
capabilities. As a result, functional hybrid PEDOT-neural cell
- couplings along the polymer backbone induce structural
electrode coatings were created that can be used as highly
disorder, limits the electrochemical response, and is con-
biomimetic conductive substrates for intracortical microelec-
tributing signicantly to polymer breakdown due to over-
trodes. Polymerization of PEDOT around living cells has
oxidation [314]. With these limitations in mind, new and been reported in vitro [318] as well as in vivo [319] through
highly stable conducting materials must be found that can living tissue. In one study, Richardson-Burns et al [318]
endure the long-term implantation lifetime as well as attack reported the electrochemical polymerization of PEDOT in the
from biological agents present in the brain tissue. presence of live neural cells that had been cultured on in-
To overcome the drawbacks of PPy, poly(3, 4-ethylene house fabricated Au/Pd sputter-coated electrodes and/or
dioxythiophene) (PEDOT) has recently been explored as an Applied Biophysics (Troy, NY) electrodes, as shown in
alternative to PPy for neural interfacing electrodes. Speci- gure 17, resulting in the formation of PEDOT lm around
cally, PEDOT is more stable to oxidation and more con- and onto adhered neural cells. Additionally, PEDOT,
ductive than PPy. Unlike PPy, undesired - couplings and PEDOT/live neurons, and neuron-templated PEDOT coatings
structural disorder are eliminated in PEDOT by blocking the on the electrodes signicantly enhanced the electrical prop-
3- and 4-positions of the monomer by the attachment of erties and increased charge transfer capacity. While in vitro
ethylenedioxy groups (gure 16). Early studies by Cui and experiments show successful electropolymerization of con-
Martin [315] explored the benets of PEDOT as coating for ducting polymers around neural cells, it is critical to apply the
neural microelectrodes. Cyclic voltammetry experiments concept in animal models to fully characterize the perfor-
demonstrated that PEDOT-coated electrodes were more stable mance of the materials to more accurately understand the
than those coated with PPy. In addition, high quality acute contribution of this strategy to the eld.
neural signals were recorded with the PEDOT-coated The rst chronic, long-term neural recording studies of
Michigan-style microelectrodes in the cerebellum of guinea PEDOT-coated electrodes were conducted by Kipke and co-
pig with higher signal amplitude than in reference experi- workers in 2006 [43, 320]. It was found that chronically
ments with un-coated microelectrodes with gold contacts. implanted control microelectrodes were unable to record well-

29
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

glycol) methacrylate (PEGMA) (gure 18(c)). Finally, a


carbon recording site was exposed at the tip of the neural
stainless-steel wire by cutting away the insulation, and the
recording site was coated with a layer of PEDOT:PSS that
was applied by electrochemical deposition (gure 18(d)).
Chronic neural recordings from the inserted implants into the
motor cortex of rats showed that the resulting microelectrodes
provided stable single-neuron recording over ve weeks in
the brain. Interestingly, the electrodes also showed reduced
neuro-inammation response compared with traditional sili-
con electrodes. Thus far, these electrodes with mechanically-
compliant coatings are the smallest implantable neural elec-
trodes that were able to record neuronal activity in animals.
Conducting polymers aim to enhance the chronic per-
formance of intracortical microelectrodes through providing a
high surface area, and more conductive materials. Also, the
charge transfer is likely improved through reduced impe-
dance, thereby providing greater selectivity for both recording
and stimulating neural applications. Despite the vast amount
of research being produced in recent years on the conducting
polymers for neural interfacing applications, the eld is still
growing and many challenges, limitations and questions
remain to be answered. One key challenge of conducting
polymers in neural interfacing applications is the preservation
of the conductivity or electrochemical stability over long
periods of time. For example, PPy and PEDOT lms have
been shown to lose up to 95% and 30% of their conductivity
when subjected to 16 h of polarization, respectively [322].
Also, conducting polymers are often brittle and the addition
Figure 17. (a) Schematic of the electrochemical deposition cell and of dopants to the system in many cases exacerbates this effect
the neural cell monolayer cultured on the surface of the metal [309]. Therefore, developing conducting polymers that are
electrode prior to polymerization. (b) PEDOT polymerized around less brittle and more malleable while maintaining the con-
living cells. Reprinted with permission from Richardson-Burns. ductivity and avoid delamination over time is required.
Reprinted with permission from [318], Copyright 2007 Elsevier.
Moreover, limited studies on the mechanical properties of
conducting polymers for the duration of implant lifetime
isolated unit activity due to a dramatically increased noise suggest that further research is needed to assess the durability
oor, while PEDOT-coated Michigan-style microelectrodes of a coated intracortical microelectrode in animal models.
consistently recorded neural activity, and showed a much This will provide an insight into possible delamination and/or
lower noise oor than un-coated gold-based controls over a mechanical erosion of conducting polymers. Another major
six-week period following implantation in three male drawback of conducting polymer systems is the diffusion of
rats [320]. the employed dopants, unreacted monomers and typical
More recently, Kozai et al took a further step towards process contaminations (e.g. solvents) into the medium,
long-lasting conjugated-polymer-based neural interfaces that which all are slightly to moderately toxic. Unfortunately,
elicit little tissue response in the brain, fabricating an inte- most of the studies that have revealed a positive biological
grated composite microelectrode consisting of electrically performance have been conducted in vitro. We believe that
conductive carbon ber core, a coating-based layer, and a the cytotoxicity of released dopants is likely a limiting factor
PEDOT:PSS-based recording pad [321]. Specically, elec- to the use of in-situ polymerization in vivo, despite the suc-
trodes were fabricated by mounting carbon ber, which cess in vitroat least with the chemistries commonly
serves as the conducting core and provides the mechanical explored. Thus, toxicity testing needs to be assessed using
backbone of the device, with a diameter of 7 m onto a in vivo implantation studies to answer whether conducting
NeuroNexus microelectrode. The carbon bers were then polymers are useful for long-term neural implants. Today,
coated with a 800 nm thin poly(p-xylylene) layer most/all of the studies have relied on electropolymerization of
(gure 18(a)), and subsequently a 50 nm thick layer of poly conducting polymers on conductive electrodes (e.g. Au). In
((p-xylylene-4-methyl-2-bromoisobutyrate)-co-(p-xylylene)), fact, the substrate to be coated needs to be conductive. So, the
both with a chemical vapor deposition (CVD) process conducting polymer is really a mediator between the brain
(gure 18(b)). The later polymer provides initiator groups for tissue and the conducting intracortical microelectrodes.
a subsequent atom transfer radical polymerization (ATRP). Therefore, it is also of particular interest to look at printed
This was used to apply a 200 nm thin layer of poly(ethylene polymer-based conductive microelectrodes for future neural

30
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

Figure 18. (a)(d) Schematic representation of the fabrication of microtherad electrodes, and (e) SEM images of a fully assembled, functional
electrode. Reprinted with permission from [321], Copyright 2012 Nature Publishing Group.

interfacing applications. Few studies have investigated how technology to long-term stable in vivo applications remains
conducting polymers tolerate sterilization. Given the corre- unclear. Finally, it is clear that long-term in vivo studies in
lation between sterilization method and the inammatory this area of research is required for a better understanding of
response to microelectrodes discussed above [160], and the the impact of conducting polymers in improving long-term
fact that not all formulations of PEDOT have the same dur- performance of intracortical microelectrodes include electro-
ability [323], the translation of any conducting polymer chemical stability, delamination, mechanical integrity, the

31
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

maintenance of an intimate contact between the electrode and the exploration of any new application for potential bioma-
surrounding neural tissue, and the ability to remove the terials, early work with CNTs towards intracortical micro-
implants without causing signicant damage to the sur- electrodes began with in vitro applications. For example,
rounding tissue. Mattson et al reported that multi-walled carbon nanotubes
(MWCNTs) could be used as platform for neuronal growth
4.8. Nanomaterials [336]. Since then, several studies have been devoted to
evaluate neuronal growth on CNTs [337341]. Wang et al
Nanomaterials have been explored in a variety of biomedical showed the rst in vitro stimulation of primary neurons with
applications due to their unique properties arising from their CNT-based electrodes [342]. The authors found that neurons
nanoscale dimensions [324326]. Nanomaterials can be use- can grow and differentiate on the microelectrode, and more
ful in cortical interfacing applications for several reasons importantly, that the neurons can be repeatedly stimulated
[327, 328]. Perhaps most importantly, nanomaterials can with CNT electrodes. In parallel, several other groups con-
interact with biological systems with a high degree of speci- rmed the possibility to stimulate the neural cells via single-
city, are able to stimulate and interact with target cells with walled and/or multi-walled carbon nanotubes in cultured
minimizing undesirable effects, and the electronic properties brain circuits [337, 343, 344]. In 2008, Keefer et al reported
of nanostructures can be tailored to match the needs asso- that CNT-coated metal microelectrodes improved both the
ciated with charge transport required for electrical/ionic level recording and electrical stimulation of neurons in culture, and
cellular interfacing. in vivo in rats and monkeys [345]. In vivo recording studies of
Charge transfer reactions involving the exchange of CNT-coated microelectrodes were conducted in the rat motor
charge between various carriers occur at the electrodepoly- cortex and monkey visual cortex. It was found that CNTs-
mer and polymertissue interfaces [329]. In particular, it has coated microelectrodes had increased the neuronal recording
been established that increasing the effective surface area of sensitivity as well as decreased neuronal noise compared to
the interface will increase the ability for charge transfer to un-coated tungsten microelectrode controls. It was possible to
occur. Also, the electrical impedance of the microelectrode is record LFPs, multiple unit activity, as well as neuronal
inversely proportional to the surface area of the recording site
spiking simultaneously with one CNT-coated microelectrode
[310]. As a result, the intrinsically large surface area of
in vivo. Additionally, Keefer et al found that the combination
nanomaterials results in high charge transfer as well as lower
of CNTs and the conducting polymer PPy increased the
overall impedance of the intracortical microelectrode.
charge transfer beyond that seen with CNTs alone. Further,
Additionally, nanoscience approaches can present sub-
CNT/PPy-coated microelectrodes had a signicantly lower
cellular stimuli that can vary from one part of the neuron to
impedance (higher electrode neuronal sensitivity) than bare
another [330]. For example, a combination of photo-
conventional tungsten and stainless steel microelectrodes. In a
lithography and layer-by-layer (LbL) self-assembly have been
similar study, Luo et al also reported that PEDOT/CNT-
used to pattern secreted phospholipase A2 (sPLA2), which
coated Pt microelectrodes showed a much lower impedance
promotes neuronal adhesion, on a non-fouling background of
poly(diallyldimethylammoniumchloride) (PDDA) [331]. The than the bare Pt electrodes as characterized by electro-
approach used by Mohammed et al facilitates nanoscale chemical impedance spectrum (EIS) in PBS [346]. Electro-
patterning with complex functional architectures that are tai- chemical impedance spectroscopy was used to show that the
lored to the needs of a particular experiment. LbL self- PEDOT/CNTs decreased the impedance of Pt microelec-
assembly has also been used by Ai et al on silicon rubber to trodes with increasing coating thickness due to the increase of
pattern alternating laminin and poly-D-lysine or bronectin/ the electroactive surface area, which results in a bigger
poly-D-lysine ultrathin layers which supported neurite out- capacitance. The PEDOT/CNT-coated electrodes exhibited
growth of cerebellar neurons [332]. Taken together, the stu- higher charge injection than traditional electrodes and in vitro
dies by Mohammad et al and Ai et al suggest that bioactive tests with neurons showed that the neurons attached tightly to
ultrathin coatings could be used to promote cell adhesion and the PEDOT/CNT surface and exhibited long neurite exten-
limit immune responses, and may facilitate improved per- sions, suggesting that PEDOT/CNT-coatings are non-neuro-
formance of intracortical microelectrodes. toxic and support the growth of neurons. Similar conducting
Additionally, several types of nanomaterials have been polymer/CNT composite coatings were also studied by sev-
utilized in neural interface devices, including carbon nano- eral other groups [347, 348]. Lu et al reported that co-
tubes, carbon nanobers, and graphene. The use of nanoma- deposited PPy/SWCNT coatings signicantly reduce the
terials and nanotools for neuroscience has been recently impedance of platinum/tungsten microelectrodes. Addition-
reviewed [327, 333]. In this Section, we therefore limit the ally, the brain tissue response of PPy/SWCNT coated
discussion to the current state of the most widely investigated microelectrodes and un-coated Pt microelectrodes were stu-
nanomaterials in the content of neural interfacing died with immunochemistry after a six-week implantation in
applications. the cortex of rats. Quantitative analysis of glial brillary
Carbon nanotubes (CNTs) are perhaps the most widely acidic protein (GFAP) expression and neuronal nuclei (NeuN)
studied class of nanomaterials for intracortical microelec- showed signicantly lower GFAP and higher NeuN counts
trodes [327, 334, 335], in view of their extraordinary strength, for the PPy/SWCNT coated microelectrodes within the rst
toughness, electrical conductivity, and surface area. As with 100 m from the implant/tissue interface [348].

32
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

Despite the perspective advantages of CNTs for intra- neural-electrical interface can be formed between the verti-
cortical microelectrode applications, the studied devices are cally aligned CNFs and a neuronal network of PC12 cells
rigid (non-compliant) and may not be optimally suited for [355]. The addition of neuronal growth factor (NGF) on the
chronic neural in vivo applications. As a result, there is an vertically aligned CNFs facilitated the formation of well-dif-
increased attention towards the development of exible ferentiated cells with mature neurites. Although, the free-
CNTs-based intracortical microelectrodes by combination of standing CNFs coated with PPy and NGF were mechanically
exible polymeric substrates and CNTs to overcome both the rigid to maintain their vertical alignment, they were found to
mechanical failure of more brittle CNTs-based devices, and be exible enough to bend toward the cell body when driven
device-mediated tissue strain. Flexible CNT-based neural by traction forces of the cells, thereby facilitating cell adhe-
electrodes were created by the combination of exible poly- sion. Thus, it was suggested that the soft PPy coating not only
meric substrates and CNT-based electrodes. Lin et al were the improved the mechanical stability by forming a core-shell
rst to fabricate a exible CNT-based electrode array for structure with the CNFs, but also promotes a better
neural recording applications [349]. In this work, the CNT mechanical contact with neuronal cells due to a reduction of
electrodes were partially embedded into a exible Parylene-C the local mechanical stresses [355].
lm using a microfabrication process based on four steps: Several studies suggest that graphene exhibits excellent
CNT growth, polymer binding, exible lm transfer, and biocompatibility, low cytotoxicity and supports neuronal
partial isolation. The exible CNT electrodes produced were growth [356358]. Therefore, graphene is another carbon
used to successfully record the spontaneous spikes from a nanomaterial that has been recently utilized in neural interface
craysh nerve cord. application. Chen et al [359] reported the fabrication of gra-
Alternatively, the direct growth of CNTs on exible phene-based neural microelectrodes, while Luo et al [360]
polyimide substrates has been reported by Hsu et al [350]. reported graphene oxide-based conducting polymer nano-
Hsu et al utilized UV-ozone exposure as a simple and low- composites for potential neural interfacing applications.
cost route to improve the interfacial properties between the Chens study showed that graphene-based microelectrodes
CNT electrodes. In culture, UV-ozone treated CNTs elec- are capable of recording neural signals in a craysh. These
trodes promoted neuron and neurite growth in close contact graphene electrodes showed biocompatibility and non-toxi-
with CNTs, suggesting the biocompatibility of modied city throughout the 16 day cell culture experiment with
CNTs for neuronal growth [350]. In a subsequent study, neuronal cells. In the second investigation, Luo et al elec-
spontaneous spikes were recorded from a craysh with a trochemically doped PEDOT with graphene oxide (GO) and
signal-to-noise (SNR) ratio of 6.2. The exible CNT elec- demonstrated that conducting PEDOT/GO nanocomposites
trodes were also used to record the electrocorticography (i.e. supported the growth of neural cells with minimal toxicity
placing the electrode in the subdural region) of a rat motor along with low electrochemical impedance [360]. To clearly
cortex with a SNR of 8.7 [351]. More examples of CNT- demonstrate the potential of this class of graphene-based
based exible electrodes for neural recording and simulating materials for neural microelectrodes, future studies need to
applications have been developed by Hanein et al investigate the chronic in vivo performance dened by both
[335, 352, 353] who transferred single-walled carbon nano- the ability to maintain a clinical viable signal quality and
tubes (SWCNTs) onto a exible PDMS substrate. Recent stimulation capabilities, as well as the biotic and abiotic
evoked electrical activity recording studies with chick retinas failure modes (dened above). Alternatively, the Martin
demonstrated the device capability for high efcacy neuronal group designed multifunctional nanobiomaterials that can be
recording applications [335]. used for coating neural microelectrodes [264, 361]. These
Carbon nanobers (CNFs), which consist of multi-walled materials signicantly decrease the electrode impedance and
graphene structures stacked on top of each other have also increase the charge density. While in vivo data are in process,
been explored as substrates for neural interfacing applica- and to our knowledge yet to be reported, the approach nicely
tions. Researchers at NASA Ames Research Center devel- demonstrates how several attractive concepts can be merged
oped forest-like vertically aligned CNFs on a Si wafer by to create smart nanobiomaterials that are soft, have a low
plasma enhanced chemical vapor deposition (PECVD) [354]. impedance, high charge density, and also can deliver anti-
After the CNF lm was submerged in a liquid and dried, the inammatory drugs to alleviate the brain immune response to
CNFs irreversibly stuck together to form microbundles. neural interfaces.
Stable 3D fuzzy lms were created by electrochemically Together with the Kipke group, Martins group also
coating the CNFs with a thin layer of electrically conducting reported the use of conducting polymer nanotubes for chronic
polypyrrole (PPy). It was found that the impedance of this neural interfaces [362]. This work showed that PEDOT
kind of electrode decreased signicantly compared to com- nanotube-coated electrodes have a markedly lower impedance
mon metal electrodes due to the large surface area of 3D over a seven-week period than conventional Michigan-style
nanostructured CNFs, which results in high ion mobility. microelectrodes. Figure 19 shows the fabrication process of
Furthermore, the PPy coating was shown to improve the such PEDOT nanotubes on the surface of Michigan-style
biocompatibility of CNF-based electrodes compared to un- microelectrodes. Martin and Kipke later investigated the
coated, and further reduces the electrode impedance by more effect of nanotube morphology on the properties of the
than 20 times due to the redox potentials of the polymer. A electrodes in vitro [363]. Their work demonstrated that the
subsequent study by the same group showed that an intimate PEDOT nanotubes decreased the impedance of the electrode

33
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

Figure 19. Schematic illustration of conducting polymer (PEDOT) nanotube fabrication on neural microelectrodes: (a), (b) electrospining of
polylactic acid (PLLA) nanobers. (c) Electrochemical deposition of PEDOT. (d) Dissolution of the PLLA core. (e), (f) Optical microscopy
images of the entire microelectrode (e) and single electrode site (f) before surface modication. (g), (h) Optical microscopy images of the
entire microelectrode (g) and single electrode site (h) after electrospining of PLLA nanobers. (i), (j) Optical microscopy images of the entire
microelectrode (i) and single electrode site (j) after electrochemical deposition of PEDOT and remove of the PLLA core. Reprinted with
permission from [362], Copyright 2009 John Wiley and Sons.

site by about two orders of magnitude, and increased the other devices discussed above, it remains unclear how func-
capacity of charge density by about three orders of magnitude tional devices of this nature can be removed from patients
compared to bare iridium microelectrodes [363]. The team without damaging surrounding tissue, or breaking the device
further showed that the mechanical properties of the con- off in the cortical tissue, due to their size and brittleness. We
ducting polymer can be tuned by their surface morphology. believe that there are still tremendous opportunities for
For instance, it was demonstrated that PPy and PEDOT nanomaterials to contribute to neural interfacing devices to
nanotubes can adhere better to the surface of the neural generate intracortical microelectrodes that can enhance, and
electrodes than lms of these polymers. Despite the many improve the current technologies. Thus, herein, we present a
incredibly attractive materials properties that nanomaterials brief outline of the most important future areas of nanoma-
have demonstrated towards long-term neural interfaces, sev- terials in the chronic neural interfacing applications that need
eral of the devices created from these materials in their current to be investigated:
form still suffer from limitation that have chronically plagued
neural electrodes (degradation and delamination). Therefore, (1) Integration of traditional approaches and nanomaterials
the combination approaches, such as thus being developed by to design new types of intracortical microelectrodes.
Martin and colleagues, represent the starting point towards (2) Development of new types of nanostructured coatings
integrating nanomaterials into intracortical microelectrodes offered by nanomaterials that could also increase the
for neural interfacing applications. charge injection capacity and reduction of impedance.
Recent research activities focused on use of nanomater- (3) Incorporation of anti-inammatory agents in the coat-
ials in the domain of neural interfacing applications has ings using layer-by-layer self-assembly technique.
contributed signicantly to our understanding of the devel- (4) Miniaturization of the traditional intracortical micro-
opment of better biocompatible intracortical microelectrodes. electrodes using smaller and more compliant exible
The application of nanomaterials in chronic neural interfaces nanomaterials, while maintaining efcient electroche-
is still in its infancy despite an impressive body of research mical function.
that is emerging, partly because of the complexities associated (5) Improvement of electrical and biological properties of
with interacting with neural cells and the mammalian nervous the neural tissue-electrode interface.
system. Moreover, there are still fundamental gaps of (6) Design of easily manufacturable, highly conductive,
knowledge regarding the potential toxicity of nanomaterials and mechanically strong and exible intracortical
within the brain that need to be addressed. Additionally, like

34
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

electrodes using nanomaterials, and conductive and not provide a global solution to all electrode types or to all
adaptive polymers. recording applications.
(7) Investigation the chronic long-term potential toxicity It is also important to note that despite the vast amount of
proles as well as delamination and degradation of research being developed in the last decade on polymeric
nanomaterials over the period of implantation. materials for neural interfacing applications, in most cases,
(8) Comprehensive long-term recording and stimulation researchers have simply used polymers developed for differ-
studies of nanomaterials in animal models. ent purposes, with the aim to re-use them for neural micro-
electrodes. Although the development of such materials-based
Finally, these exciting avenues must be tempered with
intracortical microelectrodes has contributed signicantly to
the realization that the toxicity of nanomaterials is still a
our understanding of how improve device integration and
developing eld, and that a better understanding of how
increase recording consistency, designing novel materials,
nanoscale materials interact with the central nervous system is
strategies and concepts for the specic purposes of this eld
required before one can use nanomaterials widely in neural
must be accelerated.
interfacing applications.
Despite the challenges and questions that remain, we
should be encouraged by the exciting possibilities that will be
opened by further developing our understanding of the
5. Conclusions mechanisms that inuence microelectrode performance. Fur-
thermore, due to the exciting advances in the elds of material
Undoubtedly, the recordings of consciously modulated neu- science, neural engineering and bioengineering, we should
ronal signals using intracortical microelectrodes have foster dynamic multi-disciplinary teams, in order to accu-
advanced our fundamental understanding of brain function in mulate the skills and knowledge to design, test, and integrate
both normal and diseased states. In addition, chronic micro- the next generation intracortical microelectrodes, capable of
electrode recordings provide a way for paralyzed or locked long-term clinical deployment for neuro-rehabilitative appli-
in individuals to directly control various assistive devices cations, and beyond.
through brain machine interfacing. Unfortunately, the imple-
mentation of brain machine interfacing applications has been
severely hindered due to inconsistent recording quality and
premature electrode failure. Acknowledgments
Poor recording quality and microelectrode failure are
likely a function of an array of variables resulting from both The authors gratefully acknowledge nancial support from
biotic and abiotic factors. The high degree of complexity Adolphe Merkle Foundation, the Swiss National Science
makes improving microelectrode performance a challenging Foundation (Weder, NRP 62: Smart Materials, Nr.
problem. While a number of failure modes have been iden- 406240_126046), the Department of Veterans Affairs
tied, a more in-depth mechanistic understanding is needed. (Capadona, Rehabilitation Research and Development:
A number of strategies have been studied to improve B7122R and B1495-R), Presidential Early Career Award for
microelectrode performance. Such strategies include those to Scientist and Engineers (Capadona, Rehabilitation Research
improve the stability of conducting and insulating materials as and Development: PECASE), and the Nation Institute of
well as those to improve microelectrode integration with the Health (Capadona, National Institute of Neurological Dis-
surrounding tissue. As neuro-inammation has been shown to orders and Stroke, 1R01NS082404-01A1). We also thank M
play a signicant role in each of the major microelectrode Ravikumar, K Potter-Baker, and J Nguyen for assistance in
failure modes, specic emphasis should be given to reduce gure generation. The authors have no conicts of interest
the reactive environment that results following microelectrode related to this work to disclose. None of the funding sources
implantation. aided in the collection, analysis and interpretation of data, in
Due to the variety of failure modes and the large degree the writing of the review, or in the decision to submit the
of interplay involved, it is increasingly evident that a com- paper for publication.
bination of strategies may be necessary to enable consistent,
high quality recording over a clinically relevant time frame.
To optimally implement and integrate such strategies it will
be necessary to further identify the most potent strategies for References
improving microelectrode performance. It will also be useful
to isolate possible interactions or emergent phenomena to [1] Grundfest H C B 1942 Origin, conduction and termination of
elucidate the overall design space available to engineers. Most impulses in the dorsal spino-cerebellar tract of cats
importantly, we must all consider that differences in micro- J. Neurophysiol. 5 27594
electrode design may be a major contributing source for [2] Grundfest H S R, Oettinger W H and Gurry R W 1950
discrepancies between the dominant failure mechanisms Stainless steel micro-needle electrodes mae by electrolytic
pointing Rev. Sci. Instrum. 21 3602
reported for traditional designs. Each class of microelectrode [3] Renshaw B F A and Morison B R 1940 Activity of isocortex
presents with a different primary failure mode, and time and hippocampus: electrical studies with micro-electrodes
course for failure. Therefore, one approach will most likely J. Neurophysiol. 3 74105

35
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

[4] Pancrazio J J and Peckham P H 2009 Neuroprosthetic [26] Wolbarsht M L, Macnichol E F Jr. and Wagner H G 1960
devices: how far are we from recovering movement in Glass insulated platinum microelectrode Science 132
paralyzed patients? Expert Rev. Neurotherapeutics 9 42730 130910
[5] Nicolelis M A L 2003 Brainmachine interfaces to restore [27] Evarts E V 1966 Pyramidal tract activity associated with a
motor function and probe neural circuits Nat. Rev. Neurosci. conditioned hand movement in the monkey J. Neurophysiol.
4 41722 29 101127
[6] Hochberg L R et al 2012 Reach and grasp by people with [28] Salcman M and Bak M J 1973 Design, fabrication, and in vivo
tetraplegia using a neurally controlled robotic arm. Nature behavior of chronic recording intracortical microelectrodes
485 3725 IEEE Trans. Biomed. Eng. 20 25360
[7] Hochberg L R, Serruya M D, Friehs G M, Mukand J A, [29] Salcman M and Bak M J 1976 A new chronic recording
Saleh M, Caplan A H, Branner A, Chen D, Penn R D and intracortical microelectrode Med. Biol. Eng. 14 4250
Donoghue J P 2006 Neuronal ensemble control of prosthetic [30] Schmidt E M, Bak M J and McIntosh J S 1976 Long-term
devices by a human with tetraplegia Nature 442 16471 chronic recording from cortical neurons Exp. Neurol. 52
[8] Kennedy P R and Bakay R A 1998 Restoration of neural 496506
output from a paralyzed patient by a direct brain connection [31] Burns B D, Stean J P and Webb A C 1974 Recording for
Neuroreport. 9 170711 several days from single cortical neurons in completely
[9] Kennedy P R, Bakay R A, Adams K and Goldwaithe J 2000 unrestrained cats Electroencephalogr. Clin. Neurophysiol.
Direct control of a computer from the human central nervous 36 3148
system IEEE Trans. Rehabil. Eng. 8 198202 [32] Schmidt E M, McIntosh J S and Bak M J 1988 Long-term
[10] Kennedy P R, Kirby M T, Moore M M, King B and implants of Parylene-C coated microelectrodes Med. Biol.
Mallory A 2004 Computer control using human intracortical Eng. Comput. 26 96101
local eld potentials IEEE Trans. Neur. Sys. Reh. Sep. 12 [33] Kruger J, Caruana F, Volta R D and Rizzolatti G 2010 Seven
33944 years of recording from monkey cortex with a chronically
[11] Kellis S, Miller K, Thomson K, Brown R, House P and implanted multiple microelectrode Front. Neuroeng. 3 6
Greger B 2010 Classication of spoken words using surface [34] Prasad A, Xue Q-S, Sankar V, Nishida T, Shaw G,
local eld potentials Conf. Proc. IEEE Eng. Med. Biol. Soc. Streit W J and Sanchez J C 2012 Comprehensive
2010 382730 characterization and failure modes of tungsten microwire
[12] Kellis S, Miller K, Thomson K, Brown R, House P and arrays in chronic neural implants J. Neural. Eng. 9
Greger B 2010 Decoding spoken words using local eld 056015
potentials recorded from the cortical surface J Neural Eng. 7 [35] Prasad A, Xue Q S, Dieme R, Sankar V, Mayrand R C,
056007 Nishida T, Streit W J and Sanchez J C 2014 Abiotic-biotic
[13] Lebedev M A and Nicolelis M A 2006 Brain-machine characterization of Pt/Ir microelectrode arrays in chronic
interfaces: past, present and future Trends Neurosci. 29 implants Front. Neuroeng. 7 2
53646 [36] Wise K D, Angell J B and Starr A 1970 An integrated-circuit
[14] Ward M P, Rajdev P, Ellison C and Irazoqui P P 2009 approach to extracellular microelectrodes IEEE Trans.
Toward a comparison of microelectrodes for acute and Biomed. Eng. 17 23847
chronic recordings Brain. Res. 1282 183200 [37] Wise K D and Angell J B 1975 A low-capacitance
[15] Tresco P A and Winslow B D 2011 The challenge of multielectrode probe for use in extracellular
integrating devices into the central nervous system Crit. Rev. neurophysiology IEEE Trans. Biomed. Eng. 22 2129
Biomed. Eng. 39 2944 [38] BeMent S L, Wise K D, Anderson D J, Naja K and
[16] Liu X, McCreery D B, Bullara L A and Agnew W F 2006 Drake K L 1986 Solid-state electrodes for multichannel
Evaluation of the stability of intracortical microelectrode multiplexed intracortical neuronal recording IEEE Trans.
arrays IEEE Eng. Med. Biol. Soc. 14 91100 Biomed. Eng. 33 23041
[17] Liu X, McCreery D B, Carter R R, Bullara L A, [39] Wise K D 2005 Silicon microsystems for neuroscience and
Yuen T G and Agnew W F 1999 Stability of the interface neural prostheses IEEE Eng. Med. Biol. Mag. 24 229
between neural tissue and chronically implanted intracortical [40] Hetke J F, Lund J L, Naja K, Wise K D and Anderson D J
microelectrodes IEEE Trans. Rehabil. Eng. 7 31526 1994 Silicon ribbon cables for chronically implantable
[18] Nelson M J, Pouget P, Nilsen E A, Patten C D and Schall J D microelectrode arrays IEEE Trans. Biomed. Eng. 41
2008 Review of signal distortion through metal 31421
microelectrode recording circuits and lters J Neurosci. [41] Kipke D R, Vetter R J, Williams J C and Hetke J F 2003
Methods. 169 14157 Silicon-substrate intracortical microelectrode arrays for
[19] Robinson D A 1968 The electrical properties of metal long-term recording of neuronal spike activity in cerebral
microelectrodes Proc. IEEE. 56 106571 cortex IEEE Trans. Neural Syst. Rehab. Eng.: Publ. IEEE
[20] Buzski G 2004 Large-scale recording of neuronal ensembles Eng. Med. Biol. Soc. 11 1515
Nat. Neurosci. 7 44651 [42] Vetter R J, Williams J C, Nunamaker E A and Kipke D R
[21] Tillery S I H and Taylor D M 2004 Signal acquisition and 2004 Chronic neural recording using silicon-substrate
analysis for cortical control of neuroprosthetics Curr. Opin. microelectrode arrays implanted in cerebral cortex IEEE
Neurobiol. 14 75862 Trans. Biomed. Eng. 51 896904
[22] Taylor D M, Tillery S I and Schwartz A B 2002 Direct [43] Ludwig K A, Uram J D, Yang J, Martin D C and Kipke D R
cortical control of 3D neuroprosthetic devices Science 296 2006 Chronic neural recordings using silicon microelectrode
182932 arrays electrochemically deposited with a poly(3,4-
[23] Gilja V, Chestek C A, Diester I, Henderson J M, ethylenedioxythiophene) (PEDOT) lm J. Neural. Eng. 3
Deisseroth K and Shenoy K V 2011 Challenges and 5970
opportunities for next-generation intracortically based neural [44] Campbell P K, Jones K E, Huber R J, Horch K W and
prostheses IEEE Trans. Bio-Med. Eng. 58 18919 Normann R A 1991 A silicon-based, three-dimensional
[24] Donoghue J 2008 Bridging the brain to the world: a neural interface: manufacturing processes for an intracortical
perspective on neural interface systems Neuron. 60 51121 electrode array IEEE Trans. Biomed. Eng. 38 75868
[25] Hubel D H 1957 Tungsten microelectrode for recording from [45] Negi S, Bhandari R, Rieth L and Solzbacher F 2010 In vitro
single units Science 125 54950 comparison of sputtered iridium oxide and platinum-coated

36
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

neural implantable microelectrode arrays Biomed. Mater. 5 [64] Barrese J C, Rao N, Paroo K, Triebwasser C, Vargas-Irwin C,
15007 Franquemont L and Donoghue J P 2013 Failure mode
[46] Hsu J M, Rieth L, Normann R A, Tathireddy P and analysis of silicon-based intracortical microelectrode arrays
Solzbacher F 2009 Encapsulation of an integrated neural in non-human primates J. Neural Eng. 10 066014
interface device with Parylene C IEEE Trans. Biomed. Eng. [65] Merrill D R 2010 The electrochemistry of charge injection at
56 239 the electrode/tissue interface Implantable Neural Prostheses
[47] Bhandari R, Negi S and Solzbacher F 2010 Wafer-scale 2 (Berlin: Springer) pp 85138
fabrication of penetrating neural microelectrode arrays [66] Patrick E, Orazem M E, Sanchez J C and Nishida T 2011
Biomed. Microdevices. 12 797807 Corrosion of tungsten microelectrodes used in neural
[48] Branner A S R and Normann R A 2001 Selective stimulation recording applications J. Neurosci. Methods 198 15871
of cat sciatic nerve using an array of varying-length [67] McCarthy P T, Otto K J and Rao M P 2011 Robust
microelectrodes J Neurophysiol. 85 158594 penetrating microelectrodes for neural interfaces realized
[49] Bhandari R, Negi S, Rieth L, Normann R A and by titanium micromachining Biomedical Microdev. 13
Solzbacher F A 2008 Novel method of fabricating 50315
convoluted shaped electrode arrays for neural and retinal [68] McDord J M and Fridovich I 1969 Superoxide dismutase
prostheses Sensors Actuators A 145-146 12330 J. Biol. Chem. 244 604955
[50] Bhandari R, Negi S, Rieth L and Solzbacher F A 2010 Wafer- [69] Potter K A, Buck A C, Self W K, Callanan M E, Sunil S and
scale etching technique for high aspect ratio implantable Capadona J R 2013 The effect of resveratrol on
MEMS structures. Sensors Actuators A 162 1306 neurodegeneration and blood brain barrier stability
[51] Wark H A C et al 2013 A new high-density (25 electrodes/ surrounding intracortical microelectrodes Biomaterials 34
mm2) penetrating microelectrode array for recording and 700115
stimulating sub-millimeter neuroanatomical structures [70] Potter K A, Jor M, Householder K T, Foster E J,
J. Neural. Eng. 10 Weder C and Capadona J R 2014 Curcumin-releasing
[52] Harrison R R, Kier R J, Chestek C A, Gilja V, Nuyujukian P, mechanically-adaptive intracortical implants improve the
Ryu S, Greger B, Solzbacher F and Shenoy K V 2009 proximal neuronal density and blood-brain barrier stability
Wireless neural recording with single low-power integrated Acta Biomater. 10 220922
circuit IEEE Eng. Med. Biol. Soc. 17 3229 [71] Dymond A M, Kaechele L E, Jurist J M and Crandall P H
[53] Kim S, Bhandari R, Klein M, Negi S, Rieth L, Tathireddy P, 1970 Brain tissue reaction to some chronically implanted
Toepper M, Oppermann H and Solzbacher F 2009 Integrated metals J. Neurosurg. 33 57480
wireless neural interface based on the Utah electrode array [72] Wang A et al 2007 Stability of and inammatory response to
Biomed. Microdev. 11 45366 silicon coated with a uoroalkyl self-assembled monolayer
[54] Abaya T V, Diwekar M, Blair S, Tathireddy P, Rieth L, in the central nervous system J. Biomed. Mater. Res. A 81
Clark G A and Solzbacher F 2012 Characterization of a 3D 36372
optrode array for infrared neural stimulation Biomed. Opt. [73] Hammerle H, Kobuch K, Kohler K, Nisch W, Sachs H and
Express. 3 220019 Stelzle M 2002 Biostability of micro-photodiode arrays for
[55] Warren D J, Fernandez E and Normann R A 2001 High- subretinal implantation Biomaterials 23 797804
resolution two-dimensional spatial mapping of cat striate [74] Maloney J M L S and Baldwin S P (ed) 2005 In vivo
cortex using a 100-microelectrode array Neuroscience 105 biostability of CVD silicon oxide and silicon nitride lms
1931 MRS Proc. 872 J14.3
[56] Kim S J, Manyam S C, Warren D J and Normann R A 2006 [75] Schmitt J-WS G, Fabender F, Bu G, Lth H and
Electrophysiological mapping of cat primary auditory cortex Schning M J 1999 Passivation and corrosion of
with multielectrode arrays Ann. Biomed. Eng. 34 3009 microelectrode arrays Electrochimica Acta 44 386583
[57] Suner S, Fellows M R, Vargas-Irwin C, Nakata G K and [76] Cogan S F E D, Guzelian A A, Ping Liu Y and Edell R 2003
Donoghue J P 2005 Reliability of signals from a chronically Plasma-enhanced chemical vapor deposited silicon carbide
implanted, silicon-based electrode array in non-human as an implantable dielectric coating J. Biomed. Mater. Res.
primate primary motor cortex IEEE Trans. Rehab. Eng. 13 A 67 85667
52441 [77] Loeb G E, Bak M J, Salcman M and Schmidt E M 1977
[58] Schwartz A B, Kettner R E and Georgopoulos A P 1988 Parylene as a chronically stable, reproducible microelectrode
Primate motor cortex and free arm movements to visual insulator IEEE Trans. Biomed. Eng. 24 1218
targets in three-dimensional space: part 1. Relations between [78] Winslow B D, Christensen M B, Yang W K,
single cell discharge and direction of movement J. Neurosci. Solzbacher F and Tresco P A 2010 A comparison of the
Methods 8 291327 tissue response to chronically implanted Parylene-C-coated
[59] Rousche P J and Normann R A 1998 Chronic recording and uncoated planar silicon microelectrode arrays in rat
capability of the utah intracortical electrode array in cat cortex Biomaterials 31 916372
sensory cortex J. Neurosci. Methods 82 115 [79] Bjornsson C S, Oh S J, Al-Kofahi Y A, Lim Y J, Smith K L,
[60] Kennedy P R 1989 The cone electrode: a long-term electrode Turner J N, De S, Roysam B, Shain W and Kim S J 2006
that records from neurites grown onto its recording surface Effects of insertion conditions on tissue strain and vascular
J. Neurosci. Methods 29 18193 damage during neuroprosthetic device insertion J. Neural
[61] David S and Aguayo A J 1981 Axonal elongation into Eng. 3 196207
peripheral nervous system bridges after central nervous [80] House P A, Macdonald J D, Tresco P A and Normann R A
system injury in adult rats. Science 214 9313 2006 Acute microelectrode array implantation into human
[62] Kennedy P R and Bakay R A 1997 Activity of single action neocortex: preliminary technique and histological
potentials in monkey motor cortex during long-term task considerations Neurosurg. Focus 20 E4
learning Brain Res. 760 2514 [81] Hanson S R, Harker L A, Ratner B D and Hoffman A S 1980
[63] Freire M A, Morya E, Faber J, Santos J R, Guimaraes J S, In vivo evaluation of articial surfaces with a nonhuman
Lemos N A, Sameshima K, Pereira A, Ribeiro S and primate model of arterial thrombosis J. Lab. Clin. Med. 95
Nicolelis M A 2011 Comprehensive analysis of tissue 289304
preservation and recording quality from chronic [82] Tang L 1998 Mechanisms of brinogen domains: biomaterial
multielectrode implants PLoS One. 6 e27554 interactions J Biomater. Sci. Polym. Ed. 9 125766

37
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

[83] Nilsson B, Ekdahl K N, Mollnes T E and Lambris J D 2007 [102] Shanmuganathan K, Capadona J R, Rowan S J and Weder C
The role of complement in biomaterial-induced 2010 Bio-inspired mechanically-adaptive nanocomposites
inammation Mol. Immunol. 44 8294 derived from cotton cellulose whiskers J. Mater. Chem.
[84] Anderson J M 2001 Biological responses to materials Annu. 20 180
Rev. Mater. Res. 31 81110 [103] Shanmuganathan K, Capadona J R, Rowan S J and Weder C
[85] Anderson J M, Rodriguez A and Chang D T 2008 Foreign 2010 Biomimetic mechanically adaptive nanocomposites
body reaction to biomaterials Semin. Immunol. 20 86100 Prog. Polym. Sci. 35 21222
[86] Pollock A L 2007 Early Brain Tissue Reaction Surrounding [104] Ware T, Simon D, Liu C, Musa T, Vasudevan S, Sloan A,
the Michigan-Style Microelectrode Arrays Indicates that Keefer E W, Rennaker R L and Voit W 2014 Thiolene/
Extensive Remodeling of Glial and Neuronal Populations acrylate substrates for softening intracortical electrodes
Occurs During the Foreign Body Response (Salt Lake City: J. Biomed. Mater. Res. B: Appl. Biomater. 102
Department of Bioengineering) 111
[87] Potter K A, Simon J S, Velagapudi B and Capadona J R 2012 [105] Jor M, Roberts M N, Foster E J and Weder C 2013
Reduction of autouorescence at the microelectrode-cortical Physiologically responsive, mechanically adaptive bio-
tissue interface improves antibody detection J. Neurosci. nanocomposites for biomedical applications ACS Appl.
Methods 203 96105 Mater. Interfaces 5 151726
[88] Mahi H and Rodrigue D 2012 Linear and non-linear [106] Polikov V S, Tresco P A and Reichert W M 2005 Response of
viscoelastic properties of ethylene vinyl acetate/nano- brain tissue to chronically implanted neural electrodes
crystalline cellulose composites Rheologica Acta. 51 12742 J. Neurosci. Methods 148 118
[89] Subbaroyan J, Martin D C and Kipke D R 2005 A nite- [107] Biran R, Martin D C and Tresco P A 2005 Neuronal cell loss
element model of the mechanical effects of implantable accompanies the brain tissue response to chronically
microelectrodes in the cerebral cortex J. Neural Eng. 2 implanted silicon microelectrode arrays Exp. Neurol. 195
10313 11526
[90] Kim D H, Abidian M and Martin D C 2004 Conducting [108] Biran R, Martin D C and Tresco P A 2007 The brain tissue
polymers grown in hydrogel scaffolds coated on neural response to implanted silicon microelectrode arrays is
prosthetic devices J. Biomed. Mater. Res. A 71 57785 increased when the device is tethered to the skull J. Biomed.
[91] Hess A, Capadona J, Shanmuganathan K, Hsu L, Rowan S, Mater. Res. A 82 16978
Weder C, Tyler D and Zorman C 2011 Development of a [109] Potter K A, Buck A C, Self W K and Capadona J R 2012 Stab
stimuli-responsive polymer nanocomposite toward injury and device implantation within the brain results in
biologically-optimized, MEMS-based neural probes inversely multiphasic neuroinammatory and
J. Micromech. Microeng. 21 5400917 neurodegenerative responses J. Neural. Eng. 9 046020
[92] Hess A, Dunning J, Harris J, Capadona J R, [110] Ravikumar M, Jain S, Miller R H, Capadona J R and
Shanmuganathan K, Rowan S J, Weder C, Tyler D J and Selkirk S M 2012 An organotypic spinal cord slice culture
Zorman C A 2009 A bio-inspired, chemo-responsive model to quantify neurodegeneration J. Neurosci. Methods
polymer nanocomposite for mechanically dynamic 211 2808
microsystems Solid-State Sensors, Actuators and [111] Gehrmann J, Matsumoto Y and Kreutzberg G W 1995
Microsystems Conf. Transducers 2009 Int. (Denver, 21-25 Microgliaintrinsic immuneffector cell of the brain Brain
June 2009) CO2009 pp 2247 Res. Rev. 20 26987
[93] Ware T, Simon D, Arreaga-Salas D E, Reeder J, Rennaker R, [112] Betjes M G, Tuk C W, Struijk D G, Krediet R T, Arisz L and
Keefer E W and Voit W 2012 Fabrication of responsive, Beelen R H 1993 Antigen-presenting capacity of
softening neural interfaces Adv. Funct. Mater 22 34709 macrophages and dendritic cells in the peritoneal cavity of
[94] Harris J P, Hess A E, Rowan S J, Weder C, Zorman C A, patients treated with peritoneal dialysis Clin. Exp. Immunol.
Tyler D J and Capadona J R 2011 In vivo deployment of 94 37784
mechanically adaptive nanocomposites for intracortical [113] Gregerson D S, Sam T N and McPherson S W 2004 The
microelectrodes J. Neural. Eng. 8 046010 antigen-presenting activity of fresh, adult parenchymal
[95] Goldstein S R and Salcman M 1973 Mechanical factors in the microglia and perivascular cells from retina J. Immunol. 172
design of chronic recording intracortical microelectrodes 658797
IEEE Trans. Biomed. Eng. 20 2609 [114] Mack C L, Vanderlugt-Castaneda C L, Neville K L and
[96] Sridharan A, Rajan S D and Muthuswamy J 2013 Long-term Miller S D 2003 Microglia are activated to become
changes in the material properties of brain tissue at the competent antigen presenting and effector cells in the
implanttissue interface J. Neural Eng. 10 066001 inammatory environment of the Theiler's virus model of
[97] Hess A E, Potter K A, Tyler D J, Zorman C A and multiple sclerosis J. Neuroimmunol. 144 6879
Capadona J R 2013 Environmentally-controlled microtensile [115] Poltorak M and Freed W J 1989 Immunological reactions
testing of mechanically-adaptive polymer nanocomposites induced by intracerebral transplantation: evidence that host
for ex vivo characterization JoVE 78 e50078 microglia but not astroglia are the antigen-presenting cells
[98] Harris J P, Capadona J R, Miller R H, Healy B C, Exp. Neurol. 103 22233
Shanmuganathan K, Rowan S J, Weder C and Tyler D J 2011 [116] Shaked I, Porat Z, Gersner R, Kipnis J and Schwartz M
Mechanically adaptive intracortical implants improve the 2004 Early activation of microglia as antigen-presenting
proximity of neuronal cell bodies J. Neural. Eng. 8 066011 cells correlates with T cell-mediated protection and repair
[99] Capadona J R, Tyler D J, Zorman C A, Rowan S J and of the injured central nervous system J. Neuroimmunol 146
Weder C 2012 Mechanically adaptive nanocomposites for 8493
neural interfacing MRS Bull. 37 5819 [117] Skousen J, Merriam S, Srivannavit O, Perlin G, Wise K and
[100] Capadona J R, Shanmuganathan K, Tyler D J, Rowan S J and Tresco P 2011 Reducing surface area while maintaining
Weder C 2008 Stimuli-responsive polymer nanocomposites implant penetrating prole lowers the brain foreign body
inspired by the sea cucumber dermis Science 319 1370 response to chronically implanted planar silicon
[101] Shanmuganathan K, Capadona J R, Rowan S J and Weder C microelectrode arrays Prog. Brain Res. 194C 16780
2010 Stimuli-responsive mechanically adaptive polymer [118] Goligorsky M S 2011 TLR4 and HMGB1: partners in crime?
nanocomposites ACS Appl. Mater. Inter. 2 16574 Kidney Int. 80 4502

38
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

[119] Sims G P, Rowe D C, Rietdijk S T, Herbst R and Coyle A J electrode design and chronic recording function
2010 HMGB1 and RAGE in inammation and cancer Annu. Biomaterials 34 806174
Rev. Immunol. 28 36788 [137] Skousen M B J L and Tresco P A 2014 A Strategy to
[120] Block M L, Zecca L and Hong J S 2007 Microglia-mediated passively reduce neuroinammation surrounding devices
neurotoxicity: uncovering the molecular mechanisms Nat. implanted chronically in brain tissue by manipulating device
Rev. Neurosci. 8 5769 surface permeability Biomaterials in press
[121] Chao C C, Hu S and Peterson P K 1995 Glia cytokines, and [138] Edell D J, Toi V V, McNeil V M and Clark L D 1992 Factors
neurotoxicity Crit. Rev. Neurobiol. 9 189205 inuencing the biocompatibility of insertable silicon
[122] Hanisch U K and Kettenmann H 2007 Microglia: active microshafts in cerebral cortex IEEE Trans. Biomed. Eng. 39
sensor and versatile effector cells in the normal and 63543
pathologic brain Nat. Neurosci. 10 138794 [139] Schmidt S, Horch K and Normann R 1993 Biocompatibility
[123] Mantovani A, Sozzani S, Locati M, Allavena P and Sica A of silicon-based electrode arrays implanted in feline cortical
2002 Macrophage polarization: tumor-associated tissue J. Biomed. Mater. Res. 27 13939
macrophages as a paradigm for polarized M2 mononuclear [140] Schultz R L and Willey T J 1976 The ultrastructure of the
phagocytes Trends Immunol. 23 54955 sheath around chronically implanted electrodes in brain
[124] Quagliarello V J, Wispelwey B, Long W J Jr and Scheld W M J. Neurocytol 5 62142
1991 Recombinant human interleukin-1 induces meningitis [141] Stensaas S S and Stensaas L J 1976 The reaction of the
and blood-brain barrier injury in the rat. characterization and cerebral cortex to chronically implanted plastic needles Acta
comparison with tumor necrosis factor J. Clin. Invest. 87 Neuropathol (Berl) 35 187203
13606 [142] Stensaas S S and Stensaas L J 1978 Histopathological
[125] Qin L L Y, Wang T, Wei S J, Block M L, Wilson B, evaluation of materials implanted in the cerebral cortex Acta
Liu B and Hong J S 2004 NADPH oxidase mediates Neuropathol (Berl) 41 14555
lipopolysaccharide-induced neurotoxicity and [143] Collias J C and Manuelidis E E 1957 Histopathological
proinammatory gene expression in activated microglia changes produced by implanted electrodes in cat brains;
J. Biol. Chem. 279 141521 comparison with histopathological changes in human and
[126] Qin L L Y, Cooper C, Liu B, Wilson B and Hong J S 2002 experimental puncture wounds J. Neurosurg. 14 30228
Microglia enhance beta-amyloid peptide-induced toxicity in [144] Turner J N, Shain W, Szarowski D H, Andersen M,
cortical and mesencephalic neurons by producing reactive Martins S, Isaacson M and Craighead H 1999 Cerebral
oxygen species J. Neurochem. 83 97383 astrocyte response to micromachined silicon implants Exp.
[127] Gao H M J J, Wilson B, Zhang W, Hong J S and Liu B 2002 Neurol. 156 3349
Microglial activation-mediated delayed and progressive [145] Thelin J, Jorntell H, Psouni E, Garwicz M, Schouenborg J,
degeneration of rat nigral dopaminergic neurons: relevance Danielsen N and Linsmeier C E 2011 Implant size and
to Parkinsons disease J. Neurochem. 81 128597 xation mode strongly inuence tissue reactions in the CNS
[128] Clark I A, Alleva L M and Vissel B 2010 The roles of TNF PLoS One 6 e16267
in brain dysfunction and disease Pharmacol. Ther. 128 [146] Potter-Baker K A et al 2014 A comparison of
51948 neuroinammation to implanted microelectrodes in rat and
[129] Feuerstein G Z, Liu T and Cytokines Barone FC. 1994 mouse models Biomaterials 34 563746
inammation, and brain injury: role of tumor necrosis factor- [147] Abbott N J 2002 Astrocyte-endothelial interactions and
alpha Cerebrovasc. Brain Metab. Rev. 6 34160 blood-brain barrier permeability J. Anat. 200 62938
[130] Gosselin D and Rivest S 2007 Role of IL-1 and TNF in the [148] Andjelkovic A V, Kerkovich D and Pachter J S 2000
brain: twenty years of progress on a Dr. Jekyll/Mr. Hyde Monocyte:astrocyte interactions regulate MCP-1 expression
duality of the innate immune system Brain Behav Immunol. in both cell types J. Leukoc. Biol. 68 54552
21 2819 [149] Goldstein G W 1988 Endothelial cell-astrocyte interactions. A
[131] Stamatovic S M, Keep R F, Kunkel S L and Andjelkovic A V cellular model of the blood-brain barrier Ann. New York
2003 Potential role of MCP-1 in endothelial cell tight Acad. Sci. 529 319
junctionopening: signaling via Rho and Rho kinase J. Cell [150] Huang Y H, Sinha S R, Tanaka K, Rothstein J D and
Sci. 116 461528 Bergles D E 2004 Astrocyte glutamate transporters regulate
[132] Stamatovic S M, Shakui P, Keep R F, Moore B B, metabotropic glutamate receptor-mediated excitation of
Kunkel S L, Van Rooijen N and Andjelkovic A V 2005 hippocampal interneurons J. Neurosci.: Ofcial J. Soc.
Monocyte chemoattractant protein-1 regulation of blood- Neurosci. 24 45519
brain barrier permeability J. Cereb. Blood Flow Metab. 25 [151] Schmid-Brunclik N, Burgi-Taboada C, Antoniou X,
593606 Gassmann M and Ogunshola O O 2008 Astrocyte responses
[133] Sugama S, Takenouchi T, Cho B P, Joh T H, to injury: VEGF simultaneously modulates cell death and
Hashimoto M and Kitani H 2009 Possible roles of microglial proliferation Am. J. Physiol. Regul. Integr. Comparative
cells for neurotoxicity in clinical neurodegenerative diseases Physiol. 295 R86473
and experimental animal models Inamm. Allergy Drug [152] Kim Y T, Bridge M J and Tresco P A 2007 The inuence of
Targets 8 27784 the foreign body response evoked by broblast
[134] Yadav A, Saini V and Arora S 2010 MCP-1: chemoattractant transplantation on soluble factor diffusion in surrounding
with a role beyond immunity: a review Clin. Chim. Acta; Int. brain tissue J. Control Release 118 3407
J. Clin. Chem. 411 15709 [153] Aschner M, Sonnewald U and Tan K H 2002 Astrocyte
[135] Karumbaiah L, Norman S E, Rajan N B, Anand S, Saxena T, modulation of neurotoxic injury Brain Pathol. 12 47581
Betancur M, Patkar R and Bellamkonda R V 2012 The [154] Guenard V, Frisch G and Wood P M 1996 Effects of axonal
upregulation of specic interleukin (IL) receptor antagonists injury on astrocyte proliferation and morphology in vitro:
and paradoxical enhancement of neuronal apoptosis due to implications for astrogliosis Exp. Neurol. 137 17590
electrode induced strain and brain micromotion Biomaterials [155] Louw D F, Masada T and Sutherland G R 1998 Ischemic
33 598396 neuronal injury is ameliorated by astrocyte activation Can. J.
[136] Karumbaiah L, Saxena T, Carlson D, Patil K, Patkar R, Neurol. Sci. 25 1027
Gaupp E A, Betancur M, Stanley G B, Carin L and [156] Norenberg M D 1994 Astrocyte responses to CNS injury
Bellamkonda R V 2013 Relationship between intracortical J. Neuropathol. Exp. Neurol. 53 21320

39
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

[157] McConnell G C, Schneider T M, Owens D J and signaling pathways in neuritogenesis J. Biol. Chem. 281
Bellamkonda R V 2007 Extraction force and cortical tissue 249708
reaction of silicon microelectrode arrays implanted in the rat [173] Yamada H, Fredette B, Shitara K, Hagihara K, Miura R,
brain IEEE Trans. Biomed. Eng. 54 1097107 Ranscht B, Stallcup W B and Yamaguchi Y 1997 The brain
[158] Williams J C, Hippensteel J A, Dilgen J, Shain W and chondroitin sulfate proteoglycan brevican associates with
Kipke D R 2007 Complex impedance spectroscopy for astrocytes ensheathing cerebellar glomeruli and inhibits
monitoring tissue responses to inserted neural implants neurite outgrowth from granule neurons J. Neurosci.: Off. J.
J. Neural. Eng. 4 41023 Soc. Neurosci. 17 778495
[159] Seymour J P and Kipke D R 2007 Neural probe design for [174] Aihara N, Tanno H, Hall J J, Pitts L H and Noble L J 1994
reduced tissue encapsulation in CNS Biomaterials 28 Immunocytochemical localization of immunoglobulins in
3594607 the rat brain: relationship to the blood-brain barrier J. Comp.
[160] Ravikumar M, Hageman D J, Tomaszewski W H, Neurol. 342 48196
Chandra G M, Skousen J L and Capadona J R 2014 The [175] Azzi G, Bernaudin J F, Bouchaud C, Bellon B and
effect of residual endotoxin contamination on the Fleury-Feith J 1990 Permeability of the normal rat brain,
neuroinammatory response to sterilized intracortical spinal cord and dorsal root ganglia microcirculations to
microelectrodes J. Mater. Chem. B 2 251729 immunoglobulins G. Biol. Cell. 68 316
[161] McConnell G C, Rees H D, Levey A I, Gutekunst C-A, [176] Seitz R J, Heininger K, Schwendemann G, Toyka K V and
Gross R E and Bellamkonda R V 2009 Implanted neural Wechsler W 1985 The mouse blood-brain barrier and blood-
electrodes cause chronic, local inammation that is nerve barrier for IgG: a tracer study by use of the avidin-
correlated with local neurodegeneration J. Neural Eng. 6 biotin system Acta Neuropathol. 68 1521
056003 [177] Larochelle C, Alvarez J I and Prat A 2011 How do immune
[162] Saxena T, Karumbaiah L, Gaupp E A, Patkar R, Patil K, cells overcome the blood-brain barrier in multiple sclerosis?
Betancur M, Stanley G B and Bellamkonda R V 2013 The FEBS Lett. 585 377080
impact of chronic bloodbrain barrier breach on intracortical [178] Lassmann H 2011 A dynamic view of the blood-brain
electrode function Biomaterials 34 470313 barrier in active multiple sclerosis lesions Ann. Neurol. 70
[163] Jones L L Y Y, Stallcup W B and Tuszynski M H 2002 NG2 12
is a major chondroitin sulfate proteoglycan produced after [179] Rennaker R L, Miller J, Tang H and Wilson D A 2007
spinal cord injury and is expressed by macrophages and Minocycline increases quality and longevity of chronic
oligodendrocyte progenitors J. Neurosci. Methods 22 neural recordings J. Neural. Eng. 4 L15
2792803 [180] Lee S M, Yune T Y, Kim S J, Kim Y C, Oh Y J,
[164] Jones L L M R and Tuszynski M H 2003 The chondroitin Markelonis G J and Oh T H 2004 Minocycline inhibits
sulfate proteoglycans neurocan, brevican, phosphacan, and apoptotic cell death via attenuation of TNF-alpha expression
versican are differentially regulated following spinal cord following iNOS/NO induction by lipopolysaccharide in
injury Exp. Neurol. 182 399411 neuron/glia co-cultures J. Neurochem. 91 56878
[165] Fawcett J W and Asher R A 1999 The glial scar and central [181] Kobayashi K et al 2013 Minocycline selectively inhibits M1
nervous system repair Brain Res. Bull. 49 37791 polarization of microglia Cell Death Disease 4 e525
[166] Zhong Y and Bellamkonda R V 2007 Dexamethasone-coated [182] Shain W, Spataro L, Dilgen J, Haverstick K, Retterer S,
neural probes elicit attenuated inammatory response and Isaacson M, Saltzman M and Turner J N 2003 Controlling
neuronal loss compared to uncoated neural probes Brain. cellular reactive responses around neural prosthetic devices
Res. 1148 1527 using peripheral and local intervention strategies IEEE
[167] Friedlander D R, Milev P, Karthikeyan L, Margolis R K, Trans. Neural Syst. Rehab. Eng.: Publ. IEEE Eng. Med.
Margolis R U and Grumet M 1994 The neuronal chondroitin Biol. Soc. 11 1868
sulfate proteoglycan neurocan binds to the neural cell [183] Spataro L, Dilgen J, Retterer S, Spence A J, Isaacson M,
adhesion molecules Ng-CAM/L1/NILE and N-CAM and Turner J N and Shain W 2005 Dexamethasone treatment
inhibits neuronal adhesion and neurite outgrowth J. Cell reduces astroglia responses to inserted neuroprosthetic
Biol. 125 66980 devices in rat neocortex Exp. Neurol. 194 289300
[168] Gopalakrishnan S M, Teusch N, Imhof C, Bakker M H, [184] Zhong Y, McConnell G C, Ross J D, DeWeerth S P and
Schurdak M, Burns D J and Warrior U 2008 Role of Rho Bellamkonda R V (ed) 2005 A novel dexamethasone-
kinase pathway in chondroitin sulfate proteoglycan- releasing, anti-inammatory coating for neural implants.
mediated inhibition of neurite outgrowth in PC12 cells neural engineering Conf. Proc. 2nd Int. IEEE EMBS Neural
J. Neurosci. Res. 86 221426 Eng. (Arlington, VA, 1619 March 2005) pp 5225
[169] Hynds D L and Snow D M 1999 Neurite outgrowth inhibition [185] Wadhwa R, Lagenaur C F and Cui X T 2006
by chondroitin sulfate proteoglycan: stalling/stopping Electrochemically controlled release of dexamethasone from
exceeds turning in human neuroblastoma growth cones Exp. conducting polymer polypyrrole coated electrode J. Control
Neurol. 160 24455 Release 110 53141
[170] Iijima N, Oohira A, Mori T, Kitabatake K and Kohsaka S [186] Girbovan C, Morin L and Plamondon H 2012 Repeated
1991 Core protein of chondroitin sulfate proteoglycan resveratrol administration confers lasting protection against
promotes neurite outgrowth from cultured neocortical neuronal damage but induces dose-related alterations of
neurons J. Neurochem. 56 7068 behavioral impairments after global ischemia Behav.
[171] Kufer D P, Sosa I J and Reyes O 2009 Schwann cell Pharmacol. 23 113
chondroitin sulfate proteoglycan inhibits dorsal root [187] Patel K R, Scott E, Brown V A, Gescher A J,
ganglion neuron neurite outgrowth and substrate specicity Steward W P and Brown K 2011 Clinical trials of resveratrol
via a soma and not a growth cone mechanism J. Neurosci. Ann. New York Acad. Sci. 1215 1619
Res. 87 286371 [188] Khodagholy D et al 2013 In vivo recordings of brain activity
[172] Nakanishi K, Aono S, Hirano K, Kuroda Y, Ida M, Tokita Y, using organic transistors Nat. Commun. 4 1575
Matsui F and Oohira A 2006 Identication of neurite [189] Chung K et al 2013 Structural and molecular interrogation of
outgrowth-promoting domains of neuroglycan C, a brain- intact biological systems Nature 497 3327
specic chondroitin sulfate proteoglycan, and involvement [190] Harris J P 2011 The glia-neuronal response to cortical
of phosphatidylinositol 3-kinase and protein kinase C electrodes: interactions with substrate stiffness and

40
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

electrophysiology Dissertation Case Western Reserve intracortical microelectrodesa modeling and an in vivo
University study Conf. Proc. IEEE Eng. Med. Biol. Soc.
[191] Dobrovolskaia M A and Vogel S N 2002 Toll receptors, [208] Rousche P J, Pellinen D S, Pivin D P, Williams J C,
CD14, and macrophage activation and deactivation by LPS Vetter R J and Kipke D R 2001 Flexible polyimide-based
Microbes and Infection. 4 90314 intracortical electrode arrays with bioactive capability IEEE
[192] Doyle A, Zhang G, Abdel Fattah E A, Eissa N T and Li Y P Trans. Biomed. Eng. 48 36171
2011 Toll-like receptor 4 mediates lipopolysaccharide- [209] Takeuchi S, Ziegler D, Yoshida Y, Mabuchi K and Suzuki T
induced muscle catabolism via coordinate activation of 2005 Parylene exible neural probes integrated with
ubiquitin-proteasome and autophagy-lysosome pathways microuidic channels Lab on a Chip. 5 51923
FASEB J.: Off. Publ. Federation Am. Soc. Exp. Biol. 25 [210] Fernandez L J, Altuna A, Tijero M, Gabriel G, Villa R,
99110 Rodriguez M J, Batlle M, Vilares R, Berganzo J and
[193] Lund S, Christensen K, Hedtjarn M, Mortensen A, Blanco F J 2009 Study of functional viability of SU-8-based
Hagberg H, Falsig J, Hasseldam H, Schrattenholz A, microneedles for neural applications J. Micromech.
Porzgen P and Leist M 2006 The dynamics of the LPS Microeng. 19 025007
triggered inammatory response of murine microglia under [211] Mercanzini A, Colin P, Bensadoun J C, Bertsch A and
different culture and in vivo conditions J. Neuroimmunol. Renaud P 2009 In vivo electrical impedance spectroscopy of
180 7187 tissue reaction to microelectrode arrays IEEE Trans.
[194] Winslow B D and Tresco P A 2010 Quantitative analysis of Biomed. Eng. 56 190918
the tissue response to chronically implanted microwire [212] Lu Y, Wang D, Li T, Zhao X, Cao Y, Yang H and Duan Y Y
electrodes in rat cortex Biomaterials 31 155867 2009 Poly(vinyl alcohol)/poly(acrylic acid) hydrogel
[195] Gutowski S M, Templeman K L, South A B, Gaulding J C, coatings for improving electrode-neural tissue interface
Shoemaker J T, Laplaca M C, Bellamkonda R V, Biomaterials 30 414351
Lyon L A and Garcia A J 2013 Host response to microgel [213] Lacour S, Benmerah S, Tarte E, FitzGerald J, Serra J,
coatings on neural electrodes implanted in the brain McMahon S, Fawcett J, Graudejus O, Yu Z and
J. Biomed. Mater. Res. A 13 Morrison B III 2010 Flexible and stretchable micro-
[196] Sharp A A, Ortega A M, Restrepo D, Curran-Everett D and electrodes for in vitro and in vivo neural interfaces Med.
Gall K 2009 In vivo penetration mechanics and mechanical Biol. Eng. Comput. 48 94554
properties of mouse brain tissue at micrometer scales IEEE [214] Clement R S, Singh A, Olson B, Lee K and He J (ed) 2003
Trans. Biomed Eng. 56 4553 Neural recordings from a benzocyclobutene (BCB) based
[197] Jensen W, Yoshida K and Hofmann U G 2006 In-vivo intra-cortical neural implant in an acute animal model Proc.
implant mechanics of exible, silicon-based ACREO 25th Annual Int. Conf. of the IEEE EMBS (Cancun, Mexico)
microelectrode arrays in rat cerebral cortex IEEE Trans. [215] Altuna A, Bellistri E, Cid E, Aivar P, Gal B, Berganzo J,
Biomed. Eng. 53 93440 Gabriel G, Guimera A, Villa R, Fernandez L J and
[198] Jensen W, Hofmann U G and Yoshida K (ed) 2003 Menendez de la Prida L 2013 SU-8 based microprobes for
Assessment of subdural insertion force of single-tine simultaneous neural depth recording and drug delivery in the
microelectrodes in rat cerebral cortexengineering in brain Lab on a Chip. 13 142230
medicine and biology society 2003 Proc. of the 25th Annual [216] Vernekar V N, Cullen D K, Fogleman N, Choi Y, Garcia A J,
Int. Conf. of the IEEE Allen M G, Brewer G J and LaPlaca M C 2009 SU-8 2000
[199] Egert D, Peterson R L and Naja K (ed) 2011 Parylene rendered cytocompatible for neuronal bioMEMS
microprobes with engineered stiffness and shape for applications J. Biomed. Mater. Res. A 89A 13851
improved insertion. solid-state sensors 16th Int. Conf. on [217] Lee K, Singh A, He J, Massia S, Kim B and Raupp G 2004
Actuators and Microsystems 59 June (Beijing) pp 198201 Polyimide based neural implants with stiffness improvement
[200] ChunXiang T and Jiping H (ed) 2005 Monitoring insertion Sensors Actuators B 102 6772
force and electrode impedance during implantation of [218] Lee K-K, He J, Singh A, Massia S, Ehteshami G, Kim B and
microwire electrodes 27th Annual Int. Conf. of the IEEE- Raupp G 2004 Polyimide-based intracortical neural implant
EMBS 1718 Jan. 2006 with improved structural stiffness J. Micromech. Microeng.
[201] Wester B A, Lee R H and LaPlaca M C 2009 Development 14 327
and characterization of in vivo exible electrodes compatible [219] Lee K, He J and Wang L 2004 Benzocyclobutene (BCB)
with large tissue displacements J. Neural. Eng. 6 024002 based neural implants with microuidic channel P. Ann Int.
[202] Lee H, Bellamkonda R V, Sun W and Levenston M E 2005 IEEE Embs. 26 43269
Biomechanical analysis of silicon microelectrode-induced [220] Lee K, He J, Clement R, Massia S and Kim B 2004
strain in the brain J. Neural Eng. 2 819 Biocompatible benzocyclobutene (BCB)-based neural implants
[203] Subbaroyan J 2007 Investigations of Tethering Induced Injury with micro-uidic channel Biosens. Bioelectron 20 4047
Response in Brain Tissue by Intracortical Implants Through [221] Lewitus D, Smith K L, Shain W and Kohn J 2011 Ultrafast
Modeling and In vivo Experiments (Ann Arbor, MI: resorbing polymers for use as carriers for cortical neural
University of Michigan) probes Acta Biomater. 7 248391
[204] Zhu R Y H, Varadan V K, Smith C S and Huang G L 2012 [222] Lewitus D Y, Smith K L, Shain W, Bolikal D and Kohn J
Biomechanical strain analysis at the interface of brain and 2011 The fate of ultrafast degrading polymeric implants in
nanowire electrodes on a neural probe J. Nanotechnology the brain Biomaterials 32 554350
Eng. Med 2 [223] Lewitus D, Vogelstein R J, Zhen G H, Choi Y S, Kohn J,
[205] Kim Y T, Hitchcock R W, Bridge M J and Tresco P A 2004 Harshbarger S and Jia X F 2011 Designing tyrosine-derived
Chronic response of adult rat brain tissue to implants polycarbonate polymers for biodegradable regenerative type
anchored to the skull. Biomaterials 25 222937 neural interface capable of neural recording IEEE Trans.
[206] Markwardt N T, Stokol J and Rennaker R L 2013 2nd. Sub- Neural Syst. Rehab. 19 20412
meninges implantation reduces immune response to neural [224] Kato Y, Saito I, Hoshino T, Suzuki T and Mabuchi K 2006
implants J. Neurosci. Methods. 214 11925 Preliminary study of multichannel exible neural probes
[207] Subbaroyan J and Kipke D R (ed) 2006 The role of exible coated with hybrid biodegradable polymer Proc. IEEE 1-15
polymer interconnects in chronic tissue response induced by 54447

41
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

[225] Gilgunn P J, Khilwani R, Kozai T D Y, Weber D J, Cui X T, under acute in vitro and in vivo settings Front. Neuroeng. 7
Erdos G, Ozdoganlar O B and Fedder G K (ed) 2012 An 18
ultra-compliant, scalable neural probe with molded [245] Fan Y W, Cui F Z, Hou S P, Xu Q Y, Chen L N and Lee I S
biodissolvable delivery vehicle IEEE 25th Int. Conf. on 2002 Culture of neural cells on silicon wafers with nano-
Micro Electro Mechanical Systems (MEMS) (29 January scale surface topograph J. Neurosci. Methods 120 1723
20122 February) [246] Green R A, Baek S, Poole-Warren L A and Martens P J 2010
[226] Cui X, Lee V A, Raphael Y, Wiler J A, Hetke J F, Conducting polymer-hydrogels for medical electrode
Anderson D J and Martin D C 2001 Surface modication of applications Sci. Technol. Adv. Mater. 11
neural recording electrodes with conducting polymer/ [247] Dalby M J 2007 Cellular response to low adhesion
biomolecule blends J. Biomed. Mater. Res. 56 26172 nanotopographies Int. J. Nanomed. 2 37381
[227] Rao L, Zhou H, Li T, Li C and Duan Y Y 2012 Polyethylene [248] Moxon K A, Kalkhoran N M, Markert M, Sambito M A,
glycol-containing polyurethane hydrogel coatings for McKenzie J L and Webster J T 2004 Nanostructured surface
improving the biocompatibility of neural electrodes Acta modication of ceramic-based microelectrodes to enhance
Biomater. 8 223342 biocompatibility for a direct brain-machine interface IEEE
[228] Kim D-H et al 2010 Dissolvable lms of silk broin for Trans. Biomed. Eng. 51 8819
ultrathin conformal bio-integrated electronics Nat. Mater [249] Moxon K A, Leiser S C, Gerhardt G A, Barbee K A and
[10.1038/nmat2745]. Advance online publication Chapin J K 2004 Ceramic-based multisite electrode arrays
[229] Bawa P, Pillay V, Choonara Y E and du Toit L C 2009 for chronic single-neuron recording IEEE Trans. Biomed.
Stimuli-responsive polymers and their applications in drug Eng. 51 64756
delivery Biomed. Mater. 4 022001 [250] Moxon K A, Hallman S, Aslani A, Kalkhoran N M and
[230] Alarcon CdlH, Pennadam S and Alexander C 2005 Stimuli Lelkes P I 2007 Bioactive properties of nanostructured
responsive polymers for biomedical applications Chem. Soc. porous silicon for enhancing electrode to neuron interfaces
Rev. 34 27685 J. Biomater. Sci. Polym. Ed. 18 126381
[231] Stuart M A C et al 2010 Emerging applications of stimuli- [251] Ereifej E S, Matthew H W, Newaz G, Mukhopadhyay A,
responsive polymer materials Nat. Mater. 9 10113 Auner G, Salakhutdinov I and VandeVord P J 2013
[232] Urban M W (ed) 2011 Handbook of Stimuli-Responsive Nanopatterning effects on astrocyte reactivity J. Biomed.
Materials 1st edn (New York: Wiley) Mater. Res. Part A 101A 174357
[233] Mano J F 2008 Stimuli-responsive polymeric systems for [252] Capadona J R, Petrie T A, Fears K P and Latour R A 2005
biomedical applications Adv. Eng. Mater 10 51527 Collard DM, Garca AJ. Surface-nucleated assembly of
[234] Roy D, Cambre J N and Sumerlin B S 2010 Future brillar extracellular matrices Adv Mater. 17 26048
perspectives and recent advances in stimuli-responsive [253] Bridges A W and Garca A J 2008 Anti-inammatory
materials Prog. Polym. Sci. 35 278301 polymeric coatings for implantable biomaterials and devices.
[235] Nguyen J K, Park D J, Skousen J L, Hess-Dunning A, J. Diabetes Sci. Technol. 2 98494
Tyler D J, Rowan S J, Weder C and Capadona J R 2014 [254] Leach J, Achyuta A K H and Murthy S K 2010 Bridging the
Mechanically-compliant intracortical implants reduce the divide between neuroprosthetic design, tissue engineering
neuroinammatory response J. Neural. Eng. 11 056014 and neurobiology Front. Neuroeng. 2 18
[236] Ware T et al 2012 Three-dimensional exible electronics [255] He W, McConnell G C, Schneider T M and
enabled by shape memory polymer substrates for responsive Bellamkonda R V A 2007 Novel anti-inammatory surface
neural interfaces Macromol. Mater. Eng. 297 1193202 for neural electrodes Adv. Mater. 19 352933
[237] Sharp A A, Panchawagh H V, Ortega A, Artale R, [256] Azemi E, Lagenaur C F and Cui X T 2011 The surface
Richardson-Burns S, Finch D S, Gall K, Mahajan R L and immobilization of the neural adhesion molecule L1 on
Restrepo D 2006 Toward a self-deploying shape memory neural probes and its effect on neuronal density and gliosis at
polymer neuronal electrode J. Neural. Eng. 3 L2330 the probe/tissue interface Biomaterials 32 68192
[238] Tien L W, Wu F, Tang-Schomer M D, Yoon E, [257] Azemi E, Stauffer W R, Gostock M S, Lagenaur C F and
Omenetto F G and Kaplan D L 2013 Silk as a Cui X T 2008 Surface immobilization of neural adhesion
multifunctional biomaterial substrate for reduced glial molecule L1 for improving the biocompatibility of chronic
scarring around brain-penetrating electrodes Adv. Funct. neural probes: in vitro characterization Acta Biomater.
Mater. 23 318593 (doi:10.1016/j.actbio.2008.02.028)
[239] Szarowski D H, Andersen M D, Retterer S, Spence A J, [258] Webb K, Budko E, Neuberger T J, Chen S, Schachner M and
Isaacson M, Craighead H G, Turner J N and Shain W 2003 Tresco P A 2001 Substrate-bound human recombinant L1
Brain responses to micro-machined silicon devices Brain. selectively promotes neuronal attachment and outgrowth in
Res. 983 2335 the presence of astrocytes and broblasts Biomaterials 22
[240] Stice P, Gilletti A, Panitch A and Muthuswamy J 2007 Thin 101728
microelectrodes reduce GFAP expression in the implant site [259] Taub A H, Hogri R, Magal A, Mintz M and
in rodent somatosensory cortex J. Neural. Eng. 4 4253 Shacham-Diamand Y 2012 Bioactive anti-inammatory
[241] Saddow S E 2012 Silicon Carbide Biotechnology 1st edn coating for chronic neural electrodes J. Biomed. Mater. Res.
(Waltham, MA: Elsevier.) A 100A 18548
[242] Raynor J E, Capadona J R, Collard D M, Petrie T A and [260] He W and Bellamkonda R V 2005 Nanoscale neuro-
Garcia A J 2009 Polymer brushes and self-assembled integrative coatings for neural implants Biomaterials 26
monolayers: versatile platforms to control cell adhesion to 298390
biomaterials (Review) Biointerphases 4 FA3-16 [261] Kim D H, Richardson-Burns S M, Hendricks J L,
[243] Leung B K, Biran R, Underwood C J and Tresco P A 2008 Sequera C and Martin D C 2007 Effect of immobilized nerve
Characterization of microglial attachment and cytokine growth factor on conductive polymers: electrical properties
release on biomaterials of differing surface chemistry and cellular response Adv. Funct. Mater. 17 7986
Biomaterials 29 328997 [262] Kim D H and Martin D C 2006 Sustained release of
[244] Sommakia S, Gaire J, Rickus J L and Otto K J 2014 Resistive dexamethasone from hydrophilic matrices using PLGA
and reactive changes to the impedance of intracortical nanoparticles for neural drug delivery Biomaterials 27
microelectrodes can be mitigated with polyethylene glycol 30317

42
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

[263] Zhong Y and Bellamkonda R V 2005 Controlled release of [281] Tanaka J and Maeda N 1996 Microglial ramication requires
anti-inammatory agent -MSH from neural implants nondiffusible factors derived from astrocytes Exp. Neurol.
J. Control. Release 106 30918 137 36775
[264] Abidian M R and Martin D C 2009 Multifunctional [282] Wolchok J C and Tresco P A 2010 The isolation of cell
nanobiomaterials for neural interfaces Adv. Funct. Mater. 19 derived extracellular matrix constructs using sacricial open-
57385 cell foams Biomaterials 31 9595603
[265] Mercanzini A, Reddy S T, Velluto D, Colin P, Maillard A, [283] McClelland R, Wauthier E, Uronis J and Reid L 2008
Bensadoun J C, Hubbell J A and Renaud P 2010 Controlled Gradients in the liver's extracellular matrix chemistry from
release nanoparticle-embedded coatings reduce the tissue periportal to pericentral zones: inuence on human hepatic
reaction to neuroprostheses J. Control. Release 145 196202 progenitors Tissue Eng. A 14 5970
[266] Massia S P, Holecko M M and Ehteshami G R 2004 In vitro [284] Huet C, Pisselet C, Mandon-Pepin B, Monget P and
assessment of bioactive coatings for neural implant Monniaux D 2001 Extracellular matrix regulates ovine
applications J. Biomed. Mater. Res. A 68A 17786 granulosa cell survival, proliferation and steroidogenesis:
[267] Stauffer W R and Cui X T 2006 Polypyrrole doped with 2 relationships between cell shape and function J. Endocrinol.
peptide sequences from laminin Biomaterials 27 240513 169 34760
[268] He W, McConnell G C and Bellamkonda R V 2006 [285] Sellaro T L, Ravindra A K, Stolz D B and Badylak S F 2007
Nanoscale laminin coating modulates cortical scarring Maintenance of hepatic sinusoidal endothelial cell
response around implanted silicon microelectrode arrays phenotype in vitro using organ-specic extracellular matrix
J. Neural Eng. 3 31626 scaffolds Tissue Eng. 13 230110
[269] Kolarcik C L, Bourbeau D, Azemi E, Rost E, Zhang L, [286] Singelyn J M, DeQuach J A, Seif-Naraghi S B,
Lagenaur C F, Weber D J and Cui X T 2012 In vivo effects Littleeld R B, Schup-Magofn P J and Christman K L
of L1 coating on inammation and neuronal health at the 2009 Naturally derived myocardial matrix as an injectable
electrodetissue interface in rat spinal cord and dorsal root scaffold for cardiac tissue engineering Biomaterials 30
ganglion Acta Biomater 8 356175 540916
[270] Green R A, Suaning G J, Poole-Warren L A and Lovell N H [287] Stern M M, Myers R L, Hammam N, Stern K A, Eberli D,
(ed) 2009 Bioactive conducting polymers for neural Kritchevsky S B, Soker S and Van Dyke M 2009 The
interfaces application to vision prosthesis 4th Int. IEEE/ inuence of extracellular matrix derived from skeletal
EMBS Conf. on Neural Engineering (29 April 20092 muscle tissue on the proliferation and differentiation of
May 2009) myogenic progenitor cells ex vivo Biomaterials 30 23939
[271] Kang G, Borgens R B and Cho Y 2011 Well-ordered porous [288] Zhang Y, He Y, Bharadwaj S, Hammam N, Carnagey K,
conductive polypyrrole as a new platform for neural Myers R, Atala A and Van Dyke M 2009 Tissue-specic
interfaces Langmuir 27 617984 extracellular matrix coatings for the promotion of cell
[272] Potter-Baker K A, Nguyen J K, Kovach K M, Gitomer M M, proliferation and maintenance of cell phenotype
Srail T W, Stewart W G, Skousen J L and Capadona J R Biomaterials 30 40218
2014 Development of superoxide dismutase mimetic [289] Badylak S, Obermiller J, Geddes L and Matheny R 2003
surfaces to reduce accumulation of reactive oxygen species Extracellular matrix for myocardial repair Heart Surgery
surrounding intracortical microelectrodes J. Mater. Chem. B Forum 6 E206
2 224858 [290] Brown B N, Valentin J E, Stewart-Akers A M,
[273] Chen S and Allen M G 2012 Extracellular matrix-based McCabe G P and Badylak S F 2009 Macrophage phenotype
materials for neural interfacing MRS Bull. 37 60613 and remodeling outcomes in response to biologic scaffolds
[274] Heiduschka P, Romann I, Ecken H, Schning M, with and without a cellular component Biomaterials 30
Schuhmann W and Thanos S 2001 Dened adhesion and 148291
growth of neurones on articial structured substrates [291] Allman A J, McPherson T B, Badylak S F, Merrill L C,
Electrochim. Acta. 9/1/ 47 299307 Kallakury B, Sheehan C, Raeder R H and Metzger D W
[275] Massia S P, Rao S S and Hubbell J A 1993 Covalently 2001 Xenogeneic extracellular matrix grafts elicit a TH2-
immobilized laminin peptide Tyr-Ile-Gly-Ser-Arg (YIGSR) restricted immune response Transplantation 71 163140
supports cell spreading and co-localization of the 67- [292] Allaire E, Bruneval P, Mandet C, Becquemin J P and
kilodalton laminin receptor with a-actinin and vinculin Michel J B 1997 The immunogenicity of the extracellular
J. Biol. Chem. 268 80539 matrix in arterial xenografts Surgery 122 7381
[276] Green R A, Lovell N H and Poole-Warren L A 2009 Cell [293] Allaire E, Mandet C, Bruneval P, Bensenane S,
attachment functionality of bioactive conducting polymers Becquemin J P and Michel J B 1996 Cell and extracellular
for neural interfaces Biomaterials 30 363744 matrix rejection in arterial concordant and discordant
[277] Green R A, Lovell N H and Poole-Warren L A 2010 Impact xenografts in the rat. Transplantation 62 794803
of co-incorporating laminin peptide dopants and [294] Allaire E, Guettier C, Bruneval P, Plissonnier D and
neurotrophic growth factors on conducting polymer Michel J B 1994 Cell-free arterial grafts: morphologic
properties Acta Biomater. 6 6371 characteristics of aortic isografts, allografts, and xenografts
[278] Green R A, Matteucci P B, Hassarati R T, Giraud B, in rats J. Vasc. Surg. 19 44656
Dodds C W D, Chen S, Byrnes-Preston P J, Suaning G J, [295] De Vos P, De Haan B J, Wolters G H, Strubbe J H and
Poole-Warren L A and Lovell N H 2013 Performance of Van Schilfgaarde R 1997 Improved biocompatibility but
conducting polymer electrodes for stimulating limited graft survival after purication of alginate for
neuroprosthetics J. Neural Eng. 10 016009 microencapsulation of pancreatic islets Diabetologia 40
[279] Cutler S 2003 Engineering cell adhesive surfaces that direct 26270
integrin 51 binding using a recombinant fragment of [296] Winn S R, Aebischer P and Galletti P M 1989 Brain tissue
bronectin Biomaterials 24 175970 reaction to permselective polymer capsules J. Biomed.
[280] Tanaka J, Toku K, Sakanaka M and Maeda N 1999 Mater. Res. 23 3144
Morphological differentiation of microglial cells in culture: [297] Jaeger C B, Winn S R, Tresco P A and Aebischer P 1991
involvement of insoluble factors derived from astrocytes Repair of the blood-brain barrier following implantation of
Neurosci. Res. 34 20715 polymer capsules Brain. Res. 14 June 551 16370

43
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

[298] Winn S R, Tresco P A, Zielinski B, Greene L A, [316] Xiao Y, Cui X, Hancock J M, Bouguettaya M,
Jaeger C B and Aebischer P 1991 Behavioral recovery Reynolds J R and Martin D C 2004 Electrochemical
following intrastriatal implantation of microencapsulated polymerization of poly(hydroxymethylated-3,4-
PC12 cells Exp. Neurol. 113 3229 ethylenedioxythiophene) (PEDOT-MeOH) on multichannel
[299] Bridge M J and Tresco P A 2008 In vivo solute diffusivity in neural probes Sensors Actuators B 99 43743
brain tissue surrounding indwelling neural implants ed [317] Yang J, Kim D H, Hendricks J L, Leach M, Northey R and
W M Reichert Indwelling Neural Implants: Strategies for Martin D C 2005 Ordered surfactant-templated poly(3,4-
Contending with the in vivo environment. (Boca Raton, ethylenedioxythiophene) (PEDOT) conducting polymer on
FL: CRC) microfabricated neural probes Acta Biomater. 1 12536
[300] La Flamme K E, Popat K C, Leoni L, Markiewicz E, [318] Richardson-Burns S M, Hendricks J L, Foster B, Povlich L K,
La Tempa T J, Roman B B, Grimes C A and Desai T A 2007 Kim D-H and Martin D C 2007 Polymerization of the
Biocompatibility of nanoporous alumina membranes for conducting polymer poly(3,4-ethylenedioxythiophene)
immunoisolation Biomaterials 28 263845 (PEDOT) around living neural cells Biomaterials 28
[301] Lopez C A, Fleischman A J, Roy S and Desai T A 2006 153952
Evaluation of silicon nanoporous membranes and ECM- [319] Richardson-Burns S M, Hendricks J L and Martin D C 2007
based microenvironments on neurosecretory cells Electrochemical polymerization of conducting polymers in
Biomaterials 27 307583 living neural tissue J. Neural Eng. 4 L613
[302] Galimberti D, Baron P, Meda L, Prat E, Scarpini E, [320] Ludwig K A, Langhals N B, Joseph M D,
Delgado R, Catania A, Lipton J M and Scarlato G 1999 - Richardson-Burns S M, Hendricks J L and Kipke D R 2011
MSH peptides inhibit production of nitric oxide and tumor Poly(3,4-ethylenedioxythiophene) (PEDOT) polymer
necrosis factor- by microglial cells activated with - coatings facilitate smaller neural recording electrodes
amyloid and interferon Biochem. Biophys. Res. Commun. J. Neural. Eng. 8 014001
263 2516 [321] Kozai T D Y, Langhals N B, Patel P R, Deng X P,
[303] Guimard N K, Gomez N and Schmidt C E 2007 Conducting Zhang H N, Smith K L, Lahann J, Kotov N A and
polymers in biomedical engineering Prog. Polym. Sci. 32 Kipke D R 2012 Ultrasmall implantable composite
876921 microelectrodes with bioactive surfaces for chronic neural
[304] Ravichandran R, Sundarrajan S, Venugopal J R, interfaces Nat. Materials 11 106573
Mukherjee S and Ramakrishna S 2010 Applications of [322] Yamato H, Ohwa M and Wernet W 1995 Stability of
conducting polymers and their issues in biomedical polypyrrole and poly(3,4-ethylenedioxythiophene) for
engineering J. R. Soc. Interface 7 S55979 biosensor application J. Electroanal. Chem. 397
[305] Cui X, Hetke J F, Wiler J A, Anderson D J and Martin D C 16370
2001 Electrochemical deposition and characterization of [323] Mandal H S, Knaack G L, Charkhkar H, McHail D G,
conducting polymer polypyrrole/PSS on multichannel neural Kastee J S, Dumas T C, Peixoto N, Rubinson J F and
probes Sensors and Actuators A 93 818 Pancrazio J J 2014 Improving the performance of poly(3,4-
[306] Buchko C J, Kozloff K M and Martin D C 2001 Surface ethylenedioxythiophene) for brainmachine interface
characterization of porous, biocompatible protein polymer applications Acta Biomater. 10 244654
thin lms Biomaterials 22 1289300 [324] Aguilar Z 2012 Nanomaterials for Medical Applications 1st
[307] Yang J and Martin D C 2004 Microporous conducting edn (Amsterdam: Elsevier)
polymers on neural microelectrode arrays: I electrochemical [325] Mozafari M R (ed) 2007 Nanomaterials and Nanosystems for
deposition. Sensors and Actuators B 101 13342 Biomedical Applications 1st edn (Berlin: Springer)
[308] Harris A R, Morgan S J, Chen J, Kapsa R M I, [326] Michael Giersig and Khomutov G B (ed) 2008 Nanomat.
Wallace G G and Paolini A G 2013 Conducting polymer Appl. Med. Biol. 1st edn (Berlin: Springer)
coated neural recording electrodes J. Neural Eng. 10 016004 [327] Kotov N A et al 2009 Nanomaterials for neural interfaces
[309] Green R A, Lovell N H, Wallace G G and Poole-Warren L A Adv. Mater 21 39704004
2008 Conducting polymers for neural interfaces: challenges [328] Pancrazio J J 2008 Neural interfaces at the nanoscale
in developing an effective long-term implant Biomaterials Nanomedicine 3 82330
29 33939 [329] Merrill D R, Bikson M and Jefferys J G R 2005 Electrical
[310] Indwelling Neural Implants 2008 Strategies for Contending stimulation of excitable tissue: design of efcacious and safe
with the In vivo Environment (Boca Raton, FL: CRC) protocols J. Neurosci. Methods 141 17198
[311] Kim J, Choi S, Lillehei P, King G, Watt G, Chu S, Park Y and [330] Silva G A 2006 Neuroscience nanotechnology: progress,
Thibeault S (ed) 2004 Electrochemical reconstitution of opportunities and challenges Nat. Rev. Neurosci. 7 6574
biomolecules for applications as electrocatalysts for the [331] Mohammed J S, DeCoster M A and McShane M J 2004
bionanofuel cell Smart Structures and Materials 2004:: Micropatterning of nanoengineered surfaces to study
Smart Electronics, MEMS, BioMEMS, and Nanotechnology neuronal cell attachment in vitro Biomacromolecules 5
(Bellingham, WA: Optical Engineering) 174555
[312] George P M, Lyckman A W, LaVan D A, Hegde A, Leung Y, [332] Ai H, Meng H, Ichinose I, Jones S A, Mills D K,
Avasare R, Testa C, Alexander P M, Langer R and Sur M Lvov Y M and Qiao X 2003 Biocompatibility of layer-by-
2005 Fabrication and biocompatibility of polypyrrole layer self-assembled nanolm on silicone rubber for neurons
implants suitable for neural prosthetics Biomaterials 26 J. Neurosci. Methods 9/30/ 128 18
35119 [333] Alivisatos A P et al 2013 Nanotools for neuroscience and
[313] Schmidt C E, Shastri V R, Vacanti J P and Langer R 1997 brain activity mapping ACS Nano 7 185066
Stimulation of neurite outgrowth using an electrically [334] Voge C M and Stegemann J P 2011 Carbon nanotubes in
conducting polymer Proc. Natl. Acad. Sci. 94 894853 neural interfacing applications J. Neural. Eng. 8 011001
[314] Schlenoff J B and Xu H 1992 Evolution of physical and [335] Hanein Y and Bareket-Keren L 2013 Carbon nanotube based
electrochemical properties of polypyrrole during extended multi electrode arrays for neuronal interfacing: progress and
oxidation J. Electrochem. Soc. 139 2397401 prospects Front. Neural Circuits 6
[315] Cui X and Martin D C 2003 Electrochemical deposition and [336] Mattson M, Haddon R and Rao A 2000 Molecular
characterization of poly(3,4-ethylenedioxythiophene) on functionalization of carbon nanotubes and use as substrates
neural microelectrode arrays Sensors Actuators B 89 92102 for neuronal growth J. Mol. Neurosci. 14 17582

44
J. Neural Eng. 12 (2015) 011001 M Jorfi et al

[337] Gheith M K, Pappas T C, Liopo A V, Sinani V A, Shim B S, Ozone-modied carbon nanotube electrodes for neuronal
Motamedi M, Wicksted J P and Kotov N A 2006 recording Adv. Mater. 22 217781
Stimulation of neural cells by lateral currents in conductive [351] Chen Y-C et al 2011 An active, exible carbon nanotube
layer-by-layer lms of single-walled carbon nanotubes Adv. microelectrode array for recording electrocorticograms
Mater. 18 29759 J. Neural. Eng. 8 034001
[338] Gheith M K, Sinani V A, Wicksted J P, Matts R L and [352] Gabay T, Ben-David M, Kalifa I, Sorkin R, Abrams Z,
Kotov N A 2005 Single-walled carbon nanotube Ben-Jacob E and Hanein Y 2007 Electro-chemical and
polyelectrolyte multilayers and freestanding lms as a biological properties of carbon nanotube based multi-
biocompatible platform for neuroprosthetic implants Adv. electrode arrays Nanotechnology 18 035201
Mater. 17 266370 [353] Hanein Y 2010 Carbon nanotube integration into MEMS
[339] Malarkey E B, Fisher K A, Bekyarova E, Liu W, devices Phys. Status Solidi b 247 263540
Haddon R C and Parpura V 2008 Conductive single-walled [354] Nguyen-Vu T D B, Chen H, Cassell A M, Andrews R,
carbon nanotube substrates modulate neuronal growth Nano Meyyappan M and Li J 2006 Vertically aligned carbon
Lett. 9 2648 nanober arrays: an advance toward electricalneural
[340] Lovat V, Pantarotto D, Lagostena L, Cacciari B, interfaces Small 2 8994
Grandolfo M, Righi M, Spalluto G, Prato M and Ballerini L [355] Li J, Nguyen-Vu T D B, Chen H, Cassell A M,
2005 Carbon nanotube substrates boost neuronal electrical Andrews R J and Meyyappan M 2007 Vertically aligned
signaling Nano Lett. 5 110710 carbon nanober architecture as a multifunctional 3-D neural
[341] Hu H, Ni Y, Montana V, Haddon R C and Parpura V 2004 electrical interface IEEE Trans. Biomed. Eng. 54 11218
Chemically functionalized carbon nanotubes as substrates [356] Agarwal S, Zhou X, Ye F, He Q, Chen G C K, Soo J, Boey F,
for neuronal growth Nano Lett. 4 50711 Zhang H and Chen P 2010 Interfacing live cells with
[342] Wang K, Fishman H A, Dai H and Harris J S 2006 Neural nanocarbon substrates Langmuir 26 22447
stimulation with a carbon nanotube microelectrode array [357] Li N, Zhang X, Song Q, Su R, Zhang Q, Kong T, Liu L,
Nano Lett. 6 20438 Jin G, Tang M and Cheng G 2011 The promotion of neurite
[343] Mazzatenta A, Giugliano M, Campidelli S, Gambazzi L, sprouting and outgrowth of mouse hippocampal cells in
Businaro L, Markram H, Prato M and Ballerini L 2007 culture by graphene substrates Biomaterials 32 937482
Interfacing neurons with carbon nanotubes: Electrical signal [358] Lv M, Zhang Y, Liang L, Wei M, Hu W, Li X and Huang Q
transfer and synaptic stimulation in cultured brain circuits 2012 Effect of graphene oxide on undifferentiated and
J. Neurosci. 27 69316 retinoic acid-differentiated SH-SY5Y cells line Nanoscale 4
[344] Cellot G et al 2009 Carbon nanotubes might improve 38616
neuronal performance by favouring electrical shortcuts. Nat. [359] Chen C H, Lin C T, Chen J J, Hsu W L, Chang Y C, Yeh S R,
Nanotechnology 4 12633 Li L J and Yao D J (ed) 2011 A graphene-based
[345] Keefer E W, Botterman B R, Romero M I, Rossi A F and microelectrode for recording neural signals 16th Int. Conf.
Gross G W 2008 Carbon nanotube coating improves on Solid-State Sens., Actuators and Microsystems (59
neuronal recordings Nat. Nanotechnology 3 4349 June 2011)
[346] Luo X, Weaver C L, Zhou D D, Greenberg R and Cui X T [360] Luo X, Weaver C L, Tan S and Cui X T 2013 Pure graphene
2011 Highly stable carbon nanotube doped poly(3,4- oxide doped conducting polymer nanocomposite for bio-
ethylenedioxythiophene) for chronic neural stimulation interfacing J. Mater. Chem. B 1 13408
Biomaterials 32 55517 [361] Abidian M R and Martin D C 2008 Experimental and
[347] Baranauskas G, Maggiolini E, Castagnola E, Ansaldo A, theoretical characterization of implantable neural
Mazzoni A, Angotzi G N, Vato A, Ricci D, Panzeri S and microelectrodes modied with conducting polymer
Fadiga L 2011 Carbon nanotube composite coating of neural nanotubes Biomaterials 29 127383
microelectrodes preferentially improves the multiunit signal- [362] Abidian M R, Ludwig K A, Marzullo T C, Martin D C and
to-noise ratio J. Neural. Eng. 8 066013 Kipke D R 2009 Interfacing conducting polymer nanotubes
[348] Lu Y, Li T, Zhao X Q, Li M, Cao Y L, Yang H X and with the central nervous system: chronic neural recording
Duan Y W Y 2010 Electrodeposited polypyrrole/carbon using poly(3,4-ethylenedioxythiophene) nanotubes Adv.
nanotubes composite lms electrodes for neural interfaces Mater. 21 376470
Biomaterials 31 516981 [363] Abidian M R, Corey J M, Kipke D R and Martin D C 2010
[349] Lin C-M, Lee Y-T, Yeh S-R and Fang W 2009 Flexible Conducting-polymer nanotubes improve electrical
carbon nanotubes electrode for neural recording. Biosens. properties, mechanical adhesion, neural attachment, and
Bioelectron. 24 27917 neurite outgrowth of neural electrodes Small 6 4219
[350] Hsu H-L, Teng I J, Chen Y-C, Hsu W-L, Lee Y-T, Yen S-J,
Su H-C, Yeh S-R, Chen H and Yew T-R 2010 Flexible UV-

45

Das könnte Ihnen auch gefallen