Anda di halaman 1dari 16

646707

research-article2016
NROXXX10.1177/1073858416646707The NeuroscientistLewis et al.

Review
The Neuroscientist

Brain Neuromodulation
116
The Author(s) 2016
Reprints and permissions:
Techniques: A Review sagepub.com/journalsPermissions.nav
DOI: 10.1177/1073858416646707
nro.sagepub.com

Philip M. Lewis1,2,3, Richard H. Thomson3,4,


Jeffrey V. Rosenfeld1,2,3,5, and Paul B. Fitzgerald3,4

Abstract
The modulation of brain function via the application of weak direct current was first observed directly in the early
19th century. In the past 3 decades, transcranial magnetic stimulation and deep brain stimulation have undergone
clinical translation, offering alternatives to pharmacological treatment of neurological and neuropsychiatric disorders.
Further development of novel neuromodulation techniques employing ultrasound, micro-scale magnetic fields and
optogenetics is being propelled by a rapidly improving understanding of the clinical and experimental applications
of artificially stimulating or depressing brain activity in human health and disease. With the current rapid growth in
neuromodulation technologies and applications, it is timely to review the genesis of the field and the current state of
the art in this area.

Keywords
neuromodulation, neuropsychiatry, cognitive, implant, bionics, brain

Introduction and blindness (Lewis and others 2015). Stimulation of


the somatosensory cortex is also being explored as a
A variety of techniques have been developed, or are means of providing sensory feedback to recipients of
under investigation, for influencing or modulating the motor neuroprostheses, offering the potential to pro-
activity of the human brain. This development is being vide a sense of touch to robotic limbs (Berg and oth-
propelled by rapid growth in our understanding of the ers 2013).
clinical and experimental applications of artificially Techniques for brain stimulation or neuromodulation
stimulating or depressing brain activity in human vary depending on the clinical context, the precise loca-
health and disease. Brain stimulation is being applied tion and size of targeted areas, and the desired effect.
clinically for the treatment of a variety of neurological With the current rapid growth in neuromodulation tech-
conditions (for review, see Udupa and Chen 2015) nologies and applications, it is timely to review the gen-
including Parkinsons disease (Benabid and others esis of the field and the current practice in this area. We
1987; Odekerken and others 2016), essential tremor conclude with a brief comment on future directions.
(Baizabal-Carvallo and others 2014; Benabid and oth-
ers 1991), chronic pain (Boccard and others 2014), and
epilepsy (Fisher and others 2010). Brain stimulation is
1
Department of Neurosurgery, Alfred Hospital, Melbourne, Victoria,
also finding applications in neuropsychiatry, now
Australia
available as a treatment option in the management of 2
Department of Surgery, Central Clinical School, Monash University,
severe medication-resistant depression (Fitzgerald Clayton, Victoria, Australia
2013) and obsessive-compulsive disorder (Greenberg 3
Monash Institute of Medical Engineering, Monash University, Clayton,
and others 2010). Experimentally, electrical stimula- Victoria, Australia
4
Monash Alfred Psychiatry Research Centre, Central Clinical School,
tion of the brain is being investigated in a neuropsychi-
Monash University, Melbourne, Victoria, Australia
atric context as a novel treatment for autism (Enticott 5
F. Edward Hbert School of Medicine, Uniformed Services University
and others 2014), schizophrenia (Fitzgerald and others of the Health Sciences, Bethesda, MD, USA
2008), addiction (Amiaz and others 2009), and anorexia
Corresponding Author:
nervosa (McClelland and others 2016), while it is also Philip M. Lewis, Department of Surgery, Central Clinical School,
under investigation for the treatment of sensory defi- Monash University, Level 6 Alfred Centre, Clayton, Victoria, Australia.
cits including neural deafness (Otto and others 2008) Email: philip.lewis@monash.edu

Downloaded from nro.sagepub.com at INST DE FISIOLOGIA CELU LAR on May 22, 2016
2 The Neuroscientist

In particular, Aldini recommended electrical stimula-


tion of the brain for cases of melancholy madness,
apoplexy, and even syncope, in the belief that

. . . in these maladies, the brain, being particularly affected,


may probably, by this means, be re-established in the regular
use of its functions. (Aldini 1819, p. 65)

Throughout the 19th century, the use of electricity as a


therapeutic tool in neuropsychiatry became established
(Gilman 2008), and its ready availability in the laboratory
setting facilitated the resolution of the long-standing
debate on cortical functional localization (Fritsch and
Hitzig 1870). The use of electrical stimulation for map-
ping brain function continued into the 20th century, par-
ticularly by Penfield, who did extensive studies of brain
stimulation on patients undergoing surgical excision of
epileptic foci (Penfield 1947). However, toward the end
of the 19th century noninvasive brain stimulation for
Figure 1. A patient of Aldinis receiving treatment, with the
electrical circuit completed by the patients hand touching the
treating psychiatric illness began to fall out of favor
base of the Voltaic pile. Image reproduced from Aldini (1803). (Gilman 2008).
This trend was reversed in the 1930s when Cerletti
popularized electroconvulsive therapy (ECT; Fig. 2).
The Evolution of Brain Stimulation After initial success in treating a patient with schizophre-
nia (Accornero 1988), Cerletti expanded the scope of his
Direct Electrical Stimulation
treatments to include patients with mania and severe
Electricity has occupied a place in the therapeutic arma- depression, after which the technique became popular.
mentarium of clinicians for the past two millennia, includ- While Cerletti used a 90-V direct current pulse to induce
ing for the treatment of conditions involving the head. a seizure in his first patient (Accornero 1988), modern
Indeed, Scribonius Largus, physician to the Roman ECT employs alternating current of varying frequencies,
Emperor Tiberius, used electric shocks to the forehead, applied to one or both hemispheres.
administered by the electric torpedo fish, to treat headache Lower intensity noninvasive brain stimulation or
in the first century AD (Baldwin 1992). Fast-forwarding brain polarization (Priori and others 1998) continued to
to the 17th and 18th centuries, newly developed static be used throughout the early to mid-20th century for the
electricity generation and electrochemical charge-storage treatment of insomnia in a technique referred to as elec-
devices allowed extensive experimentation with and, ulti- trosleep (Nias 1976). This involved delivering short
mately, the widespread use of electricity as a therapeutic pulses of weak DC current across the temples; notably,
tool (Priestley 1767). The discovery by Galvani that unlike ECT electrosleep did not induce seizures. In 1998,
nerves and muscles were electrically excitable (Piccolino Priori etal confirmed that low-intensity brain polariza-
1997) led to Volta developing the first electrochemical tion alters cortical excitability, prompting further investi-
battery while investigating Galvanis theory of animal gation into its applications in neurology. The technique,
electricity (Piccolino 2000). Galvanis nephew Aldini contemporaneously referred to as transcranial direct cur-
later carried out numerous studies on the effects of elec- rent stimulation (tDCS), is currently being investigated in
tricity, or galvanism on the human body, during which he a variety of neurological contexts, including but not lim-
also investigated its therapeutic potential (Parent 2004). In ited to, improving cognitive performance (Floel 2014),
1803, Aldini described the successful treatment of a treating depression (Brunoni and others 2013) and
patient with severe melancholy by electrical stimulation enhancing motor recovery after brain injury (Liew and
of the head (Aldini 1803) (Fig. 1). others 2014).
In his 1819 book written in English, Aldini further out- The application of invasive brain stimulation to the
lined his thoughts on the therapeutic potential of galva- treatment of neurological disorders began in earnest in the
nism, stating that it will be beneficial to apply the 1950s, particularly after the development of stereotactic
galvanic fluid, as a mechanical stimulant in diseases methods for accurately placing electrodes deep within the
where an excitement of the nervous and muscular system brain substance (Spiegel and others 1947). From the 1950s
is necessary (Aldini 1819, p. 61). to the 1970s, a number of groups investigated deep brain

Downloaded from nro.sagepub.com at INST DE FISIOLOGIA CELU LAR on May 22, 2016
Lewis et al. 3

stimulation of the precentral gyrus (i.e., motor cortex)


could provide symptomatic relief for patients with central
pain (Brown 2001). Cortical stimulation is now also per-
formed therapeutically for tinnitus, depression and post-
stroke motor rehabilitation (Tronnier and Rasche 2013).
Of particular interest is the development of closed-loop,
or responsive neuromodulation systems for the auto-
mated termination of epileptic seizures, a technology
which also holds promise for the unattended treatment of
medically refractory major depression (Sun and Morrell
2014).
Neurostimulation of somatosensory and visual corti-
ces with neuroprosthetics continues to progress in the
experimental setting, however full clinical translation of
this research is yet to be achieved.

Magnetic Stimulation
Two decades after Fritsch and Hitzig used direct electri-
cal stimulation of the exposed cortex to map the canine
motor cortex, dArsonval demonstrated that the same
principles of electromagnetic induction Faraday had
exploited to generate electricity, could stimulate neural
tissue without requiring any physical connection to the
target. Notably, this discovery was made while dArsonval
was investigating the ability of high-strength alternating
Figure 2. Cerletti and Binis first electroshock device on display magnetic fields to induce heating in living animals
in the Museum of the History of Medicine, University of Rome, (Geddes 1999). Volunteers placed inside the coil reported
Italy. Image Francesco Pallone, reproduced with permission seeing phosphenes, experiencing vertigo and occasion-
under the terms of Creative Commons Licence CC-BY-SA-3.0
ally syncope (Geddes 1999).
from Wikimedia Commons. (https://commons.wikimedia.org/
wiki/File:Macchina_elettroshock_Ugo_Cerletti.jpg). DArsonval published in French, therefore his work
was largely overlooked by English and German research-
ers. Beer (1902) independently designed a similar device
stimulation (DBS) for the treatment or diagnosis of psy- for treating depression, however it was not widely used.
chiatric (Delgado and others 1952) and movement disor- Thompson (1910) also developed a stimulating coil large
ders (Bekhtereva and others 1963; Sem-Jacobsen 1966), enough to fit a human head, which was capable of induc-
epilepsy (Cooper 1978), and chronic pain (Hosobuchi and ing sensations of light and taste. Despite others develop-
others 1973). The demonstrated effectiveness, reversibil- ing similar devices (Barlow and others 1947; Dunlap
ity and lower morbidity of DBS for Parkinsonian tremor 1911; Magnusson and Stevens 1911), techniques for and
as compared with lesional surgery (Benabid and others applications of magnetic brain stimulation did not signifi-
1991) has led to its subsequent uptake as a surgical treat- cantly advance for the greater part of the 20th century.
ment of choice for this condition. DBS continues to be
explored as a therapeutic option in a variety of other con- Transcranial Magnetic Stimulation.The first transcranial
ditions, discussed in further detail later. magnetic stimulation (TMS) device capable of evoking
In parallel with the development of DBS, stimulation cortical activity was developed by Barker and colleagues
of the cortex for the treatment of neurological dysfunc- (Barker and others 1985) (Fig. 3). Reports of its potential
tion was being actively explored. For example, visual in neuropsychiatry quickly followed, with a number of
cortex stimulation, which was known to reproducibly studies suggesting a role for TMS in clinical depression
elicit small visual percepts or phosphenes (Penfield (George and others 1995; Grisaru and others 1994;
1947), was investigated from the late 1950s onward as a Hoflich and others 1993; Kolbinger and others 1995;
means of providing functional visual perception to the Pascual-Leone and others 1996). Extensive research in
blind (Lewis and others 2015). the period since has resulted in the application of both
The therapeutic applications of cortical stimulation conventional suprathreshold TMS and more recently,
were extended in 1991 with the discovery that epidural low-intensity TMS for the investigation and treatment of

Downloaded from nro.sagepub.com at INST DE FISIOLOGIA CELU LAR on May 22, 2016
4 The Neuroscientist

physical cathode is therefore referred to as cathodic


stimulation (Brocker and Grill 2013).
Beyond the direct activation of neurons by the applica-
tion of electric fields, modulation and/or propagation of
the stimulus can occur through trans-synaptic mecha-
nisms, which may be excitatory or inhibitory in nature.
The relative contributions of both factors to the spread of
activation and the behavioral manifestations of cortical
stimulation remains the subject of some debate (Histed
and others 2013; Tehovnik and Slocum 2013).

Epidural and Subdural Cortical Stimulation. Stimulation of


the cortical surface can be achieved by placing electrodes
on the dura (epidurally), or directly on the cortex itself
Figure 3. Image of Barker and colleagues first transcranial (subdurally). Both methods have been applied in a variety
magnetic stimulation (TMS) device. Reproduced from Barker of neurological conditions including chronic pain (Lefau-
and others (1985), with permission. cheur and others 2009; Tsubokawa and others 1991),
movement disorders (Moro and others 2011; Woolsey
a wide variety of neurological conditions, with the most and others 1979), tinnitus (De Ridder and others 2007),
widely studied indication being treatment-resistant major medically refractory partial seizures (Bergey and others
depressive disorder. 2015), and for the treatment of depression (Hajcak and
others 2010). Reported stimulation sites include motor
cortex, Heschls gyrus, speech areas, and the dorsolateral
Contemporary Neuromodulation prefrontal cortex (Tronnier and Rasche 2013).
Techniques Cortical stimulation is commonly used by neurosur-
geons during awake craniotomy to identify sensorimotor
Established
cortex and speech areas (Hervey-Jumper and others
Implanted Forms of Direct Electrical Stimulation.Despite 2015), while subcortical stimulation is increasingly used
its long history of use in medicine, the precise mecha- to avoid damage to deeper motor pathways during sur-
nisms underlying the efficacy of direct (i.e., invasive) gery for glioma resection (Schucht and others 2013).
electrical brain stimulation remain the subject of ongo- Epidural electrode placement forces electrical charge
ing investigation. From a biophysical standpoint, the to flow through the dura and cerebrospinal fluid before
electrical and structural inhomogeneity of brain tissue, reaching the cortex, which reduces stimulus focality and
particularly the variable directionality and excitability of increases stimulus thresholds relative to subdural elec-
axons, dendrites, and neuronal somata render accurate trodes (Bezard and others 1999; Kim and others 2011).
modeling of the effects of applied electric fields on brain Indeed, computational modeling studies have shown that
tissue a challenging task (Meffin and others 2014; 50% or less of the total current output during epidural
Miranda and others 2007). Notwithstanding these uncer- cortical stimulation (ECS) reaches the cortex, with the
tainties about stimulation of bulk brain tissue, the funda- bulk of the remainder shunted through cerebrospinal fluid
mental effects of externally applied electric fields on (CSF; Kim and others 2011; Manola and others 2005;
individual neurons are well described. By placing a neg- Wongsarnpigoon and Grill 2008). In patients with corti-
atively or positively charged electrode directly over or cal atrophy, the problem of current shunting is magnified
among the target neurons, those in the vicinity of the by the increased volume of CSF between the electrode
electrode become hyper- or depolarized, due to the and the cortical surface, which limits the utility of ECS in
combined influence of electronic current flow and altera- these patients (Tronnier and Rasche 2013).
tions in the conductance of membrane-bound, voltage- While subdural electrodes therefore offer the advan-
sensitive ion channels (Fig. 4; Brocker and Grill 2013). tage of more focal stimuli at lower currents, the requisite
This results in the suppression or elicitation respectively, opening of the dura increases the risk of both CSF
of action potentials in neuronal elements close to the leak and infection. Moreover, the threshold intensity for
electrode(s). Hyperpolarization in the vicinity of an elec- stimulus-induced seizures is expectedly lower for subdu-
trode can also produce depolarization and subsequent ral electrodes (Bezard and others 1999).
neuronal activation distant to the electrode via virtual
cathodes, resulting in anodic stimulation if the depolar- Intracortical Stimulation.Intracortical electrodes were
ization reaches threshold. Stimulation proximal to a investigated in the 1960s, as a means with which to

Downloaded from nro.sagepub.com at INST DE FISIOLOGIA CELU LAR on May 22, 2016
Lewis et al. 5

Figure 4. Illustration depicting cathodic versus anodic monopolar stimulation. During cathodic stimulation (A, left), current is
drawn from the axon of a neuron proximal to the electrode, resulting in depolarization. This results in current being drawn into
the axon from surrounding tissue distant to the electrode, hyperpolarizing the axon membrane and creating virtual anodes
at these sites. Note however that the strength of hyperpolarization at the virtual anodes is much weaker (approximately 5
times less) than the depolarization proximal to the electrode (cathode). Thus, during anodic stimulation (B, right), the same
phenomenon occurring in reverse results in remote depolarization at virtual cathodes. Given the lower amplitude of
depolarization at the virtual cathodes, a greater stimulus strength is required for this to result in neuronal firing. Note that the
electrodes depicted contain multiple electrodes, thus stimulation could be performed in a bipolar fashion, with one electrode
acting as the cathode, with the other as the anode. Figure adapted from Brocker and Grill (2013), with permission.

selectively stimulate small populations of neurons and at A drawback to the penetration of cortical tissue
lower stimulation currents (Doty 1965; Stoney and others required for ICMS is the host response to the indwelling
1968). Smaller electrodes that penetrate the cortical sur- foreign body, which can result in a fibrous (Woolley and
face also reveal laminar variations in behavioral responses others 2013) and/or glial (Polikov and others 2005;
(motor and sensory) and stimulus thresholds, which can Woolley and others 2013) encapsulation of the electrodes.
vary substantially between the cortical surface (layer I) This glial scar separates the electrode from viable neu-
and layer V (Koivuniemi and others 2011; Tehovnik and rons and increases the impedance of the electrodetissue
others 2003). The underlying principles of intracortical interface, although it may resolve over a period of months
microstimulation (ICMS) are common to that of epidural (Wang and others 2013). Future novel electrode designs
and subdural stimulation; however, the electrodes are may mitigate this phenomenon (Lewis and others 2015).
much smaller and are usually insulated, with a small area
of exposed electrode material through which stimulus Deep Brain Stimulation.Deep brain stimulation (DBS)
current is delivered (the stimulating surface). This surface involves the stereotactic implantation of lead-mounted
may be at or offset from the electrode tip (Fig. 5), and electrodes (Fig. 7) into deeper brain structures implicated
may have physical dimensions similar in scale to a single in the genesis of neurological disease. A large number of
pyramidal cell body (i.e., <100 m; Wang and others stimulation targets have been identified with the choice
2013). of target dependent on the underlying condition; how-
This fact, combined with laminar variations in neu- ever, there remains uncertainty about the most effective
ronal architecture and connectivity throughout the cor- target for many of the conditions treated with DBS. Com-
tex renders both the population of neurons stimulated mon targets in the treatment of movement disorders such
(Fig. 6), and thus the behavioral consequences of as Parkinsons disease, essential tremor, and dystonia
ICMS, very sensitive to small changes in electrode include the subthalamic nucleus, globus pallidus inter-
position (Overstreet and others 2013). nus, and the ventral intermediate nucleus of the thalamus

Downloaded from nro.sagepub.com at INST DE FISIOLOGIA CELU LAR on May 22, 2016
6 The Neuroscientist

Figure 5. Scanning electron micrograph of two intracortical microelectrode designs, the left (a), showing the conventional
arrangement of an exposed electrode tip through which stimulus current is delivered. The novel design on the right (b)
incorporates a 70-m length annulus of exposed electrode material, located approximately 500 m from the insulated electrode
tip. Figure 5a adapted from (Normann and others 1999), with permission. Figure 5b supplied courtesy of the Monash Vision
Group.

(Baizabal-Carvallo and others 2014; Odekerken and oth- development of DBS leads that incorporate smaller and
ers 2016; Pouclet-Courtemanche and others 2016). Con- greater numbers of electrodes, allowing for a more
versely, DBS for obsessive compulsive disorder may nuanced and targeted approach to stimulation.
target the anterior limb of the internal capsule, nucleus
accumbens or the inferior thalamic peduncle (Greenberg Transcranial Magnetic Stimulation. As described earlier, the
and others 2010; Jimenez-Ponce and others 2009). For a basic principles of magnetic neurostimulation derive
complete list of disorders and corresponding stimulation from Faradays experiments in the early 1830s, which
targets, the reader is referred to the recent review by demonstrated that an alternating current in a primary cir-
(Udupa and Chen 2015). cuit will induce the same in an isolated secondary circuit
The precise mechanisms by which DBS exerts its ther- if the two are in close proximity. During TMS, a signifi-
apeutic effect are still being determined; however, there is cant electrical charge is stored in capacitors, which is
growing recognition that the influence of DBS on brain periodically discharged through conducting coils to pro-
networks, of which the chosen targets form a component, duce a time-varying magnetic field. This magnetic field
is key to clinical efficacy (Alhourani and others 2015). induces an electric field in the brain, which itself consti-
Importantly, these networks are formed not only from tutes an electrically conductive medium analogous to a
fibers projecting directly to and from stimulation targets wire. Thus current flow occurs in the brain, which if suf-
and other brain regions but also from fibers of passage, ficiently strong, will depolarise neurons in the tissue
being axons in the vicinity of the target but that do not located just under the coil.
synapse directly with it (Agnesi and others 2013). Given TMS can be used to noninvasively stimulate neural
the structural and behavioral complexity of these net- tissue due to the lack of resistance to the passage of a
works, the task of determining precisely how DBS magnetic field across tissue including the skull. The mag-
achieves its therapeutic effect is a formidable one indeed. netic field strength, however, is significantly reduced in
As mentioned briefly earlier, DBS electrodes are typi- relationship to the distance between the stimulating target
cally implanted using stereotactic systems, ensuring pre- and the magnetic source, requiring a very strong (>1
cise placement within deep targets that may be very tesla) field strength at the face of the coil.
small; in the case of the ventrointermediate nucleus, the Commercially available TMS devices typically pro-
anteroposterior and mediolateral dimensions may be as duce two pulse types: a biphasic pulse or a monophasic
small as 2 4 mm, respectively (Vassal and others 2012). pulse, with a third variation called the half-sine pulse
Current-generation DBS electrodes are typically quad- (Fig. 9). A biphasic pulse is approximately sinusoidal and
ripolar in design (Fig. 8), allowing for precise configura- is generally of shorter duration than a monophasic pulse,
tion of stimulation site, focality, and polarity (Marks which involves a rapid rise to peak values, followed by a
2015). Further refinements may be realized through the slow decay back to zero. In commercially available

Downloaded from nro.sagepub.com at INST DE FISIOLOGIA CELU LAR on May 22, 2016
Lewis et al. 7

(Fitzgerald and others 2006). The classical observation


has been that low frequency (~1 Hz) stimulation for a
period of 15 minutes or longer induces a transient inhibi-
tion, or a decrease in activity, of the cortex (Chen and
others 1997), whereas stimulation at frequencies above 1
Hz has been shown to induce increased cortical activation
(Siebner and others 2000). The mechanisms underlying
such inhibition remain unclear although there are simi-
larities to long-term depression, a cellular experimental
phenomenon wherein repeated low frequency stimulation
reduces activity in individual synapses (Chen and others
1997). Conversely, some authors suggest that the
increased cortical activity in response to higher-frequency
rTMS may be due to a transient increase in the efficacy of
excitatory synapses (Pascual-Leone and others 1994),
analogous to the cellular phenomena of long-term poten-
tiation. Importantly, others have shown that the individ-
ual variations in response to rTMS can be significant;
indeed, some study subjects show an inverse response
(i.e., decreasing cortical excitability) to increasing rTMS
frequency, or even demonstrate no frequency dependence
at all (Hamada and others 2013; Maeda and others 2000).
Attempts to improve the efficacy of TMS have resulted
in the development of more complex rTMS stimulus pat-
terns that more closely mimic intrinsic patterns of neuro-
nal firing. For example, theta burst stimulation (TBS),
which involves the application of short-duration high-
frequency 50-Hz trains at 5 Hz or theta frequency, has
been shown to induce cortical plasticity in a much shorter
timeframe compared to conventional rTMS (Huang and
Figure 6. The results of a computational modeling study,
showing the total number of cells recruited by an intracortical others 2005). Notably however, the aforementioned indi-
electrode advanced through each cortical layer (A, top) and vidual variability in response to rTMS has also been seen
the percentage of total neuronal recruitment accounted in studies of TBS (Hamada and others 2013).
for by pyramidal neurons (B, bottom), as stimulus strength Stimulating at high frequencies has also been shown to
is increased. This demonstrates that increasing stimulus produce transient functional lesions in cortical areas
strength not only increases the volume of tissue stimulated, receiving stimulation (Flitman and others 1998; Pascual-
but also alters the distribution of neuronal subtypes activated.
Leone and others 1991). Therefore, rTMS may be used as
Figure reproduced from (Overstreet and others 2013) with
permission. a neurophysiological probe to test the functional integrity
of different cortical regions by either activating these
regions or inhibiting them.
stimulators, several types of coils are typically used. These The greatest area of application for rTMS techniques
include circular and figure-of-eight shaped coils. In gen- currently is in the treatment of neuropsychiatric disor-
eral, figure-of-eight shaped coils produce a stronger more ders. A robust series of studies has demonstrated that
focussed magnetic field with better spatial resolution of rTMS has significant therapeutic benefit in the treatment
activation compared to circular coils (Ueno and others of major depressive disorder. Studies have demonstrated
1988). Considerable development is now occurring of a the efficacy of both high frequency stimulation applied to
range of deeper brain stimulation coils capable of provid- the left dorsolateral prefrontal cortex and low frequency
ing TMS to a range of brain targets, not just on the cortical stimulation applied to the right dorsolateral prefrontal
surface. cortex (Berlim and others 2013; Berlim and others 2014;
Repetitive transcranial magnetic stimulation (rTMS) Fitzgerald and others 2003; Fitzgerald and Daskalakis
involves applying TMS stimulation pulses repetitively to 2011; George and others 2010; OReardon and others
the cortex, usually at a fixed stimulation frequency 2007). This body of research has led to the translation of
(Wassermann 1998). rTMS can either activate or inhibit rTMS into clinical practice where it is now approved for
cortical activity depending on stimulation frequency use, and increasingly used in clinical practice in a

Downloaded from nro.sagepub.com at INST DE FISIOLOGIA CELU LAR on May 22, 2016
8 The Neuroscientist

Figure 7. (a) Coronal magnetic resonance imaging (MRI) showing bilateral deep-brain stimulation electrodes, implanted for the
treatment of Parkinsons disease. (b) Chest radiograph showing typical placement of the subcutaneously implanted stimulator
hardware.

application of a low-amplitude (1-2 mA) DC to the brain


through two surface electrodes placed on the scalp (Paulus
2003). In the most commonly used method, electrode pads
covered with sponges are connected to a low-voltage
stimulation device. Stimulation is usually applied continu-
ally for a period of time, commonly between 15 and 20
minutes.
Since the resurgence of interest in the application of
tDCS in late 1990s, a series of neurophysiological studies
have demonstrated the capacity of tDCS to modulate brain
activity, opening its use to wider experimental and clinical
applications (Bikson and others 2012; Liebetanz and oth-
ers 2002; Nitsche and others 2003). Studies have demon-
strated that there are effects on cortical excitability under
both the stimulating anode (positive electrode) and cathode
(negative electrode) that are divergent in polarity; typi-
cally, an increase in cortical excitability is seen under the
anode, and a decrease under the cathode (Paulus 2003).
While this polarity model might be useful for predicting
Figure 8. An example of a quadripolar, or four-electrode, response in a general sense, it may be overly simplistic.
deep brain stimulation (DBS) lead. All rights reserved. Used Exposure of neurons to weak DC current influences neuro-
with the permission of Medtronic. nal behavior through a complex milieu of short- and long-
term effects at a local and regional level. These include the
growing number of countries. Research continues to aforementioned simple alterations in neuronal firing pat-
explore the potential therapeutic benefit of rTMS in a terns and cortical excitability that occur in a polarity-
number of other disorders, including obsessive compul- dependent manner (the somatic doctrine; Bikson and
sive disorder, schizophrenia, posttraumatic stress disor- others 2012), as well as other synaptically mediated
der, chronic pain, and substance abuse (Fitzgerald 2011). changes that themselves are implicated in the observed
longer term effects of tDCS on cortical excitability, net-
Transcranial Direct Current Stimulation. Transcranial direct work behavior and thus neuronal plasticity. (Bikson and
current stimulation (tDCS) is a method for modifying others 2012; Stagg and Nitsche 2011). From a clinical
the behavior of neurons and/or neuronal networks using standpoint, it has been shown that the response rate to
nondepolarizing electrical current. It involves the tDCS may only be as high as 50% (Wiethoff and others

Downloaded from nro.sagepub.com at INST DE FISIOLOGIA CELU LAR on May 22, 2016
Lewis et al. 9

Figure 9. Typical transcranial magnetic stimulation (TMS) pulse shapes. (a) Monophasic pulse. (b) Half-sine pulse. (c) Biphasic
pulse. Reproduced from Sommer and others (2006), with permission.

2014). Clearly more research is required to better under- tDCS, a direct nonvarying current is typically applied at a
stand the demographic and neurophysiological factors that single stimulation intensity. It remains unclear whether
predict the likely response to stimulation. this is the most efficient way of inducing changes in corti-
Notwithstanding the uncertainty surrounding individ- cal excitability with scalp administered electrical stimu-
ual responses, the ability of tDCS to modulate brain func- lation, and a variety of alternative approaches are under
tion in an excitatory or inhibitory direction in a noninvasive investigation. For example, during intermittent DCS, the
fashion has led to its rapid expansion as a tool in cognitive stimulation pulse is interrupted periodically at a specific
neuroscience. Stimulation may be applied during or prior interval and frequency (transcranial pulsed current stimu-
to the performance of a cognitive task as a way of tempo- lation (Fitzgerald 2014; Jaberzadeh and others 2014)). Ini-
rarily enhancing or inhibiting performance. Studies have tial research with this approach suggests that it might have
demonstrated that single sessions of tDCS can produce greater effects on corticospinal excitability when applied
transient improvement in a range of cognitive functions, to the motor cortex than standard tDCS (Jaberzadeh and
for example, working memory (Hoy and others 2013); others 2014). The second approach with a fixed stimula-
however, these improvements are limited in healthy con- tion frequency is transcranial alternating current stimula-
trols compared with patients (Hoy and others 2013). tion (tACS). tACS uses an alternating current, centered
Research is now exploring whether these benefits can be either on zero or with a current offset. tACS is an attrac-
applied in longer treatment paradigms in disorders such as tive brain stimulation approach as it can be potentially
Alzheimers disease and schizophrenia. With regards to applied at a frequency relevant to modulating intrinsic
the use of tDCS for cognitive enhancement in healthy neural oscillations (Frohlich 2014) although it is not clear
individuals, aside from questions of efficacy there are a whether this always will result in enhanced effects on
myriad of ethical considerations yet to be fully addressed. brain activity (Brignani and others 2013). A third variant
Noteworthy is that a recent survey of tDCS researchers of electrical stimulation is the application of transcranial
highlighted significant concerns about safety and misuse random noise stimulation (tRNS) where electrical activity
of tDCS by nonexperts (Riggall and others 2015). is modulated in a random fashion (potentially to overcome
Other potential applications of tDCS in neuropsychia- homoeostatic responses to externally applied current).
try include the treatment of depression; parallel to the tRNS can be used to alter cortical excitability (Terney and
situation with rTMS, the greatest body of research is others 2008) and its application in the treatment of schizo-
focused on the use of tDCS to modulate mood. To date a phrenia is being explored (Palm and others 2013).
series of relatively small studies have explored the effects
of tDCS in clinical depression. These were recently sum-
Novel Neuromodulation Techniques
marized in a meta-analysis, which suggests that tDCS has
antidepressant efficacy, but that further research is A variety of novel neuromodulation techniques are cur-
required to allow adequate translation of this into clinical rently being developed that show promise as future meth-
practice (Shiozawa and others 2014). ods of choice, depending on the application.

Other Noninvasive Electrical Stimulation Approaches. Low-Intensity Magnetic Fields.Chance observations of


Although often confused with tDCS, there are a number improved mood after magnetic resonance imaging (MRI)
of other forms of noninvasive electrical stimulation used scanning in depressed patients participating in a medica-
to potentially modulate cortical excitability. During tion trial, led to a formal investigation of the phenomenon

Downloaded from nro.sagepub.com at INST DE FISIOLOGIA CELU LAR on May 22, 2016
10 The Neuroscientist

Figure 10. (a) Commercially available, 500-m-diameter microcoil used to successfully stimulate retinal ganglion cells
magnetically. (b) The same coil after removal of insulation, showing the copper wires. (c) Computer simulation showing the
electric field distribution around a model of the same coil. Figure adapted from Bonmassar and others (2012), with permission.

by Rohan and others (2004). The authors of that study demonstrating modulation of brain activity using fields
found that, despite the relatively weak intracranial elec- one-tenth the strength of TMS (Gonzalez-Rosa and oth-
tric field induced by the MRI (0.7 V/m during MRI vs ers 2015; Oliviero and others 2011).
1-500 V/m for rTMS), 77% of the study subjects who
underwent MRI reported improved mood. In contrast, Micromagnetic Stimulation.Micromagnetic stimulation
only 30% of subjects who received a sham MRI reported refers to the generation of very small alternating mag-
mood improvement (Rohan and others 2004). These find- netic fields to stimulate neuronal firing, with the stimulat-
ings led to further investigation into what was subse- ing device implanted within or close to the relevant neural
quently termed low-field magnetic stimulation (LFMS), target. While the fundamental principles are the same as
in a double-blind, randomized, sham-controlled study of TMS, the efficacy of micromagnetic stimulation of neural
the mood-improving effects of LFMS in depressed tissue was only very recently demonstrated (Bonmassar
patients. Using a custom-built, tabletop LFMS device to and others 2012; Park and others 2013) (Fig. 10). Park
deliver the stimuli, the authors earlier findings were and colleagues demonstrated that stimulation of the audi-
reproduced in the larger cohort (n = 63), with significant tory nucleus in a hamster could be achieved using a com-
improvements in mood seen in LFMS-treated subjects mercially-available microcoil 600 m in length, excited
compared with controls (Rohan and others 2014). with a 300-mV, 50-s pulse. The advantages of reducing
In a similar fashion, transcranial pulsed electromag- the scale of a magnetic stimulation device are clear: A
netic fields (T-PEMF), adapted from its use in osteoar- reduction in the volume of stimulated tissue will offer
thritis treatment, also induces low-intensity subthreshold improved selectivity and potentially treatment efficacy.
electric fields in brain tissue (0.22 V/m) but employs a Moreover, the tissue encapsulation of an implanted
different waveform to LFMS (Martiny and others 2010). micromagnetic stimulating element should be overcome
The technique has also shown efficacy for the treatment by increasing the stimulus strength. Once a steady-state
of depression in double-blind, randomized controlled tri- biological response has been reached, such stimulating
als (Martiny and others 2010; Straaso and others 2014). elements should show stable long-term performance.
Further refinements to low-field magnetic stimulation Magnetic stimulation also demonstrates orientational
techniques may be possible by synchronizing stimulus selectivity, thereby using multiple such stimulating ele-
waveforms to the recipients individual alpha frequency, ments could permit even further control of the stimula-
as determined by EEG (Leuchter and others 2015). By tion of fibers with varying orientations.
targeting the resonant frequency of a particular cortical Unsolved problems with current methods of generat-
circuit, it may be possible to stimulate and/or entrain ing micromagnetic fields for neuromodulation include
these circuits using much lower energy than conventional high power consumption (and therefore heat generation),
suprathreshold TMS techniques (Leuchter and others which may preclude the development of an implantable
2015). device (Luan and others 2014).
Another alternative to dynamic techniques such as Ongoing work in this field may address these prob-
rTMS is static field stimulation, with several authors lems, for example using exotic materials to improve the

Downloaded from nro.sagepub.com at INST DE FISIOLOGIA CELU LAR on May 22, 2016
Lewis et al. 11

efficiency of magnetic stimulation, as recently demon- achieved using light. This ability is conferred by the deliv-
strated by Guduru and others (2015). The authors of that ery into a host organism of genes expressing for light-
study administered 10 g of 30-nm-diameter magneto- sensitive ion channels called opsins, of which rhodopsin is
electric particles into the tail vein of a mouse, forcing one that has been studied extensively (Boyden and others
them to cross the blood-brain barrier using a strong, 2005; Zemelman and others 2002). The development of
externally applied static magnetic field. The nanoparti- technology to support optogenetic neuromodulation is
cles reduced the strength of the external magnetic field progressing rapidly (Lee and others 2015), however sig-
required to modulate neural activity by 2 orders of mag- nificant challenges remain before the technique will see
nitude (Guduru and others 2015). A significant drawback translation into clinical practice. Williams and Denison
to micromagnetic stimulation is the need for cranial sur- (2013) summarized these challenges, which include the
gery to perform hardware implantation. It remains to be development of techniques for safe delivery and integra-
seen whether or not the risks of surgery will be out- tion of opsins into the target tissue, increasing the sensitiv-
weighed by the efficacy of such stimulation when com- ity of opsins to achieve low-power stimulation and
pared against alternative noninvasive techniques currently integrating the light delivery hardware with the target tis-
under development. sue in a durable manner. The recent development of flex-
ible, thin microprobes incorporating light-emitting diodes
Ultrasound. Ultrasound is emerging as a promising tech- and microfluidic channels may represent a significant step
nology for highly spatially focused, noninvasive neuro- forward in achieving this goal (Jeong and others 2015).
modulation. The excitation of peripheral nerves by
ultrasound was first observed by Harvey (1929). Later
studies confirmed that focused ultrasound (FUS) could Summary and Outlook
modulate the activity of mammalian brain tissue in vitro The development of techniques for influencing the activ-
(Rinaldi and others 1991) and in vivo (Fry and others ity of the human brain is occurring rapidly, with clinical
1958; Velling and Shklyaruk 1988). More recent work translation well underway. Further work in this field will
has shown alterations in human sensory evoked EEG not only refine existing techniques but also reveal new
responses during exposure to transcranial FUS (tFUS; methods for modulating the behavior of neural circuitry
Legon and others 2014), and the possibility of using tFUS with improved spatial, temporal, and functional selectiv-
as a diagnostic or therapeutic tool in neurology and/or ity, possibly requiring a multimodal approach. With con-
neuropsychiatry is being actively explored. temporary technology, significant improvements in the
Unlike very high-powered tFUS, which exploits the treatment of neurological and neuropsychiatric disease
thermal effects of ultrasound to ablate tissue, low-powered have already been realized, beyond that achievable using
or nonthermal tFUS for modulation of neural activity is purely pharmaceutical means. Further developments in
thought to act via the mechanical effects of ultrasound on this field will undoubtedly translate to superior clinical
nerve membranes (Tyler 2011). These effects include efficacy and an even more widespread adoption of neuro-
cavitation-induced cyclic deformation (expansion and con- modulation technologies in human health and disease.
traction) of the neuronal cell membranes (Krasovitski and
others 2011; Tyler 2011), acoustic pressure-induced mem- Authors Note
brane deformation and fluid disturbances such as microjets
Jeffrey V. Rosenfeld and Paul B. Fitzgerald are joint senior authors.
or streaming (Tyler 2011).
Ultrasound-induced neuromodulation has been
Declaration of Conflicting Interests
achieved at a wide range of frequencies, however Tufail
etal (2011) recommend frequencies between 0.2 and 0.7 The author(s) declared no potential conflicts of interest with respect
MHz, offering the best compromise between spatial to the research, authorship, and/or publication of this article.
selectivity, tissue absorption and penetration of the intact
skull (Tufail and others 2011). Notably however, one Funding
group reported that brain exposure to 8 MHz ultrasound The author(s) disclosed receipt of the following financial sup-
through the frontal bone can affect mood in humans port for the research, authorship, and/or publication of this arti-
(Hameroff and others 2013). An important consideration cle: PBF has received equipment for research from Medtronic
in this context is the fact that the long-term effects of non- Ltd, Magventure A/S, and Brainsway Ltd and funding for
thermal tFUS are as yet unknown. research from Cervel Neurotech.

Optogenetic Techniques. Optogenetic neuromodulation is a References


promising technique wherein spatially and temporally Accornero F. 1988. An eyewitness account of the discovery of
precise activation or inhibition of neural circuitry can be electroshock. Convuls Ther 4(1):409.

Downloaded from nro.sagepub.com at INST DE FISIOLOGIA CELU LAR on May 22, 2016
12 The Neuroscientist

Agnesi F, Johnson MD, Vitek JL. 2013. Deep brain stimulation: and meta-analysis of randomized, double-blind and sham-
how does it work? Handb Clin Neurol 116:3954. controlled trials. Psychol Med 44(2):22539.
Aldini J. 1803. An account of the late improvements in galva- Bezard E, Boraud T, Nguyen JP, Velasco F, Keravel Y, Gross
nism. London: Wiles & Taylor. 254 p. C. 1999. Cortical stimulation and epileptic seizure: a study
Aldini J. 1819. General views on the application of galvanism of the potential risk in primates. Neurosurgery 45(2):
to medical purposes. London, England: W. Clowes. 105 p. 34650.
Alhourani A, McDowell MM, Randazzo MJ, Wozny TA, Bikson M, Reato D, Rahman A. 2012. Cellular and network
Kondylis ED, Lipski WJ, and others. 2015. Network effects effects of transcranial direct current stimulation. In: Rossini
of deep brain stimulation. J Neurophysiol 114(4):210517. PM, editor. Transcranial brain stimulation. Boca Raton,
Amiaz R, Levy D, Vainiger D, Grunhaus L, Zangen A. 2009. FL: CRC Press. p 5591.
Repeated high-frequency transcranial magnetic stimulation Boccard SG, Fitzgerald JJ, Pereira EA, Moir L, Van Hartevelt
over the dorsolateral prefrontal cortex reduces cigarette TJ, Kringelbach ML, and others. 2014. Targeting the affec-
craving and consumption. Addiction 104(4):65360. tive component of chronic pain: a case series of deep brain
Baizabal-Carvallo JF, Kagnoff MN, Jimenez-Shahed J, Fekete stimulation of the anterior cingulate cortex. Neurosurgery
R, Jankovic J. 2014. The safety and efficacy of thalamic 74(6):62835.
deep brain stimulation in essential tremor: 10 years and Bonmassar G, Lee SW, Freeman DK, Polasek M, Fried SI, Gale
beyond. J Neurol Neurosurg Psychiatry 85(5):56772. JT. 2012. Microscopic magnetic stimulation of neural tis-
Baldwin B. 1992. The career and work of Scribonius Largus. sue. Nat Commun 3:921.
Rheinisches Museum fr Philologie 135(1):7482. Boyden ES, Zhang F, Bamberg E, Nagel G, Deisseroth K. 2005.
Barker AT, Jalinous R, Freeston IL. 1985. Non-invasive Millisecond-timescale, genetically targeted optical control
magnetic stimulation of human motor cortex. Lancet of neural activity. Nat Neurosci 8(9):12638.
1(8437):11067. Brignani D, Ruzzoli M, Mauri P, Miniussi C. 2013. Is transcra-
Barlow HB, Kohn HL, Walsh EG. 1947. Visual sensations nial alternating current stimulation effective in modulating
aroused by magnetic fields. Am J Physiol 148:3725. brain oscillations? PLoS One 8(2):e56589.
Beer B. 1902. Ueber das Auftraten einer objective Brocker DT, Grill WM. 2013. Principles of electrical stimula-
Lichtempfindung in magnetischen Felde. Klin Wochenschr tion of neural tissue. Handb Clin Neurol 116:318.
15:1089. Brown JA. 2001. Motor cortex stimulation. Neurosurg Focus
Bekhtereva NP, Grachev KV, Orlova AN, Latsuk SL. 1963. 11(3):E5.
Utilization of multiple electrodes implanted in the sub- Brunoni AR, Valiengo L, Baccaro A, Zanao TA, de Oliveira
cortical structure of the human brain for the treatment of JF, Goulart A, and others. 2013. The sertraline vs. electri-
hyperkinesis [in Russian]. Zh Nevropatol Psikhiatr Im S S cal current therapy for treating depression clinical study:
Korsakova 63:38. results from a factorial, randomized, controlled trial. JAMA
Benabid AL, Pollak P, Gervason C, Hoffmann D, Gao DM, Psychiatry 70(4):38391.
Hommel M, and others. 1991. Long-term suppression of Chen R, Classen J, Gerloff C, Celnik P, Wassermann EM,
tremor by chronic stimulation of the ventral intermediate Hallett M, and others. 1997. Depression of motor cortex
thalamic nucleus. Lancet 337(8738):4036. excitability by low-frequency transcranial magnetic stimu-
Benabid AL, Pollak P, Louveau A, Henry S, de Rougemont J. lation. Neurology 48(5):1398403.
1987. Combined (thalamotomy and stimulation) stereo- Cooper IS. 1978. Cerebellar stimulation in man. New York,
tactic surgery of the VIM thalamic nucleus for bilateral NY:Raven Press.
Parkinson disease. Appl Neurophysiol 50(1-6):3446. De Ridder D, De Mulder G, Verstraeten E, Seidman M, Elisevich
Berg JA, Dammann JF 3rd, Tenore FV, Tabot GA, Boback JL, K, Sunaert S, and others. 2007. Auditory cortex stimulation
Manfredi LR, and others. 2013. Behavioral demonstration for tinnitus. Acta Neurochir Suppl 97(Pt 2):45162.
of a somatosensory neuroprosthesis. IEEE Trans Neural Delgado JMR, Hamlin H, Chapman WP. 1952. Technique
Syst Rehabil Eng 21(3):5007. of intracranial electrode implacement for recording and
Bergey GK, Morrell MJ, Mizrahi EM, Goldman A, King- stimulation and its possible therapeutic value in psychotic
Stephens D, Nair D, and others. 2015. Long-term treatment patients. Confinia Neurol 12(56):3159.
with responsive brain stimulation in adults with refractory Doty RW. 1965. Conditioned reflexes elicited by electri-
partial seizures. Neurology 84(8):8107. cal stimulation of the brain in macaques. J Neurophysiol
Berlim MT, van den Eynde F, Daskalakis ZJ. 2013. Clinically 28:62340.
meaningful efficacy and acceptability of low-frequency Dunlap K. 1911. Visual sensations from the alternating mag-
repetitive transcranial magnetic stimulation (rTMS) for netic field. Science 33:6871.
treating primary major depression: a meta-analysis of Enticott PG, Fitzgibbon BM, Kennedy HA, Arnold SL, Elliot
randomized, double-blind and sham-controlled trials. D, Peachey A, and others. 2014. A double-blind, random-
Neuropsychopharmacology 38(4):54351. ized trial of deep repetitive transcranial magnetic stimula-
Berlim MT, van den Eynde F, Tovar-Perdomo S, Daskalakis tion (rTMS) for autism spectrum disorder. Brain Stimul
ZJ. 2014. Response, remission and drop-out rates following 7(2):20611.
high-frequency repetitive transcranial magnetic stimulation Fisher R, Salanova V, Witt T, Worth R, Henry T, Gross R, and
(rTMS) for treating major depression: a systematic review others. 2010. Electrical stimulation of the anterior nucleus

Downloaded from nro.sagepub.com at INST DE FISIOLOGIA CELU LAR on May 22, 2016
Lewis et al. 13

of thalamus for treatment of refractory epilepsy. Epilepsia Greenberg BD, Gabriels LA, Malone DA Jr, Rezai AR, Friehs
51(5):899908. GM, Okun MS, and others. 2010. Deep brain stimulation
Fitzgerald PB. 2011. The emerging use of brain stimulation of the ventral internal capsule/ventral striatum for obses-
treatments for psychiatric disorders. Aust N Z J Psychiatry sive-compulsive disorder: worldwide experience. Mol
45(11):92338. Psychiatry 15(1):6479.
Fitzgerald PB. 2013. Non-pharmacological biological treatment Grisaru N, Yaroslavsky U, Abarbanel JM, Lamberg T, Belmaker
approaches to difficult-to-treat depression. Med J Aust RH. 1994. Transcranial magnetic stimulation in depression
199(6 Suppl):S4851. and schizophrenia. Eur Neuropsychopharmacol 4:2878.
Fitzgerald PB. 2014. Transcranial pulsed current stimulation: a Guduru R, Liang P, Hong J, Rodzinski A, Hadjikhani A,
new way forward? Clin Neurophysiol 125(2):2179. Horstmyer J, and others. 2015. Magnetoelectric spin
Fitzgerald PB, Brown TL, Marston NA, Daskalakis ZJ, De on stimulating the brain. Nanomedicine (Lond) 10(13):
Castella A, Kulkarni J. 2003. Transcranial magnetic stimu- 205161.
lation in the treatment of depression: a double-blind, pla- Hajcak G, Anderson BS, Arana A, Borckardt J, Takacs I,
cebo-controlled trial. Arch Gen Psychiatry 60(10):10028. George MS, and others. 2010. Dorsolateral prefrontal
Fitzgerald PB, Daskalakis ZJ. 2011. The effects of repeti- cortex stimulation modulates electrocortical measures of
tive transcranial magnetic stimulation in the treatment of visual attention: evidence from direct bilateral epidural
depression. Expert Rev Med Devices 8(1):8595. cortical stimulation in treatment-resistant mood disorder.
Fitzgerald PB, Fountain S, Daskalakis ZJ. 2006. A comprehen- Neuroscience 170(1):2818.
sive review of the effects of rTMS on motor cortical excit- Hamada M, Murase N, Hasan A, Balaratnam M, Rothwell JC.
ability and inhibition. Clin Neurophysiol 117(12):258496. 2013. The role of interneuron networks in driving human
Fitzgerald PB, Herring S, Hoy K, McQueen S, Segrave R, motor cortical plasticity. Cereb Cortex 23(7):1593605.
Kulkarni J, and others. 2008. A study of the effectiveness Hameroff S, Trakas M, Duffield C, Annabi E, Gerace MB, Boyle
of bilateral transcranial magnetic stimulation in the treat- P, and others. 2013. Transcranial ultrasound (TUS) effects
ment of the negative symptoms of schizophrenia. Brain on mental states: a pilot study. Brain Stimul 6(3):40915.
Stimul 1(1):2732. Harvey EN. 1929. The effect of high frequency sound waves
Flitman SS, Grafman J, Wassermann EM, Cooper V, OGrady on heart muscle and other irritable tissues. Am J Physiol
J, Pascual-Leone A, and others. 1998. Linguistic process- 91:28490.
ing during repetitive transcranial magnetic stimulation. Hervey-Jumper SL, Li J, Lau D, Molinaro AM, Perry DW,
Neurology 50(1):17581. Meng L, and others. 2015. Awake craniotomy to maximize
Floel A. 2014. tDCS-enhanced motor and cognitive function in glioma resection: methods and technical nuances over a
neurological diseases. Neuroimage 85(Pt 3):93447. 27-year period. J Neurosurg 123(2):32539.
Fritsch G, Hitzig E. 1870. Electric excitability of the cerebrum Histed MH, Ni AM, Maunsell JH. 2013. Insights into cortical
(Crump T, Lama S, Trans.). Epilepsy Behav 15(2):12330. mechanisms of behavior from microstimulation experi-
Frohlich F. 2014. Endogenous and exogenous electric fields as ments. Prog Neurobiol 103:11530.
modifiers of brain activity: rational design of noninvasive Hoflich G, Kasper S, Hufnagel A, Ruhrmann S, Moller H-J.
brain stimulation with transcranial alternating current stim- 1993. Application of transcranial magnetic stimulation in
ulation. Dialogues Clin Neurosci 16(1):93102. drug-resistant major depressiona report of two cases.
Fry FJ, Ades HW, Fry WJ. 1958. Production of reversible Hum Psychopharmacol 8:361365.
changes in the central nervous system by ultrasound. Hosobuchi Y, Adams JE, Rutkin B. 1973. Chronic thalamic
Science 127(3289):834. stimulation for the control of facial anesthesia dolorosa.
Geddes LA. 1999. Retrospectroscope. dArsonval, physician Arch Neurol 29(3):15861.
and inventor. IEEE Eng Med Biol Mag 18(4):11822. Hoy KE, Emonson MR, Arnold SL, Thomson RH, Daskalakis
George MS, Lisanby SH, Avery D, McDonald WM, Durkalski ZJ, Fitzgerald PB. 2013. Testing the limits: investigating
V, Pavlicova M, and others. 2010. Daily left prefrontal the effect of tDCS dose on working memory enhancement
transcranial magnetic stimulation therapy for major depres- in healthy controls. Neuropsychologia 51(9):177784.
sive disorder: a sham-controlled randomized trial. Arch Huang YZ, Edwards MJ, Rounis E, Bhatia KP, Rothwell JC.
Gen Psychiatry 67(5):50716. 2005. Theta burst stimulation of the human motor cortex.
George MS, Wassermann EM, Williams WA, Callahan A, Neuron 45(2):2016.
Ketter TA, Basser P, and others. 1995. Daily repetitive Jaberzadeh S, Bastani A, Zoghi M. 2014. Anodal transcranial
transcranial magnetic stimulation (rTMS) improves mood pulsed current stimulation: a novel technique to enhance
in depression. Neuroreport 6(14):18536. corticospinal excitability. Clin Neurophysiol 125(2):
Gilman SL. 2008. Electrotherapy and mental illness: then and 34451.
now. Hist Psychiatry 19(75 Pt 3):33957. Jeong JW, McCall JG, Shin G, Zhang Y, Al-Hasani R, Kim
Gonzalez-Rosa JJ, Soto-Leon V, Real P, Carrasco-Lopez C, M, and others. 2015. Wireless optofluidic systems for pro-
Foffani G, Strange BA, and others. 2015. Static magnetic grammable in vivo pharmacology and optogenetics. Cell
field stimulation over the visual cortex increases alpha 162(3):662-674.
oscillations and slows visual search in humans. J Neurosci Jimenez-Ponce F, Velasco-Campos F, Castro-Farfan G,
35(24):918293. Nicolini H, Velasco AL, Salin-Pascual R, and others. 2009.

Downloaded from nro.sagepub.com at INST DE FISIOLOGIA CELU LAR on May 22, 2016
14 The Neuroscientist

Preliminary study in patients with obsessive-compulsive control: electrical potential field, activating functions and
disorder treated with electrical stimulation in the inferior responses of simple nerve fibre models. Med Biol Eng
thalamic peduncle. Neurosurgery 65(6 Suppl):2039. Comput 43(3):33543.
Kim D, Jun SC, Kim HI. 2011. Computational study of subdu- Marks WJ. 2015. Deep brain stimulation management. New
ral and epidural cortical stimulation of the motor cortex. York, NY: Cambridge University Press.
Conf Proc IEEE Eng Med Biol Soc 2011:72269. Martiny K, Lunde M, Bech P. 2010. Transcranial low voltage
Koivuniemi A, Wilks SJ, Woolley AJ, Otto KJ. 2011. pulsed electromagnetic fields in patients with treatment-
Multimodal, longitudinal assessment of intracortical resistant depression. Biol Psychiatry 68(2):1639.
microstimulation. Prog Brain Res 194:13144. McClelland J, Kekic M, Campbell IC, Schmidt U. 2016.
Kolbinger HM, Holflich G, Hufnagel A, Moller H-J, Kasper Repetitive transcranial magnetic stimulation (rTMS) treat-
S. 1995. Transcranial magnetic stimulation (TMS) in ment in enduring anorexia nervosa: a case series. Eur Eat
the treatment of major depressiona pilot study. Hum Disord Rev 24(2):15763.
Psychopharmacol 10:30510. Meffin H, Tahayori B, Sergeev EN, Mareels IM, Grayden DB,
Krasovitski B, Frenkel V, Shoham S, Kimmel E. 2011. Burkitt AN. 2014. Modelling extracellular electrical stimu-
Intramembrane cavitation as a unifying mechanism for lation: part 3. Derivation and interpretation of neural tissue
ultrasound-induced bioeffects. Proc Natl Acad Sci U S A equations. J Neural Eng 11(6):065004.
108(8):325863. Miranda PC, Correia L, Salvador R, Basser PJ. 2007. Tissue het-
Lee S, Williams P, Braine C, Lin DT, John S, Irazoqui P. 2015. erogeneity as a mechanism for localized neural stimulation
A miniature, fiber-coupled, wireless, deep-brain opto- by applied electric fields. Phys Med Biol 52(18):560317.
genetic stimulator. IEEE Trans Neural Syst Rehabil Eng Moro E, Schwalb JM, Piboolnurak P, Poon YY, Hamani C,
23(4):65564. Hung SW, and others. 2011. Unilateral subdural motor
Lefaucheur JP, Drouot X, Cunin P, Bruckert R, Lepetit H, cortex stimulation improves essential tremor but not
Creange A, and others. 2009. Motor cortex stimulation Parkinsons disease. Brain 134(Pt 7):2096105.
for the treatment of refractory peripheral neuropathic pain. Nias DK. 1976. Therapeutic effects of low-level direct electri-
Brain 132(Pt 6):146371. cal currents. Psychol Bull 83(5):76673.
Legon W, Sato TF, Opitz A, Mueller J, Barbour A, Williams Nitsche MA, Fricke K, Henschke U, Schlitterlau A, Liebetanz
A, and others. 2014. Transcranial focused ultrasound D, Lang N, and others. 2003. Pharmacological modula-
modulates the activity of primary somatosensory cortex in tion of cortical excitability shifts induced by transcranial
humans. Nat Neurosci 17(2):3229. direct current stimulation in humans. J Physiol 553(Pt 1):
Leuchter AF, Cook IA, Feifel D, Goethe JW, Husain M, 293301.
Carpenter LL, and others. 2015. Efficacy and safety of Normann RA, Maynard EM, Rousche PJ, Warren DJ. 1999. A
low-field synchronized transcranial magnetic stimulation neural interface for a cortical vision prosthesis. Vision Res
(sTMS) for treatment of major depression. Brain Stimul 39(15):257787.
8(4):78794. OReardon JP, Solvason HB, Janicak PG, Sampson S, Isenberg
Lewis PM, Ackland HM, Lowery AJ, Rosenfeld JV. 2015. KE, Nahas Z, and others. 2007. Efficacy and safety of
Restoration of vision in blind individuals using bionic transcranial magnetic stimulation in the acute treatment of
devices: a review with a focus on cortical visual prostheses. major depression: a multisite randomized controlled trial.
Brain Res 1595:5173. Biol Psychiatry 62(11):120816.
Liebetanz D, Nitsche MA, Tergau F, Paulus W. 2002. Odekerken VJ, Boel JA, Schmand BA, de Haan RJ, Figee M,
Pharmacological approach to the mechanisms of transcra- van den Munckhof P, and others. 2016. GPi vs STN deep
nial DC-stimulation-induced after-effects of human motor brain stimulation for Parkinson disease: three-year follow-
cortex excitability. Brain 125(Pt 10):223847. up. Neurology 86(8):75561.
Liew SL, Santarnecchi E, Buch ER, Cohen LG. 2014. Non- Oliviero A, Mordillo-Mateos L, Arias P, Panyavin I, Foffani
invasive brain stimulation in neurorehabilitation: local and G, Aguilar J. 2011. Transcranial static magnetic field
distant effects for motor recovery. Front Hum Neurosci stimulation of the human motor cortex. J Physiol 589(Pt
8:378. 20):494958.
Luan S, Williams I, Nikolic K, Constandinou TG. 2014. Otto SR, Shannon RV, Wilkinson EP, Hitselberger WE,
Neuromodulation: present and emerging methods. Front McCreery DB, Moore JK, and others. 2008. Audiologic
Neuroeng 7:27. outcomes with the penetrating electrode auditory brainstem
Maeda F, Keenan JP, Tormos JM, Topka H, Pascual-Leone A. implant. Otol Neurotol 29(8):114754.
2000. Interindividual variability of the modulatory effects Overstreet CK, Klein JD, Helms Tillery SI. 2013. Computational
of repetitive transcranial magnetic stimulation on cortical modeling of direct neuronal recruitment during intracorti-
excitability. Exp Brain Res 133(4):42530. cal microstimulation in somatosensory cortex. J Neural
Magnusson CE, Stevens HC. 1911. Visual sensations caused Eng 10(6):066016.
by the changes in the strength of a magnetic field. Am J Palm U, Hasan A, Keeser D, Falkai P, Padberg F. 2013.
Physiol 29:12436. Transcranial random noise stimulation for the treatment of
Manola L, Roelofsen BH, Holsheimer J, Marani E, Geelen J. negative symptoms in schizophrenia. Schizophr Res 146(1-
2005. Modelling motor cortex stimulation for chronic pain 3):3723.

Downloaded from nro.sagepub.com at INST DE FISIOLOGIA CELU LAR on May 22, 2016
Lewis et al. 15

Parent A. 2004. Giovanni Aldini: from animal electricity to Sem-Jacobsen CW. 1966. Depth-electrographic observa-
human brain stimulation. Can J Neurol Sci 31(4):57684. tions related to Parkinsons disease. Recording and elec-
Park HJ, Bonmassar G, Kaltenbach JA, Machado AG, Manzoor trical stimulation in the area around the third ventricle. J
NF, Gale JT. 2013. Activation of the central nervous sys- Neurosurg 24(1 Suppl):388-402.
tem induced by micro-magnetic stimulation. Nat Commun Shiozawa P, Fregni F, Bensenor IM, Lotufo PA, Berlim MT,
4:2463. Daskalakis JZ, and others. 2014. Transcranial direct current
Pascual-Leone A, Gates JR, Dhuna A. 1991. Induction of stimulation for major depression: an updated systematic
speech arrest and counting errors with rapid-rate transcra- review and meta-analysis. Int J Neuropsychopharmacol
nial magnetic stimulation. Neurology 41(5):697702. 17(9):144352.
Pascual-Leone A, Rubio B, Pallardo F, Catala MD. 1996. Siebner HR, Peller M, Willoch F, Minoshima S, Boecker H,
Rapid-rate transcranial magnetic stimulation of left dor- Auer C, and others. 2000. Lasting cortical activation after
solateral prefrontal cortex in drug-resistant depression. repetitive TMS of the motor cortex: a glucose metabolic
Lancet 348(9022):2337. study. Neurology 54(4):95663.
Pascual-Leone A, Valls-Sole J, Wassermann EM, Hallett M. Sommer M, Alfaro A, Rummel M, Speck S, Lang N, Tings T,
1994. Responses to rapid-rate transcranial magnetic stimu- and others. 2006. Half sine, monophasic and biphasic tran-
lation of the human motor cortex. Brain 117(Pt 4):84758. scranial magnetic stimulation of the human motor cortex.
Paulus W. 2003. Transcranial direct current stimulation (tDCS). Clin Neurophysiol 117(4):83844.
Suppl Clin Neurophysiol 56:24954. Spiegel EA, Wycis HT, Marks M, Lee AJ. 1947. Stereotaxic
Penfield W. 1947. Some observations on the cerebral cortex of apparatus for operations on the human brain. Science
man. Proc R Soc Lond B Biol Sci 134(876):32947. 106(2754):34950.
Piccolino M. 1997. Luigi Galvani and animal electricity: two Stagg CJ, Nitsche MA. 2011. Physiological basis of transcranial
centuries after the foundation of electrophysiology. Trends direct current stimulation. Neuroscientist 17(1):3753.
Neurosci 20(10):4438. Stoney SD Jr, Thompson WD, Asanuma H. 1968. Excitation
Piccolino M. 2000. The bicentennial of the Voltaic battery of pyramidal tract cells by intracortical microstimulation:
(1800-2000): the artificial electric organ. Trends Neurosci effective extent of stimulating current. J Neurophysiol
23(4):14751. 31(5):65969.
Polikov VS, Tresco PA, Reichert WM. 2005. Response of Straaso B, Lauritzen L, Lunde M, Vinberg M, Lindberg L,
brain tissue to chronically implanted neural electrodes. J Larsen ER, and others. 2014. Dose-remission of pulsating
Neurosci Methods 148(1):118. electromagnetic fields as augmentation in therapy-resistant
Pouclet-Courtemanche H, Rouaud T, Thobois S, Nguyen J-M, depression: a randomized, double-blind controlled study.
Brefel-Courbon C, Chereau I, and others. 2016. Long-term Acta Neuropsychiatr 26(5):2729.
efficacy and tolerability of bilateral pallidal stimulation to Sun FT, Morrell MJ. 2014. Closed-loop neurostimulation: the
treat tardive dyskinesia. Neurology 86(7):6519. clinical experience. Neurotherapeutics 11(3):55363.
Priestley J. 1767. The history and present state of electricity: Tehovnik EJ, Slocum WM. 2013. Two-photon imaging and the
with original experiments. London, England: Printed for activation of cortical neurons. Neuroscience 245:1225.
J. Dodsley, J. Johnson and T. Cadell. 503 p. Tehovnik EJ, Slocum WM, Schiller PH. 2003. Saccadic eye
Priori A, Berardelli A, Rona S, Accornero N, Manfredi M. movements evoked by microstimulation of striate cortex.
1998. Polarization of the human motor cortex through the Eur J Neurosci 17(4):8708.
scalp. Neuroreport 9(10):225760. Terney D, Chaieb L, Moliadze V, Antal A, Paulus W. 2008.
Riggall K, Forlini C, Carter A, Hall W, Weier M, Partridge B, Increasing human brain excitability by transcranial
and others. 2015. Researchers perspectives on scientific high-frequency random noise stimulation. J Neurosci
and ethical issues with transcranial direct current stimula- 28(52):1414755.
tion: an international survey. Sci Rep 5:10618. Thompson SP. 1910. A physiological effect of an alternating
Rinaldi PC, Jones JP, Reines F, Price LR. 1991. Modification magnetic field. Proc R Soc London (Biol) B82:396399.
by focused ultrasound pulses of electrically evoked Tronnier V, Rasche D. 2013. Epidural and subdural stimulation.
responses from an in vitro hippocampal preparation. Brain Handb Clin Neurol 116:34351.
Res 558(1):3642. Tsubokawa T, Katayama Y, Yamamoto T, Hirayama T, Koyama
Rohan M, Parow A, Stoll AL, Demopulos C, Friedman S, S. 1991. Chronic motor cortex stimulation for the treatment
Dager S, and others. 2004. Low-field magnetic stimulation of central pain. Acta Neurochir Suppl (Wien) 52:1379.
in bipolar depression using an MRI-based stimulator. Am J Tufail Y, Yoshihiro A, Pati S, Li MM, Tyler WJ. 2011.
Psychiatry 161(1):938. Ultrasonic neuromodulation by brain stimulation with tran-
Rohan ML, Yamamoto RT, Ravichandran CT, Cayetano KR, scranial ultrasound. Nat Protoc 6(9):145370.
Morales OG, Olson DP, and others. 2014. Rapid mood- Tyler WJ. 2011. Noninvasive neuromodulation with ultra-
elevating effects of low field magnetic stimulation in sound? A continuum mechanics hypothesis. Neuroscientist
depression. Biol Psychiatry 76(3):18693. 17(1):2536.
Schucht P, Moritz-Gasser S, Herbet G, Raabe A, Duffau H. 2013. Udupa K, Chen R. 2015. The mechanisms of action of deep
Subcortical electrostimulation to identify network subserv- brain stimulation and ideas for the future development.
ing motor control. Hum Brain Mapp 34(11):302330. Prog Neurobiol 133:2749.

Downloaded from nro.sagepub.com at INST DE FISIOLOGIA CELU LAR on May 22, 2016
16 The Neuroscientist

Ueno S, Tashiro T, Harada K. 1988. Localized stimulation of Repetitive Transcranial Magnetic Stimulation, June 5-7,
neural tissue in the brain by means of a paired configuration 1996. Electroencephalogr Clin Neurophysiol 108(1):116.
of time-varying magnetic fields. J Appl Phys 64:58624. Wiethoff S, Hamada M, Rothwell JC. 2014. Variability in
Vassal F, Coste J, Derost P, Mendes V, Gabrillargues J, Nuti C, response to transcranial direct current stimulation of the
and others. 2012. Direct stereotactic targeting of the ven- motor cortex. Brain Stimul 7(3):46875.
trointermediate nucleus of the thalamus based on anatomic Williams JC, Denison T. 2013. From optogenetic technologies to
1.5-T MRI mapping with a white matter attenuated inver- neuromodulation therapies. Sci Transl Med 5(177):177ps6.
sion recovery (WAIR) sequence. Brain Stimul 5(4):62533. Wongsarnpigoon A, Grill WM. 2008. Computational modeling
Velling VA, Shklyaruk SP. 1988. Modulation of the functional of epidural cortical stimulation. J Neural Eng 5(4):44354.
state of the brain with the aid of focused ultrasonic action. Woolley AJ, Desai HA, Otto KJ. 2013. Chronic intracortical
Neurosci Behav Physiol 18(5):36975. microelectrode arrays induce non-uniform, depth-related
Wang C, Brunton E, Haghgooie S, Cassells K, Lowery A, tissue responses. J Neural Eng 10(2):026007.
Rajan R. 2013. Characteristics of electrode impedance and Woolsey CN, Erickson TC, Gilson WE. 1979. Localization in
stimulation efficacy of a chronic cortical implant using somatic sensory and motor areas of human cerebral cortex
novel annulus electrodes in rat motor cortex. J Neural Eng as determined by direct recording of evoked potentials and
10(4):046010. electrical stimulation. J Neurosurg 51(4):476506.
Wassermann EM. 1998. Risk and safety of repetitive transcra- Zemelman BV, Lee GA, Ng M, Miesenbock G. 2002. Selective
nial magnetic stimulation: report and suggested guide- photostimulation of genetically chARGed neurons. Neuron
lines from the International Workshop on the Safety of 33(1):1522.

Downloaded from nro.sagepub.com at INST DE FISIOLOGIA CELU LAR on May 22, 2016

Anda mungkin juga menyukai