You are on page 1of 13

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/265474562

Modulation of brain plasticity in stroke: A novel model for neurorehabilitation

Article in Nature Reviews Neurology · September 2014


DOI: 10.1038/nrneurol.2014.162 · Source: PubMed

CITATIONS READS

627 4,323

11 authors, including:

Giovanni Di Pino Giovanni Pellegrino


Università Campus Bio-Medico di Roma Western University
143 PUBLICATIONS 4,790 CITATIONS 119 PUBLICATIONS 2,667 CITATIONS

SEE PROFILE SEE PROFILE

Giovanni Assenza Fioravante Capone


Università Campus Bio-Medico di Roma Università Campus Bio-Medico di Roma
153 PUBLICATIONS 3,194 CITATIONS 162 PUBLICATIONS 4,160 CITATIONS

SEE PROFILE SEE PROFILE

All content following this page was uploaded by Giovanni Assenza on 25 March 2015.

The user has requested enhancement of the downloaded file.


REVIEWS
Modulation of brain plasticity in stroke:
a novel model for neurorehabilitation
Giovanni Di Pino, Giovanni Pellegrino, Giovanni Assenza, Fioravante Capone, Florinda Ferreri,
Domenico Formica, Federico Ranieri, Mario Tombini, Ulf Ziemann, John C. Rothwell and
Vincenzo Di Lazzaro
Abstract | Noninvasive brain stimulation (NIBS) techniques can be used to monitor and modulate the
excitability of intracortical neuronal circuits. Long periods of cortical stimulation can produce lasting effects
on brain function, paving the way for therapeutic applications of NIBS in chronic neurological disease. The
potential of NIBS in stroke rehabilitation has been of particular interest, because stroke is the main cause
of permanent disability in industrial nations, and treatment outcomes often fail to meet the expectations of
patients. Despite promising reports from many clinical trials on NIBS for stroke recovery, the number of studies
reporting a null effect remains a concern. One possible explanation is that the interhemispheric competition
model—which posits that suppressing the excitability of the hemisphere not affected by stroke will enhance
recovery by reducing interhemispheric inhibition of the stroke hemisphere, and forms the rationale for
many studies—is oversimplified or even incorrect. Here, we critically review the proposed mechanisms of
synaptic and functional reorganization after stroke, and suggest a bimodal balance–recovery model that
links interhemispheric balancing and functional recovery to the structural reserve spared by the lesion. The
proposed model could enable NIBS to be tailored to the needs of individual patients.
Di Pino, G. et al. Nat. Rev. Neurol. advance online publication 9 September 2014; doi:10.1038/nrneurol.2014.162

Introduction
Stroke is among the principal causes of death worldwide, The lack of effective neurorepair after stroke and the
and although most stroke survivors achieve at least some limitations of functional recovery attained by physio­
spontaneous recovery after the ictus, it remains the main therapy have led researchers to consider alternative
Institute of Neurology, cause of permanent disability in Europe and the USA.1–3 approaches that improve the scope for recovery by
Campus Bio-Medico The only existing treatment for stroke is tissue plasmino­ enhancing the natural plasticity of the sensorimotor
University, Via Álvaro
del Portillo 200,
gen activator, which can salvage tissue at risk in the system. If recovery depends on ‘relearning’ new patterns
00128 Rome, Italy penumbra of a brain infarct and reduce future disability of activity to maximize the use of remaining u ­ ndamaged
(G.D.P., G.P., G.A., F.C., if administered within a few hours after stroke onset.4 brain, interventions that increase plasticity, and thereby
F.F., F.R., M.T., V.D.L.).
Laboratory of No treatments specifically designed to restore function the ability to learn, should improve recovery. Two
Biomedical Robotics through repair of damaged tissue have been developed approaches are of great current interest: pharmaco­logical
and Biomicrosystem,
Center for Integrated to date. Current best practice in stroke management is to interventions and noninvasive brain stimulation (NIBS).
Research, Campus Bio- reduce initial impact, take precautions to avoid the further In this Review, we focus on NIBS. Although NIBS has
Medico University, Via
Álvaro del Portillo 28,
burden of complications, and maximize f­unctional ability been used to improve gait, neglect symptoms and lan­
00128 Rome, Italy through extensive physiotherapy. guage after stroke, we will concentrate here on its appli­
(D.F.). Department of Although early rehabilitative interventions can improve cation to recovery of arm and hand function, because
Neurology & Stroke,
and Hertie Institute for arm function above what would be achieved by natural most studies have assessed the efficacy of NIBS on upper
Clinical Brain Research, recovery alone,5 the majority of patients are still unable limb function.
Eberhard Karls-
University Tübingen,
to use the affected hand and/or arm for simple activities More than 350 articles have been published on trans­
Hoppe-Seyler-Strasse 3, of daily living 6 months after the stroke.6 Robot-assisted cranial stimulation in stroke since 2012. These include
D‑72076 Tübingen, therapy has been proposed as an excellent low-cost way to more than 50 small clinical trials that evaluated the
Germany (U.Z.). Sobell
Department of Motor increase the amount of therapy that an individual patient potential for NIBS to enhance recovery of hand and arm
Neuroscience and receives, because a robot can deliver a higher amount function. Recent meta-analyses disagree on the effective­
Movement Disorders,
UCL Institute of
of exercise in the same time than can a human physio­ ness of these techniques: two Cochrane reviews do not
Neurology, Queen therapist,7 but in controlled trials robot-assisted therapy support the use of repetitive transcranial magnetic stimu­
Square, London has shown disappointingly little additional benefit over lation (rTMS)10 or transcranial direct current stimu­lation
WC1N 3BG, UK (J.C.R.).
­conventional regimes of intensive physiotherapy.8,9 (tDCS)11 in stroke rehabilitation, whereas two other sys­
Correspondence to: tematic reviews conclude that NIBS can have a moder­
V.D.L.
V.DiLazzaro@ Competing interests ate benefit in terms of stroke recovery, with few major
unicampus.it The authors declare no competing interests. adverse effects.12,13 Here, we discuss the factors that have

NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION | 1


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Key points the gluta­matergic synapse, hypoxic failure leads to focal


post­synap­tic hyperpolarization18,19 followed by a massive
■■ Noninvasive brain stimulation (NIBS) is a promising approach to enhance
hypoxic depolarization of the postsynaptic terminal.20
recovery after stroke, but its beneficial effect is limited and the technique is
not yet ready for broad clinical use Even though inhibitory synapses are more resistant to
■■ We suggest that the disappointments in NIBS trials are related to over-reliance hypoxia than are excitatory synapses,21 synaptic disinhib­
on the interhemispheric competition and vicariation models of recovery, which ition fails before synaptic excitation because of reduced
are oversimplified and do not apply to all patients with stroke excitatory input to inhibitory interneurons,22 poten­
■■ The concept of ’structural reserve’ integrates the effects that interhemispheric tially c­ ontributing to excitotoxicity that causes delayed
inhibition and vicariation exert on the unlesioned residual network cell death.
■■ We propose a unified ‘bimodal balance–recovery model’ that takes into account
During days and weeks following stroke, slower syn­
this individual residual network
■■ The model could be used to tailor treatment for individual patients and increase
aptic changes can emerge. A critical period of approxi­
the efficacy of NIBS in stroke rehabilitation mately 90 days of increased synaptic plasticity is thought
to parallel the initial period of rapid behavioural recov­
ery.5,23 Two factors exert opposing effects during this
Box 1 | TMS protocols for assessment of brain excitability and connectivity period. First, in mouse models of stroke, a sustained
increase in tonic GABAergic signalling—lasting over
TMS has been widely used to investigate excitability and connectivity in M1 after
a month—can reduce plasticity; during this period
stroke. Single-pulse, paired-pulse and repetitive TMS protocols probe different
aspects of excitatory and inhibitory function (Figure 1). behavioural recovery can be enhanced by blocking
extrasynaptic GABA receptors.24 Simultaneously, a sus­
A single pulse of TMS applied over M1 can produce a peripheral muscular
response, or MEP. TMS that triggers MEPs is called suprathreshold TMS, whereas tained increase in glutamatergic excitability mediated by
TMS that is not of sufficient intensity to trigger MEPs is called subthreshold TMS. AMPA receptors promotes plasticity and the release of
The threshold and recruitment of MEPs at different intensities of stimulation ­brain-derived neurotropic factor.25
provide information about the level of corticospinal excitability. Data from rodent studies have shown that stroke-
The effect of GABA receptor type A‑modulated inhibitory circuits on corticospinal related changes can also involve brain regions in the
neurons can be probed by short-interval intracortical inhibition paradigm hemisphere unaffected by stroke. These changes
(Figure 1a).146,147 The role of GABA receptor type B‑expressing interneurons can be include bihemispheric reductions in phasic (synaptic)
probed by the controlateral cortical silent period protocol, in which a single TMS GABAergic inhibition,26 in parallel with downregula­
pulse is delivered during tonic contralateral hand/arm contraction,148 or by the tion of GABAA receptor binding and GABAA receptor
long-interval intracortical inhibition protocol.149 Other inhibitory mechanisms can
subunit expression.27 Changes in calcium currents have
be evaluated with short-latency afferent inhibition, which depends on GABAergic
and cholinergic circuits.150 Studies using these protocols have elucidated the also been demonstrated in the noninfarcted tissue of the
involvement of GABAergic circuits in intracortical inhibition,148,151,152 whereas unaffected hemisphere.28
the origin and nature of the increased excitability of the intracortical facilitation
(Figure 1b) is still poorly defined. Excitability and connectivity in stroke
TMS can also be paired with neuroimaging or electrophysiological techniques.153,154 Several methods and protocols are available for assess­
TMS–EEG could aid probing of the functional impact of a stroke lesion on the ing the altered excitability and connectivity after stroke,
connectivity of the whole brain, even when the lesion is not located at the site of both in acute and chronic phases. (Box 1, Figure 1). In
stimulation.155 In TMS–EEG, a TMS pulse evokes an initial activation of the target this article, we have adopted the following classification
area, followed by later effects that are attributable to activity triggered by axonally of the phases of stroke: hyperacute: the first 6 h post-
and synaptically propagated signals.145
ictus; acute: 6–24 h post-ictus; subacute: 24 h to 6 weeks
Abbreviations: GABA, γ-aminobutyric acid; M1, primary motor cortext; MEP, motor evoked
potential; TMS, transcranial magnetic stimulation. ­post-ictus; chronic: greater than 6 weeks post-ictus.12,29
During the acute phase after stroke, TMS of the
affected hemisphere is often unable to elicit motor
contributed to the slow progress of accepting NIBS as evoked potentials (MEPs), even at maximum deliverable
a part of routine clinical practice, and suggest ways to stimulation intensity. In patients with preserved MEPs,
develop the use of NIBS in stroke rehabilitation. the motor thresholds are generally higher and MEPs are
smaller compared with those recorded from the unaffec­
Synaptic dysfunction in stroke ted hemisphere or from healthy individuals. Within the
Noninvasive neuromodulatory strategies are thought to first few months, MEPs may reappear and gradually
alter synaptic efficacy in glutamatergic and γ-aminobutyric increase in amplitude,30–33 while the motor threshold
acid-mediated (GABAergic) circuits,14 which are essen­ tends to decrease.34–38 Many authors have reported that
tial for motor learning. Knowing how stroke alters these TMS measures of corticospinal tract integrity evaluated
circuits, and determining the time course over which the early after stroke,35,39–41 and the improvement of TMS
stroke-related changes occur, might aid the development measures of corticospinal integrity during the first few
of more-effective n ­ euromodulatory strategies. months33,39 of recovery, both correlate with the long-
Brain infarcts lead to an imbalance in energy demand term functional outcome. Absence of MEPs in the first
and blood supply. The majority of energy consumed few hours or days following stroke is associated with
by the brain is devoted to synaptic transmission and poor clinical recovery,42 although exceptions have been
the postsynaptic effects of glutamate,15 and interrup­ reported.36,44–46 Presence or absence of MEPs in arm
tion of synaptic transmission accounts for the electrical muscles within the first week after stroke is an impor­
silence in the penumbra after stroke.16,17 At the level of tant component of the ‘PREP algorithm’ proposed by

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

a Subthreshold TMS TMS ◀ Figure 1 | TMS paired-pulse protocols. The approach is also
outlined in Box 1. a | TMS paired-pulse protocols for testing
– intracortical inhibitory circuitry. In SICI, a subthreshold TMS
1–5 ms
SICI ISI stimulus is followed by a subthreshold TMS pulse after a
Conditioning stimulus Test stimulus 1–5 ms delay. In LICI, a suprathreshold conditioning
TMS Suprathreshold TMS stimulus precedes the test stimulus by 50–200 ms. In cSP,
a TMS pulse is delivered during tonic contralateral hand/
– arm contraction. b | Intracortical facilitation is measured by
50–200 ms
LICI ISI ICF, in which a subthreshold conditioning pulse is followed
by a TMS pulse after a 8–30 ms delay. c | Protocols for
20% max Contralateral M1 TMS testing afferent inhibition. In SAI and LAI, a peripheral
contraction suprathreshold
stimulation stimulus is paired with a TMS pulse on M1. In SAI, ISI is
Silent period – shorter (N20lat + 2–5 ms) than in LAI (N20lat + 100–300 ms)
EMGA
cSP ISI d | iSP and IHI protocols evaluate transcallosal inhibitory
drive. Abbreviations: cSP, contralateral cortical silent period;
EMGA, electromyographic activity; ICF, intracortical
facilitation; IHI, interhemispheric inhibition; ISI, interstimulus
b Subthreshold TMS TMS interval; iSP, ipsilateral cortical silent period; LAI, long-
latency afferent inhibition; LICI, long-interval intracortical
+
8–30 ms inhibition; M1, primary motor cortex; MNS, median nerve
ICF ISI stimulation; N20lat, latency of the N20 component of
somatosensory evoked potentials; SAI, short-latency
afferent inhibition; SICI, short-interval intracortical inhibition;
TMS, transcranial magnetic stimulation.
c Suprathreshold MNS TMS


N20lat + 2–5 ms Short-interval intracortical inhibition (SICI)36,48–50 and
SAI ISI
long-interval intracortical inhibition (LICI)50—­indicators
MNS Suprathreshold TMS of GABAA and GABAB receptor-mediated inhib­ition
(Figure 1a)—are suppressed, whereas intracortical

N20lat + 100–300 ms facilitation (ICF; Figure 1b) remains normal.48,50,51 In a
LAI ISI recent study from our group, short-latency afferent inhib­
ition (SAI; Figure 1c)—a marker of central cholinergic
activity—­was suppressed in the acute phase of stroke,
Max Ipsilateral M1 with increased suppression of SAI correlating with better
d suprathreshold TMS
contraction
stimulation motor function recovery at 6 months.52 Although motor
EMGA Silent period – threshold and MEP amplitude in the unaffected hemi­
iSP ISI sphere are generally within normal limits,32,36,43,45,46,51,53–55
there are some reports of a reduction of SICI.36,51,54,56
TMS can be used to investigate interhemispheric con­
Ipsilateral M1 TMS TMS Contralateral M1
subthreshold stimulation nectivity between the two motor cortices, using a paired-
stimulation coil method to test interhemispheric inhib­ition (IHI;

10–40 ms
IHI ISI
Figure 1d). In stroke survivors, IHI from the unaffected
hemisphere to the affected hemisphere abnormally per­
sists at the onset of attempted contraction of the paretic
limb,31,57 and could potentially interfere with initiation
of movement.
Longitudinal studies describing detailed neurophysio­
logical and clinical data over the first few months after
stroke are scarce, but in one small study, neurophysio­
logical measures of corticospinal integrity—such as MEP
TMS MNS EMGA and motor threshold—in the affected hemisphere were
found to correlate with motor function in the acute and
– Protocol to measure inhibition + Protocol to measure facilitation subacute phase, whereas at >3 months, motor function
correlated better with measures of intracortical excit­
ability (SICI, LICI and ICF) than with measures of corti­
Stinear et al.,47 in which recovery prospects of individual cospinal integrity.50 Although this study comprised only
patients can be forecast through a combination of clinical 10 patients, the data are consistent with the hypothesis
upper limb strength measures, TMS response, and MRI that intracortical and corticospinal excitability probe
­measures of corticospinal tract asymmetry. different aspects of the recovery process. In the acute
After stroke, the balance between inhibition and excita­ period, motor performance relies on remaining cor­
tion in the affected hemisphere shifts towards excitation, ticospinal connections of the affected hemisphere,
while activity in the local inhibitory circuits is reduced. while sub­s equent recovery may depend more on the

NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION | 3


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Acute stroke that exceeds the activation seen in healthy individuals


performing movements of the same difficulty.36,49,54,56
aH uH The extent of this activity seems to depend on the degree
SICI Normal MEP
LICI Normal MTs
of functional impairment, being greatest in patients with
Normal ICF SICI the greatest impairment.
IHI aH uH IHI uH aH Is increased activation of the premotor cortex, SMA
∆power
and unaffected hemisphere an epiphenomenon, or does
activity in those areas compensate for the lost motor func­
tion and, if so, through what mechanisms? Attempts to
Bad prognosis Good prognosis address these questions have used TMS to disrupt activ­
ity in selected cortical areas to see if there is any impact
aH uH aH uH on movement of the paretic arm. In patients with sub­
No MEP MEP LTP-like LTD-like
MTs MTs SAI cortical stroke, disruptive TMS impairs simple reaction
∆power MEP present time of the paretic hand when applied over the dorsal
premotor cortex (PMd) of both the unaffec­ted66 and the
Figure 2 | Neurophysiological parameters in acute stroke predict prognosis. affected hemisphere.55 In well-recovered patients with
Schematic illustration of the typical changes in neurophysiological parameters that chronic stroke, complex finger movements were also dis­
characterize the acute phase in stroke, and predictors of bad and good recovery. rupted by TMS to M1 and the superior parietal lobe of
Abbreviations: aH, affected hemisphere; ICF, intracortical facilitation; IHI,
the unaffected hemisphere.67 The conclusion is that activ­
interhemispheric inhibition; LICI, long-interval intracortical inhibition; LTD, long-term
depression; LTP, long-term potentiation; MEP, motor-evoked potential; MT, motor ity in these areas can be compensatory and contribute
threshold; SAI, short-latency afferent inhibition; SICI, short-interval intracortical to recovery of movement. No similar evidence exists for
inhibition; uH, unaffected hemisphere; Δpower, power spectral density in the delta involvement of other brain areas, particularly in less well-
band (<4 Hz) EEG. recovered patients; however, several clinical case reports
indicate that new infarcts affecting the previously unaffec­
recruitment of alternative cortical networks in both ted hemisphere worsen the primary hemiparesis.68–71
hemispheres.50 According to this view, intracortical dis­ Similar data have been reported in animals. For example,
inhibition of the affected hemisphere (as indicated by application of lidocaine to the unaffected hemisphere
reduced SICI, LICI and SAI) might facilitate remain­ of rats 3–4 weeks after occlusion of the middle cerebral
ing motor output in the acute stage, and subsequently artery on the contralateral side reduced performance of
facilitate synaptic plasti­city to promote r­eorganization the paretic forelimb.72 Interestingly, the effect was most
of cortical networks in the subacute phase.58 dramatic in animals with large lesions and was much less
Taken together, the results from the studies reviewed noticeable in animals with small lesions.
above suggest that reduced corticospinal excitability in Direct projections from the PMd to the lower motor
the affected hemisphere increases intracortical excitabil­ neurons are scarce,73,74 making it unclear how increased
ity in both hemispheres and changes the balance of inter­ activity in the PMd can be transformed into functional
hemispheric interactions (Figure 2). However, despite the benefit. One possible mechanism is involvement of pro­
large number of investigations performed so far, different jections from the PMd to reticulospinal motor pathways
groups have reported highly variable and even opposite in the brainstem, some of which innervate distal arm and
results, leading to some uncertainty about the clinical hand muscles.75 Increased activity in the PMd might also
significance of these findings. simply increase the drive to any remaining corticospinal
projections from the affected hemisphere, thereby boost­
Plasticity changes during recovery ing the output projecting to the hand. Increased activity
Neuronal reorganization and plasticity that follow stroke in M1 of the unaffected hemisphere might contribute to
begin in the very early stages, continue for several weeks, recovery via the small percentage of uncrossed cortico­
and involve brain regions distant to the affected site. spinal fibres that can innervate paretic muscles on the
Imaging studies (PET, EEG and functional MRI) have ipsilateral side. Finally, if the SMA contributes to recov­
revealed widespread changes in patterns of brain acti­ ery of motor function, its extensive input directly to the
vation during simple movements of the affected hand corticospinal tract could enable this region to provide
after stroke; these alterations evolve over a timescale important input to spinal motor neurons. In line with this
that is consistent with a gradual reorganization of the hypothesis, findings from squirrel monkeys indicate that
sensori­motor system. Activation in the primary motor 3 months after stroke, expansion of the distal representa­
cortex (M1) of the affected hemisphere is reduced, tion of the paretic forelimb into the SMA of the affected
and—­depending on the size and location of the ischae­ hemisphere is proportional to the size of M1 lesion.76 Of
mic lesion—the activity can be relocated to more-­ note, in rats, recovery from a unilateral cerebral lesion
posterior 59–61 or more-anterior regions,46 particularly can be accompanied by sprouting of corticospinal fibres
towards the premotor cortex and supplementary motor from the intact hemisphere at the level of the spinal cord:
area (SMA), which are often spared by a typical middle the fibres branch and re-cross the spinal cord to inner­
cerebral artery occlusion.62–65 vate neurons on the paretic side.77 However, no evidence
The unaffected hemisphere can also show widespread for spinal cord level reinnervation from the contralateral
activation accompanying movements of the paretic hand affected hemisphere exists in human patients.

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Current models of functional recovery


The studies described above that used TMS to disrupt
the function of motor areas suggest that activity in the
unaffec­ted hemisphere is likely to contribute to func­
Max
tional recovery after stroke; this pattern of reorganization
High
probability
is referred to as the vicariation model, in which activity
in residual networks substitutes for those functions lost
by damaged areas. In practice, this presumably occurs
via the intact, unaffected hemisphere.83 However, an
Recovery

influential model of hemispheric interactions—the


1 interhemispheric competition model—suggests exactly
3 the opposite. This model assumes the presence of a
mutual, balanced inhibition between the hemispheres
in the healthy brain. Damage to one hemisphere in
Min
Low stroke disrupts this balance, with reduced inhibition of
2 probability
Max the unaffec­ted hemisphere by the affected hemisphere,
Str
uc
tur ncing resulting in increased inhibition of the affected hemi­
al ala sphere by the unaffected hemisphere. The affected
res ic b
er v pher
e mis ­h emisphere is thus thought to be ‘double-­d isabled’,
rhe
Inte because ipsilateral damage is coupled with excess
Max Min
­inhibition from the o­ pposite hemisphere.
Figure 3 | The bimodal balance–recovery model. Structural reserve is the quantity
of strategic neural pathways and relays that are spared by the lesion and can In healthy individuals, IHI decreases and reverses to
reallocate previous or outsource new functions. Patients with high structural reserve facilitation shortly before the onset of unilateral index
often achieve better functional recovery. In such cases, the balance of activity finger movements. In relatively well-recovered stroke
between the two hemispheres tends toward the previous equilibrium (1), whereas patients who are able to make index finger movements
persistence of interhemispheric imbalance is a predictor of worse outcome (2). with the affected hand, this mechanism fails because the
When structural reserve is low—such as in a subclass of patients with more-severe inhibition–facilitation reversal is lacking, potentially
impairment—and the sensorimotor network is far from reaching a physiological
interfering with movement of the paretic side.57,84 These
restitutio ad integrum, persistence of interhemispheric imbalance promotes vicarious
activity of the unaffected hemisphere (3), allowing compensatory plasticity. Colour
data are in line with the interhemispheric competition
code indicates frequency distribution of an event in a population of stroke patients, model; however, the results suggest abnormal control of
which is equivalent to the probability that a given individual is represented by that interhemispheric interaction during task performance
point of the surface. ‘Hot’ colours indicate higher probability than ‘cold’ colours. The only. It is important to note that resting levels of IHI are
probability distribution presented here follows a bimodal statistical distribution, generally reported to be within the normal range after
which arises from the superimposed distributions of two populations: patients with stroke, whereas the interhemispheric competition model
high reserve and high balancing versus patients with low reserve and low balancing. would predict IHI from the affected to the unaffected
hemisphere to be reduced, and IHI from the unaffected
Although the factors that determine the reorganiza­ to the affected hemisphere to be increased.
tion of activity after stroke remain unknown, bilateral Other data support the interhemispheric competition
activity accompanies certain unilateral movements even model. In healthy volunteers, reducing somatosensory
in healthy individuals. Simple movements, for example, input from one hand by cutaneous anaesthesia improves
moving the wrist or one finger, involve unilateral activity performance in the unanesthetized hand.85 Similarly, in
in the contralateral primary sensorimotor and premotor patients with chronic stroke, cutaneous anaesthesia of
cortex, whereas more-complex or sequential movements the intact hand results in behavioural gains in the paretic
can involve the same areas bilaterally, together with hand that briefly outlast the duration of the anaesthesia.86
the SMA and possibly other areas. The complexity of the The two models of reorganization—vicariation and
task correlates with the amount of involvement of non- interhemispheric competition—lead to opposite predic­
primary and ipsilateral areas of cortex.78,79 It seems pos­ tions about whether the optimal neuromodulatory treat­
sible that attempted movements of a paretic limb could ments for an individual patient would be inhibitory or
be processed in the brain in the same way as complex excitatory. Interhemispheric competition predicts that
movements of the limb in healthy individuals, thereby disruption of activity in the unaffected hemisphere would
recruiting similar areas to produce movement. be beneficial for stroke recovery by releasing the affected
Reorganization of activity can also have maladaptive hemisphere from abnormal inhib­ition by the unaffec­
effects. Stroke often hampers bimanual performance,80,81 ted hemisphere. Conversely, the vicariation model pre­
an impairment that has recently been linked with dicts that such a strategy would be counter­productive,
reduced task-related facilitatory activity from SMA onto because it would interfere with c­ ompensatory activity in
M1 in the affected hemisphere and disrupted control of the ­unaffected hemisphere.
inhibitory connections between the M1 regions of the
two hemispheres. Thus, impaired bimanual perfor­ The bimodal balance–recovery model
mance could be explained by abnormal interhemispheric We suggest that neither the interhemispheric competi­
­interaction of primary and supplementary motor areas.82 tion model nor the vicariation model is sufficient on its

NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION | 5


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Box 2 | Strategies for neuromodulation in humans Some evidence supporting the bimodal balance–­
recovery model has come from studies assessing the
Noninvasive neuromodulatory techniques are divided into inhibitory and excitatory
protocols on the basis of their effects on the amplitudes of motor evoked potentials
efficacy of different neuromodulatory techniques in
(MEPs; Figure 4).156 The most extensively used protocols in humans include promoting recovery of motor function after stroke.
repetitive transcranial magnetic stimulation (rTMS; Figure 4a), paired associative Fractional anisotropy of the internal capsule, a micro­
stimulation (PAS; Figure 4b) and transcranial direct current stimulation (tDCS; structural marker of damage to descending motor path­
Figure 4c). ways,89 has been demonstrated to differentiate patients
rTMS can increase excitability of corticospinal neurons directly (suprathreshold who respond better to an inhibitory cathodal tDCS
>5 Hz rTMS) or decrease excitability through cortical interneurons projecting stimulation protocol applied to the unaffected hemi­
to corticospinal cells (suprathreshold low-frequency rTMS).14,120 Intermittent sphere from patients who benefit more from excitatory
and continuous theta-burst stimulation (iTBS and cTBS) are subthreshold rTMS stimulation of the affected hemisphere.90 Patients with
paradigms (that is, the stimulator delivers an intensity below the one needed to
extensive damage to the corticospinal tract of the affected
evoke the MEP); iTBS increases and cTBS decreases corticospinal excitability.158 The
balance of long-term potentiation (LTP)-like plasticity in the affected hemisphere and
hemisphere responded poorly to inhibitory (cathodal)
the long-term depression (LTD)-like plasticity in the unaffected hemisphere evoked by stimulation of the unaffected hemisphere, whereas
TBS predicts motor recovery at 6 months after stroke.159 patients with smaller lesions responded well.
PAS induces LTP/LTD-like changes by pairing an electrical stimulus to a peripheral These findings are in line with previous publications
nerve with a time-locked TMS pulse to the contralateral primary motor cortex suggesting that interhemispheric competition would
(M1).160 Both intracortical and thalamocortical inhibitory and excitatory networks dominate in patients with more-limited damage in the
are thought to be involved in the alterations of synaptic strength.105,161–164 The affected hemisphere, but would be less relevant in patients
interstimulus interval (ISI) determines whether the modulation is excitatory with more-severe damage to the affected hemisphere.91–94
or inhibitory. Similarly, TMS–fMRI studies of functional connectivity
tDCS delivers a low-intensity constant current through the scalp to the brain, between the motor areas of the two hemispheres have
and often exerts polarity-specific modulation of corticospinal excitability.165 shown that in severely affected patients, the influence
tDCS can activate LTP-like and LTD-like processes166 and many neurotransmitter
of the PMd of the unaffected hemisphere on M1 of the
systems.167,168 Unlike TMS, tDCS does not trigger action potentials; instead, it is
thought to cause a small change (in the order of 1 mV) in the membrane potential
affected hemisphere is facilitatory, whereas it is inhibitory
of cortical neurons.169 in patients with minimal impairment 95 and in healthy
individuals.96,97 Finally, an fMRI study has demonstrated
Repetitive electrical stimulation (rES) of peripheral nerves or muscles could
increase the efficacy of rehabilitation after stroke; it is portable and does not that inhibition from the unaffected to the affected hemi­
require the patient’s active participation,170–175 and has been demonstrated to sphere is greater during paretic hand movements in
improve the motor performance of the paretic limb.176 relatively well-recovered patients than in patients who
show poorer recovery, consistent with the notion that the
interhemispheric competition model predicts recovery in
own to explain recovery in all patients, owing to vari­ patients with high functional reserve.98
ability in time elapsed from the stroke event, residual
functions, and lesion size and location. Towards more-successful clinical trials
Our proposal is to combine these models into a Repetitive activation of synapses results in strengthen­
new model—the bimodal balance–recovery model ing (long-term potentiation; LTP) or weakening (long-
(Figure 3)—by introducing a new parameter, structural term depression; LTD) of synaptic connections and
reserve, which describes the extent to which neural path­ efficacy.99–101 Although LTP and LTD have been mostly
ways and relays spared by the lesion contribute to recov­ studied in vitro, synaptic connections in the human brain
ery in an individual patient. The amount of structural can be activated with noninvasive brain s­ timulation
reserve determines whether interhemispheric imbalance (Box 2, Figure 4).102–105
dominates over vicariation: if the structural reserve is Several recent studies have addressed the topic of
high, the interhemispheric competition model can neuromodulation and stroke recovery, but the evi­
predict recovery better than vicariation model, which is dence supporting the use of NIBS in stroke rehabilita­
more useful in predicting recovery in patients with little tion remains inconsistent. In 2012, two meta-analyses
structural reserve. of clinical trials concluded that both rTMS and tDCS
Integrity of brain regions in both hemispheres is can enhance motor recovery after stroke; the effect
likely to influence structural reserve, but undoubtedly sizes were substantial (0.4–0.6) and no major adverse
the most important contributory factor is the remaining effects were observed.12,13 In contrast with those promis­
function of the motor areas and corticospinal tract in ing results, however, two systematic Cochrane reviews
the affected hemisphere. Preservation of these structures published in 2013 concluded that good-quality evidence
in the affected hemisphere correlates with functional for the efficacy of rTMS10 and tDCS11 to improve motor
recovery and involvement of the stroke hemisphere in function of patients with stroke is lacking. Below, we
movement control.87,88 Greater damage produces worse discuss the factors that are likely to contribute to the
deficits, and is accompanied by increased levels of activ­ inconsistent findings.
ity in the unaffec­ted hemisphere, which in most cases
will represent vicariation of function. Hence, the level of The need for patient stratification
lateralization of neural activity alone is not always able In the NIBS trials conducted to date, similar neuro­
to predict the response to rehabilitation.88 modulation was often delivered regardless of lesion site

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

a rTMS over the M1 of the affected hemisphere in patients



1–4 Hz Total time 20 min
with subcortical but not cortical stroke lesions.114 The
rTMS 1s 1s 1s
5–20 Hz Total time 6 min extent of the damage is also highly important. To the
+
best of our knowledge, apart from a single case,115 the lit­
erature is devoid of positive responses to NIBS in stroke
TMS Suprathreshold
patients with severe impairments. Finally, it should be
Subthreshold noted that the stroke phase (acute, subacute or chronic)
TMS can determine the brain state and the plastic changes that
5 Hz are ongoing or already achieved. Nevertheless, similar

cTBS Total time 40 s protocols, for instance, continuous theta burst stimula­
TBS 2s 8s
tion, have been used in the ­subacute113,116 and chronic107
iTBS Total time 190 s
160
ms
+ stages after stroke.
5 Hz 20 20
ms ms
50 ms Refining stimulation techniques and protocols
In most trials, the NIBS technique used is chosen on
b
the basis of ease of use, patient comfort, availability of the
TMS + MNS n=1 n=2 n=3 n = 90
stimulation equipment, and effects on cortico­spinal
ISI = 10 ms
Total time 45 min
– excitability, irrespective of the underlying mechanism
PAS of action. We argue, however, that the choice of NIBS
Total time 67.5 min + should depend on the predicted mechanism of recov­
TMS + MNS
ery in each individual patient. The ideal NIBS protocol
ISI = 25 ms ISI 20 s depends on the patient’s functional reserve (which deter­
mines whether or not the model of interhemispheric
rivalry is applicable), type of stroke (subcortical versus
c – – – – – – – – – – – – – cortical; or ischaemic versus haemorrhagic), and stroke
c-tDCS Total time 15–60 min stage (acute, subacute or chronic).
tDCS
a-tDCS Total time 15–60 min +
NIBS techniques also have high interindividual vari­
+ + + + + + + + + + + +
ability: in many cases, stimulation parameters chosen to
tDCS increase corticospinal excitability can have a inhibitory
effect,117–120 possibly because of the variable effects of the
protocol on the mixed populations of cortical neurons,120
which might depend on genetic factors.121 As discussed
above, stroke may increase the variation in response
to NIBS, particularly if additional pharmacological
­treatment has been introduced.122
Heterogeneity of the stimulation protocols, with
TMS tDCS MNS respect to the number of sessions delivered107,113,116 and
the timing of the concomitant physical therapy,109,110
– Protocol to measure inhibition + Protocol to measure facilitation
adds to the confusing complexity. Trials of tDCS in
stroke are hampered by these issues and show high inter­
Figure 4 | Noninvasive neuromodulatory techniques. a | Depending on the study variability regarding electrode placing, intensity
stimulation frequency, suprathreshold rTMS can increase (>5 Hz rTMS) or decrease
of the delivered current, session schedule, and type of
(suprathreshold <5 Hz rTMS) the excitability of corticospinal neurons (Box 2). TBS
protocols are subthreshold rTMS paradigms that, depending on the pattern stroke.110,123 Only a few investigations have linked stimu­
(intermittent versus continuous) of the stimulation, can either increase (iTBS) or lation parameters to a neurophysiological evaluation
decrease (cTBS) corticospinal excitability. b | In the PAS protocol, median nerve of the motor system and have addressed the effects of
stimulation is combined with TMS to the contralateral M1. c | tDCS can exert the intervention on both excitability–connectivity and
polarity-specific modulation of corticospinal excitability; in the M1, a‑tDCS increases ­clinical outcome.109,116
corticospinal excitability, whereas c‑tDCS decreases it. Abbreviations: a‑tDCS,
anodal tDCS; cTBS, continuous theta burst stimulation; c‑tDCS, cathodal tDCS;
Heterogeneity in outcome measures
ISI, interstimulus intervals; iTBS, intermittent theta burst stimulation; M1, primary
Outcome measures vary widely between studies, ranging
motor cortex; PAS, paired associative stimulation; rTMS, repetitive TMS; TBS, theta
burst stimulation; tDCS, transcranial direct current stimulation; TMS, transcranial from measures of dexterity (Jebsen–Taylor Test,109 Action
magnetic stimulation. Research Arm Test,90,114 Purdue Pegboard Test,124 Box
and Block Test 125) to measures of strength (handgrip,126
pinchforce106), reaction time,127 and disability or stroke
or stroke aetiology. Unfortunately, this drawback has not severity (Barthel ,128 Fugl–Meyer,129 National Institutes of
been overcome by the most recent trials. Patients with Health Stroke Scale125) Meta-analyses equate the hetero­
cortical and subcortical stroke,106–111 or ischaemic and geneity of the outcome measures in different trials and
haemorrhagic stroke,109–111 are often merged into the same express the effects of the interventions as z‑scores, but
group, even though such factors can influence recovery: the different outcome measures make comparison of the
one study has described a positive response to facilitatory outcomes between studies very challenging.

NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION | 7


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Our conclusion is that noninvasive neuromodulation cortical excitability and time-resolved effective con­
is not yet ready to be implemented in stroke rehabilita­ nectivity.141–143 TMS–EEG could, therefore, provide new
tion in the current clinical practice, for several reasons. insights into the interaction between brain areas during
First, the concept of interhemispheric imbalance, which motor function, 142,144,145 and elucidate how the areas
has been employed as the rationale in most of the studies, involved in motor control react to acute damage, such
is likely to be too simplistic to be successfully applied as stroke.
in each individual patient. Second, very few trials have
used exactly the same protocols, meaning that estimat­ Conclusions
ing the real effectiveness and reproducibility is often The interhemispheric competition and vicariation
impossible. Third, the use of different outcome measures models of motor recovery after stroke lead to oppos­
means that studies are not readily comparable. Last, the ing conclusions about the best NIBS interventions. We
number of negative ­standardized clinical trials cannot suggest introduction of a weighting term, ‘structural
be overlooked.90,125,130 reserve’, which describes the remaining functional
motor output, and propose a unifying bimodal balance–­
Individual multimodal diagnostic approach recovery model, which determines the amount of recov­
The bimodal balance–recovery model proposed in ery limited by interhemispheric competition and the
this Review (Figure 3) implies that we cannot hope to recovery supported by vicariation. This threshold varies
improve the function of the affected hand in patients from patient to patient and enables us to determine how
with stroke through a one-size-fits-all-strategy,90 and best to apply NIBS interventions on an individual basis.
that multiple factors, such as the severity of clinical For such patient-tailored treatment, development of
impairment, determine the strategies and success of standardized methods of meas­uring structural reserve,
­neuromodulatory treatment.131 such as clinical, anatomical and functional measures
The idea of a patient-tailored approach is not new: a of motor connectivity, is important. Future studies will
similar idea has been proposed by Pascual-Leone and refine the bimodal balance–recovery model and test
colleagues132,133 and Byblow and Stinear.47 An individu­ whether the efficacy of individualized NIBS therapy is
alized, multimodal pre-therapy diagnosis is envisaged, superior to the current methods.
involving the clinical history of the patient, time elapsed Our analysis has a number of unavoidable limita­
after the stroke, topography of the lesion, and type and tions. Besides the factors included in our model, several
severity of functional impairment. Stinear and Byblow additional factors will contribute to the efficacy of
proposed a simple diagnostic algorithm to predict func­ NIBS, including the timeliness of the intervention, age,
tional recovery at 6 months on the basis of measures sex, comorbidities, concurrent pharmacological treat­
collected during the first 7–10 days after stroke.88 We ments, the ischaemic versus haemorrhagic nature of the
propose that a similar algorithm should be developed to lesion, and whether the stroke affects the dominant or
facilitate an individualized approach to NIBS. the nondominant hemisphere. However, we believe that
The algorithm of Stinear and Byblow incorporates in the future, these factors can be successfully integrated
clinical strength measures, response to TMS, and frac­ into the proposed bimodal balance–recovery model
tional anisotropy measures on MRI. The proposed NIBS and the ­a ssociated concept of individually tailored
algorithm could rely on similar measures. However, NIBS treatment.
given that functional brain connectivity might also Finally, there is no comprehensive agreement on the
be relevant, other methods could prove valuable. For optimal combination of NIBS parameters, the duration
example, EEG could provide a useful and cheap func­ of the NIBS treatment, and clinical outcome measures
tional way to measure the state of remaining brain areas (force versus dexterity; time of follow-up) in evaluating
and aid the selection of the NIBS protocol. EEG can be the efficacy of the therapy. We envision that progress
used to explore resting-­state activity and connectivity in the near future will provide in-depth knowledge of
of the brain and to provide immediate information on stroke rehab­i litation, and that individualized neuro­
the functional integrity of cortical tissue. Delta waves modulation will result in notable enhancements in
increase bilaterally in acute stroke, but are raised particu­ rehabilitation success.
larly over the affected hemisphere, where they are related
to the acute clinical impairment.134 Symmetry of spec­
Review criteria
tral power between the two hemispheres is associated
with mild neurological deficits,135 and a sudden emer­ We searched the literature for recent trials and meta-
analyses assessing the efficacy of noninvasive brain
gence of delta waves in the unaffected hemisphere136–138
stimulation. Rather than adding to the growing body of
or of an asymmetry across the hemispheres139 can reveal existing systemic reviews of clinical trials, our intention
acute worsening with a poor prognosis. In the chronic is to introduce a scientific rationale for improving the
phase, the reduction of the beta band (12.5–30.0 Hz) outcome of those trials. We do not claim to have read all
oscillatory activity in the motor cortex correlates with relevant literature; however, we have tried to incorporate
motor impairment.140 the whole range of present viewpoints into a coherent
Further information on connectivity might be and logical structure. Thus, our scope was extremely
obtained with TMS–EEG co-recording, which offers broad and relied extensively on the long experience of
the authors in this field.
the possibility for noninvasive probing of the brain’s

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

1. Kolominsky-Rabas, P. L., Weber, M., Gefeller, O., 19. Somjen, G. G. (Ed.) Ions in the Brain: Normal not dexterity. Clin. Neurophysiol. 113,
Neundoerfer, B. & Heuschmann, P. U. Function, Seizures, and Stroke: Normal Function, 2025–2029 (2002).
Epidemiology of ischemic stroke subtypes Seizures, and Stroke (Oxford University Press, 38. Turton, A., Wroe, S., Trepte, N., Fraser, C. &
according to TOAST criteria: incidence, 2004). Lemon, R. Contralateral and ipsilateral EMG
recurrence, and long-term survival in ischemic 20. Gerard, R. The response of nerve to oxygen lack. responses to transcranial magnetic stimulation
stroke subtypes: a population-based study. Am. J. Physiol. 92, 498–541 (1930). during recovery of arm and hand function after
Stroke 32, 2735–2740 (2001). 21. Zhu, P. & Krnjević, K. Anoxia selectively stroke. Electroencephalogr. Clin. Neurophysiol.
2. Roger, V. L. et al. Heart disease and stroke depresses excitatory synaptic transmission in 101, 316–328 (1996).
statistics. A report from the American Heart hippocampal slices. Neurosci. Lett. 166, 27–30 39. Catano, A., Houa, M. & Noël, P. Magnetic
Association. Circulation 123, e18–e209 (2011). (1994). transcranial stimulation: dissociation of
3. Veerbeek, J. M., Kwakkel, G., van Wegen, E. E., 22. Khazipov, R., Bregestovski, P. & Ben-Ari, Y. excitatory and inhibitory mechanisms in acute
Ket, J. C. & Heymans, M. W. Early prediction of Hippocampal inhibitory interneurons are strokes. Electroencephalogr. Clin. Neurophysiol.
outcome of activities of daily living after stroke: functionally disconnected from excitatory inputs 105, 29–36 (1997).
a systematic review. Stroke 42, 1482–1488 by anoxia. J. Neurophysiol. 70, 2251–2259 40. Palliyath, S. Role of central conduction time and
(2011). (1993). motor evoked response amplitude in predicting
4. Lansberg, M. G., Bluhmki, E. & Thijs, V. N. 23. Murphy, T. H. & Corbett, D. Plasticity during stroke outcome. Electromyogr. Clin. Neurophys.
Efficacy and safety of tissue plasminogen stroke recovery: from synapse to behaviour. 40, 315–320 (2000).
activator 3 to 4.5 hours after acute ischemic Nat. Rev. Neurosci. 10, 861–872 (2009). 41. Rapisarda, G., Bastings, E., de Noordhout, A. M.,
stroke: a meta-analysis. Stroke 40, 2438–2441 24. Clarkson, A. N., Huang, B. S., MacIsaac, S. E., Pennisi, G. & Delwaide, P. Can motor recovery in
(2009). Mody, I. & Carmichael, S. T. Reducing excessive stroke patients be predicted by early transcranial
5. Maulden, S. A., Gassaway, J., Horn, S. D., GABA-mediated tonic inhibition promotes magnetic stimulation? Stroke 27, 2191–2196
Smout, R. J. & DeJong, G. Timing of initiation functional recovery after stroke. Nature 468, (1996).
of rehabilitation after stroke. Arch. Phys. Med. 305–309 (2010). 42. Hendricks, H. T., Zwarts, M. J., Plat, E. F. &
Rehabil. 86, S34–S40 (2005). 25. Carmichael, S. T. Brain excitability in stroke: the van Limbeek, J. Systematic review for the early
6. Dobkin, B. H. Clinical practice. Rehabilitation yin and yang of stroke progression. Arch. Neurol. prediction of motor and functional outcome after
after stroke. N. Engl. J. Med. 352, 1677–1684 69, 161 (2012). stroke by using motor-evoked potentials. Arch.
(2005). 26. Buchkremer-Ratzmann, I., August, M., Phys. Med. Rehabil. 83, 1303–1308 (2002).
7. Wagner, T. H. et al. An economic analysis of Hagemann, G. & Witte, O. W. Electrophysiological 43. Byrnes, M. L., Thickbroom, G. W., Phillips, B. A.
robot-assisted therapy for long-term upper-limb transcortical diaschisis after cortical & Mastaglia, F. L. Long-term changes in motor
impairment after stroke. Stroke 42, 2630–2632 photothrombosis in rat brain. Stroke 27, cortical organisation after recovery from
(2011). 1105–1111 (1996). subcortical stroke. Brain Res. 889, 278–287
8. Lo, A. C. et al. Robot-assisted therapy for long- 27. Schiene, K. et al. Neuronal hyperexcitability and (2001).
term upper-limb impairment after stroke. N. Engl. reduction of GABAA-receptor expression in the 44. Pennisi, G. et al. Transcranial magnetic
J. Med. 362, 1772–1783 (2010). surround of cerebral photothrombosis. J. Cereb. stimulation after pure motor stroke. Clin.
9. Klamroth-Marganska, V. et al. Three-dimensional, Blood Flow Metab. 16, 906–914 (1996). Neurophysiol. 113, 1536–1543 (2002).
task-specific robot therapy of the arm after 28. Bruehl, C., Neumann-Haefelin, T. & Witte, O. 45. Pennisi, G. et al. Absence of response to early
stroke: a multicentre, parallel-group randomised Enhancement of whole cell calcium currents transcranial magnetic stimulation in ischemic
trial. Lancet Neurol. 13, 159– 166 (2014). following transient MCAO. Brain Res. 884, stroke patients: prognostic value for hand motor
10. Hao, Z., Wang, D., Zeng, Y. & Liu, M. Repetitive 129–138 (2000). recovery. Stroke 30, 2666–2670 (1999).
transcranial magnetic stimulation for improving 29. Bahn, M. M., Oser, A. B. & Cross, D. T. CT 46. Delvaux, V. et al. Post-stroke reorganization of
function after stroke. Cochrane Database of and MRI of stroke. J. Magn. Reson. Imaging 6, hand motor area: a 1‑year prospective follow-up
Systematic Reviews, Issue 5. Art. No.: 833–845 (1996). with focal transcranial magnetic stimulation.
Cd008862. http://dx.doi.org/10.1002/ 30. Cicinelli, P., Traversa, R. & Rossini, P. Post-stroke Clin. Neurophysiol. 114, 1217–1225 (2003).
14651858.CD008862.pub2. reorganization of brain motor output to the hand: 47. Stinear, C. M., Barber, P. A., Petoe, M., Anwar, S.
11. Elsner, B., Kugler, J., Pohl, M. & Mehrholz, J. a 2–4 month follow-up with focal magnetic & Byblow, W. D. The PREP algorithm predicts
Transcranial direct current stimulation (tDCS) transcranial stimulation. Electroencephalogr. potential for upper limb recovery after stroke.
for improving function and activities of daily Clin. Neurophysiol. 105, 438–450 (1997). Brain 135, 2527–2535 (2012).
living in patients after stroke. Cochrane 31. Traversa, R., Cicinelli, P., Bassi, A., Rossini, P. M. 48. Liepert, J., Storch, P., Fritsch, A. & Weiller, C.
Database of Systematic Reviews, Issue 11. & Bernardi, G. Mapping of motor cortical Motor cortex disinhibition in acute stroke.
Art. No.: Cd009645. http://dx.doi.org/ reorganization after stroke. A brain stimulation Clin. Neurophysiol. 111, 671–676 (2000).
10.1002/14651858.CD009645.pub2. study with focal magnetic pulses. Stroke 28, 49. Cicinelli, P. et al. Interhemispheric asymmetries of
12. Adeyemo, B. O., Simis, M., Macea, D. D. & 110–117 (1997). motor cortex excitability in the postacute stroke
Fregni, F. Systematic review of parameters of 32. Traversa, R., Cicinelli, P., Pasqualetti, P., Filippi, M. stage: a paired-pulse transcranial magnetic
stimulation, clinical trial design characteristics, & Rossini, P. M. Follow-up of interhemispheric stimulation study. Stroke 34, 2653–2658 (2003).
and motor outcomes in non-invasive brain differences of motor evoked potentials from the 50. Swayne, O. B., Rothwell, J. C., Ward, N. S.
stimulation in stroke. Front. Psychiatry 3, 88 ‘affected’and ‘unaffected’ hemispheres in & Greenwood, R. J. Stages of motor output
(2012). human stroke. Brain Res. 803, 1–8 (1998). reorganization after hemispheric stroke
13. Hsu, W. Y., Cheng, C. H., Liao, K. K., Lee, I. H. 33. Traversa, R. et al. Neurophysiological follow-up suggested by longitudinal studies of cortical
& Lin, Y. Y. Effects of repetitive transcranial of motor cortical output in stroke patients. Clin. physiology. Cereb. Cortex 18, 1909–1922 (2008).
magnetic stimulation on motor functions in Neurophysiol. 111, 1695–1703 (2000). 51. Bütefisch, C. M., Netz, J., Wessling, M.,
patients with stroke: a meta-analysis. Stroke 43, 34. Catano, A., Houa, M., Caroyer, J., Ducarne, H. Seitz, R. J. & Hömberg, V. Remote changes
1849–1857 (2012). & Noel, P. Magnetic transcranial stimulation in in cortical excitability after stroke. Brain 126,
14. Di Lazzaro, V. et al. I‑wave origin and modulation. acute stroke: early excitation threshold and 470–481 (2003).
Brain Stimul. 5, 512–525 (2012). functional prognosis. Electroencephalogr. Clin. 52. Di Lazzaro, V. et al. The level of cortical afferent
15. Attwell, D. & Laughlin, S. B. An energy budget for Neurophysiol. 101, 233–239 (1996). inhibition in acute stroke correlates with long-
signaling in the grey matter of the brain. J. Cereb. 35. Heald, A., Bates, D., Cartlidge, N., French, J. & term functional recovery in humans. Stroke 43,
Blood Flow Metab. 21, 1133–1145 (2001). Miller, S. Longitudinal study of central motor 250–252 (2012).
16. Astrup, J., Siesjo, B. K. & Symon, L. Thresholds conduction time following stroke. 2. Central 53. Foltys, H. et al. Motor representation in patients
in cerebral ischemia: the ischemic penumbra. motor conduction measured within 72 h after rapidly recovering after stroke: a functional
Stroke 12, 723–725 (1981). stroke as a predictor of functional outcome at magnetic resonance imaging and transcranial
17. Symon, L. The relationship between CBF, evoked 12 months. Brain 116, 1371–1385 (1993). magnetic stimulation study. Clin. Neurophysiol.
potentials and the clinical features in cerebral 36. Manganotti, P. et al. Motor disinhibition in 114, 2404–2415 (2003).
ischaemia. Acta Neurol. Scand. Suppl. 78, affected and unaffected hemisphere in the early 54. Shimizu, T. et al. Motor cortical disinhibition
175–190 (1980). period of recovery after stroke. Clin. Neurophysiol. in the unaffected hemisphere after unilateral
18. Krnjević, K. & Xu, Y. Mechanisms underlying 113, 936–943 (2002). cortical stroke. Brain 125, 1896–1907 (2002).
anoxic hyperpolarization of hippocampal 37. Thickbroom, G. W., Byrnes, M. L., Archer, S. A. & 55. Fridman, E. A. et al. Reorganization of the human
neurons. Can. J. Physiol. Pharmacol. 68, Mastaglia, F. L. Motor outcome after subcortical ipsilesional premotor cortex after stroke. Brain
1609–1613 (1990). stroke: MEPs correlate with hand strength but 127, 747–758 (2004).

NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION | 9


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

56. Liepert, J., Hamzei, F. & Weiller, C. Motor cortex 74. Boudrias, M.‑H., McPherson, R. L., Frost, S. B. & learning and recovery from stroke. Front. Syst.
disinhibition of the unaffected hemisphere after Cheney, P. D. Output properties and organization Neurosci. 4, 146 (2010).
acute stroke. Muscle Nerve 23, 1761–1763 of the forelimb representation of motor areas on 95. Bestmann, S. et al. The role of contralesional
(2000). the lateral aspect of the hemisphere in rhesus dorsal premotor cortex after stroke as studied
57. Murase, N., Duque, J., Mazzocchio, R. & macaques. Cerebr. Cortex 20, 169–186 (2010). with concurrent TMS–fMRI. J. Neurosci. 30,
Cohen, L. G. Influence of interhemispheric 75. Baker, S. N. The primate reticulospinal tract, 11926–11937 (2010).
interactions on motor function in chronic stroke. hand function and functional recovery. J. Physiol. 96. Mochizuki, H., Huang, Y.‑Z. & Rothwell, J. C.
Ann. Neurol. 55, 400–409 (2004). 589, 5603–5612 (2011). Interhemispheric interaction between human
58. Jacobs, K. M. & Donoghue, J. P. Reshaping 76. Eisner-Janowicz, I. et al. Early and late changes dorsal premotor and contralateral primary motor
the cortical motor map by unmasking latent in the distal forelimb representation of the cortex. J. Physiol. 561, 331–338 (2004).
intracortical connections. Science 251, supplementary motor area after injury to frontal 97. Koch, G. et al. Time course of functional
944–947 (1991). motor areas in the squirrel monkey. connectivity between dorsal premotor and
59. Pineiro, R., Pendlebury, S., Johansen-Berg, H. & J. Neurophysiol. 100, 1498–1512 (2008). contralateral motor cortex during movement
Matthews, P. Functional MRI detects posterior 77. Brus-Ramer, M., Carmel, J. B., Chakrabarty, S. selection. J. Neurosci. 26, 7452–7459 (2006).
shifts in primary sensorimotor cortex activation & Martin, J. H. Electrical stimulation of spared 98. Lomarev, M., Kim, D., Richardson, S. P., Voller, B.
after stroke: evidence of local adaptive corticospinal axons augments connections with & Hallett, M. Safety study of high-frequency
reorganization? Stroke 32, 1134–1139 (2001). ipsilateral spinal motor circuits after injury. transcranial magnetic stimulation in patients
60. Rossini, P. et al. Hand motor cortical area J. Neurosci. 27, 13793–13801 (2007). with chronic stroke. Clin. Neurophysiol. 118,
reorganization in stroke: a study with fMRI, MEG 78. Verstynen, T., Diedrichsen, J., Albert, N., 2072–2075 (2007).
and TCS maps. Neuroreport 9, 2141–2146 Aparicio, P. & Ivry, R. B. Ipsilateral motor cortex 99. Magee, J. C. & Johnston, D. A synaptically
(1998). activity during unimanual hand movements controlled, associative signal for Hebbian
61. Calautti, C., Leroy, F., Guincestre, J.‑Y. & relates to task complexity. J. Neurophysiol. 93, plasticity in hippocampal neurons. Science 275,
Baron, J.‑C. Displacement of primary 1209–1222 (2005). 209–213 (1997).
sensorimotor cortex activation after subcortical 79. Hummel, F., Kirsammer, R. & Gerloff, C. 100. Bi, G. & Poo, M. Synaptic modification by
stroke: a longitudinal PET study with clinical Ipsilateral cortical activation during finger correlated activity: Hebb’s postulate revisited.
correlation. Neuroimage 19, 1650–1654 sequences of increasing complexity: Annu. Rev. Neurosci. 24, 139–166 (2001).
(2003). representation of movement difficulty or memory 101. Bi, G. Q. & Poo, M. M. Synaptic modifications
62. Seitz, R. J. et al. Role of the premotor cortex in load? Clin. Neurophys. 114, 605–613 (2003). in cultured hippocampal neurons: dependence
recovery from middle cerebral artery infarction. 80. Rose, D. & Winstein, C. The co-ordination of on spike timing, synaptic strength, and
Arch. Neurol. 55, 1081–1088 (1998). bimanual rapid aiming movements following postsynaptic cell type. J. Neurosci. 18,
63. Chollet, F. et al. The functional anatomy of motor stroke. Clin. Rehabil. 19, 452–462 (2005). 10464–10472 (1998).
recovery after stroke in humans: a study with 81. Lewis, G. N. & Byblow, W. D. Bimanual 102. Hoogendam, J. M., Ramakers, G. M. &
positron emission tomography. Ann. Neurol. 29, coordination dynamics in poststroke Di Lazzaro, V. Physiology of repetitive
63–71 (1991). hemiparetics. J. Mot. Behav. 36, 174–188 (2004). transcranial magnetic stimulation of the human
64. Weiller, C., Chollet, F., Friston, K. J., Wise, R. J. 82. Grefkes, C. et al. Cortical connectivity after brain. Brain Stimul. 3, 95–118 (2010).
& Frackowiak, R. S. Functional reorganization subcortical stroke assessed with functional 103. Huang, Y.‑Z., Rothwell, J. C., Chen, R.‑S., Lu, C.‑S.
of the brain in recovery from striatocapsular magnetic resonance imaging. Ann. Neurol. 63, & Chuang, W.‑L. The theoretical model of theta
infarction in man. Ann. Neurol. 31, 463–472 236–246 (2008). burst form of repetitive transcranial magnetic
(1992). 83. Finger, S. In Handbook of Clinical Neurology stimulation. Clin. Neurophysiol. 122, 1011–1018
65. Weiller, C., Ramsay, S., Wise, R., Friston, K. & Vol. 95 Ch. 51 (eds Aminoff, J. A. et al.) 833–841 (2011).
Frackowiak, R. Individual patterns of functional (Elsevier, 2009). 104. Wolters, A. et al. A temporally asymmetric
reorganization in the human cerebral cortex after 84. Duque, J. et al. Transcallosal inhibition in chronic Hebbian rule governing plasticity in the human
capsular infarction. Ann. Neurol. 33, 181–189 subcortical stroke. Neuroimage 28, 940–946 motor cortex. J. Neurophysiol. 89, 2339–2345
(1993). (2005). (2003).
66. Johansen-Berg, H. et al. The role of ipsilateral 85. Werhahn, K. J., Mortensen, J., Van Boven, R. W., 105. Stefan, K., Kunesch, E., Cohen, L. G.,
premotor cortex in hand movement after stroke. Zeuner, K. E. & Cohen, L. G. Enhanced tactile Benecke, R. & Classen, J. Induction of plasticity
Proc. Natl Acad. Sci. 99, 14518–14523 (2002). spatial acuity and cortical processing during in the human motor cortex by paired associative
67. Lotze, M. et al. The role of multiple acute hand deafferentation. Nat. Neurosci. 5, stimulation. Brain 123, 572–584 (2000).
contralesional motor areas for complex hand 936–938 (2002). 106. Conforto, A. B. et al. Transcranial magnetic
movements after internal capsular lesion. 86. Floel, A. et al. Influence of somatosensory input stimulation in mild to severe hemiparesis early
J. Neurosci. 26, 6096–6102 (2006). on motor function in patients with chronic after stroke: a proof of principle and novel
68. Jang, S. H. A review of the ipsilateral motor stroke. Ann. Neurol. 56, 206–212 (2004). approach to improve motor function. J. Neurol.
pathway as a recovery mechanism in patients 87. Ward, N. S. & Cohen, L. G. Mechanisms 259, 1399–1405 (2012).
with stroke. NeuroRehabilitation 24, 315–320 underlying recovery of motor function after 107. Talelli, P. et al. Theta burst stimulation in
(2009). stroke. Arch. Neurol. 61, 1844–1848 (2004). the rehabilitation of the upper limb:
69. Ago, T. et al. Deterioration of pre-existing 88. Stinear, C. M. et al. Functional potential in a semirandomized, placebo-controlled trial in
hemiparesis brought about by subsequent chronic stroke patients depends on corticospinal chronic stroke patients. Neurorehabil. Neural
ipsilateral lacunar infarction. J. Neurol. tract integrity. Brain 130, 170–180 (2007). Repair 26, 976–987 (2012).
Neurosurg. Psychiatry 74, 1152–1153 (2003). 89. Lindenberg, R. et al. Structural integrity of 108. Lindenberg, R., Zhu, L. L. & Schlaug, G.
70. Song, Y.‑M., Lee, J.‑Y., Park, J.‑M., Yoon, B.‑W. & corticospinal motor fibers predicts motor Combined central and peripheral stimulation
Roh, J.‑K. Ipsilateral hemiparesis caused by a impairment in chronic stroke. Neurology 74, to facilitate motor recovery after stroke: the
corona radiata infarct after a previous stroke on 280–287 (2010). effect of number of sessions on outcome.
the opposite side. Arch. Neurol. 62, 809–811 90. Bradnam, L. V., Stinear, C. M., Barber, P. A. & Neurorehabil. Neural Repair 26, 479–483
(2005). Byblow, W. D. Contralesional hemisphere control (2012).
71. Yamamoto, S., Takasawa, M., Kajiyama, K., of the proximal paretic upper limb following 109. Avenanti, A., Coccia, M., Ladavas, E.,
Baron, J.‑C. & Yamaguchi, T. Deterioration of stroke. Cerebr. Cortex 22, 2662–2671 (2012). Provinciali, L. & Ceravolo, M. Low-frequency
hemiparesis after recurrent stroke in the 91. Werhahn, K. J., Conforto, A. B., Kadom, N., rTMS promotes use-dependent motor plasticity
unaffected hemisphere: three further cases with Hallett, M. & Cohen, L. G. Contribution of the in chronic stroke: a randomized trial. Neurology
possible interpretation. Cerebrovasc. Dis. 23, ipsilateral motor cortex to recovery after chronic 78, 256–264 (2012).
35–39 (2006). stroke. Ann. Neurol. 54, 464–472 (2003). 110. Kakuda, W. et al. A multi-center study on low-
72. Biernaskie, J., Szymanska, A., Windle, V. & 92. Seghier, M. L. Laterality index in functional MRI: frequency rTMS combined with intensive
Corbett, D. Bi-hemispheric contribution to methodological issues. Magn. Res. Imaging 26, occupational therapy for upper limb hemiparesis
functional motor recovery of the affected 594–601 (2008). in post-stroke patients. J. Neuroeng. Rehabil. 9,
forelimb following focal ischemic brain injury 93. Johansen-Berg, H. Functional imaging of stroke 4 (2012).
in rats. Eur. J. Neurosci. 21, 989–999 (2005). recovery: what have we learnt and where do we 111. Chang, W. H. et al. rTMS with motor training
73. Lemon, R. N. Descending pathways in motor go from here? Int. J. Stroke 2, 7–16 (2007). modulates cortico-basal ganglia-thalamocortical
control. Annu. Rev. Neurosci. 31, 195–218 94. Johansen-Berg, H., Scholz, J. & Stagg, C. J. circuits in stroke patients. Restor. Neurol.
(2008). Relevance of structural brain connectivity to Neurosci. 30, 179–189 (2012).

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

112. Seniów, J. et al. Transcranial magnetic 127. Hummel, F. C. & Cohen, L. G. Non-invasive 144. Nikulin, V. V., Kicic, D., Kahkonen, S.
stimulation combined with physiotherapy in brain stimulation: a new strategy to improve & Ilmoniemi, R. J. Modulation of
rehabilitation of poststroke hemiparesis: neurorehabilitation after stroke? Lancet Neurol. electroencephalographic responses to
a randomized, double-blind, placebo-controlled 5, 708–712 (2006). transcranial magnetic stimulation: evidence
study. Neurorehabil. Neural Repair 26, 128. Khedr, E. M., Ahmed, M. A., Fathy, N. & for changes in cortical excitability related to
1072–1079 (2012). Rothwell, J. C. Therapeutic trial of repetitive movement. Eur. J. Neurosci. 18, 1206–1212
113. Cazzoli, D. et al. Theta burst stimulation reduces transcranial magnetic stimulation after acute (2003).
disability during the activities of daily living in ischemic stroke. Neurology 65, 466–468 (2005). 145. Ferreri, F. et al. Human brain connectivity during
spatial neglect. Brain 135, 3426–3439 (2012). 129. Nair, D. G., Renga, V., Lindenberg, R., Zhu, L. single and paired pulse transcranial magnetic
114. Ameli, M. et al. Differential effects of high- & Schlaug, G. Optimizing recovery potential stimulation. Neuroimage 54, 90–102 (2011).
frequency repetitive transcranial magnetic through simultaneous occupational therapy and 146. Kujirai, T. et al. Corticocortical inhibition in
stimulation over ipsilesional primary motor non-invasive brain-stimulation using tDCS. human motor cortex. J. Physiol. 471, 501–519
cortex in cortical and subcortical middle Restor. Neurol. Neurosci. 29, 411–420 (2011). (1993).
cerebral artery stroke. Ann. Neurol. 66, 130. Grefkes, C. & Fink, G. R. Disruption of motor 147. Di Lazzaro, V. et al. Direct demonstration of
298–309 (2009). network connectivity post-stroke and its the effect of lorazepam on the excitability of the
115. Boggio, P. S. et al. Hand function improvement noninvasive neuromodulation. Curr. Opin. Neurol. human motor cortex. Clin. Neurophys. 111,
with low-frequency repetitive transcranial 25, 670–675 (2012). 794–799 (2000).
magnetic stimulation of the unaffected 131. Schlaug, G., Renga, V. & Nair, D. Transcranial 148. Calancie, B., Nordin, M., Wallin, U. &
hemisphere in a severe case of stroke. direct current stimulation in stroke recovery. Hagbarth, K.‑E. Motor-unit responses in human
Am. J. Phys. Med. Rehabil. 85, 927–930 Arch. Neurol. 65, 1571–1576 (2008). wrist flexor and extensor muscles to transcranial
(2006). 132. Hummel, F. C. et al. Controversy: noninvasive cortical stimuli. J. Neurophysiol. 58, 1168–1185
116. Koch, G. et al. Theta-burst stimulation of the left and invasive cortical stimulation show efficacy in (1987).
hemisphere accelerates recovery of hemispatial treating stroke patients. Brain Stim. 1, 370–382 149. Valls-Solé, J., Pascual-Leone, A.,
neglect. Neurology 78, 24–30 (2012). (2008). Wassermann, E. M. & Hallett, M. Human motor
117. Di Lazzaro, V. et al. Modulation of motor cortex 133. Plow, E. B., Carey, J. R., Nudo, R. J., & Pascual- evoked responses to paired transcranial
neuronal networks by rTMS: comparison of local Leone, A. Invasive cortical stimulation to magnetic stimuli. Electroencephalogr. Clin.
and remote effects of six different protocols of promote recovery of function after stroke: Neurophysiol. 85, 355–364 (1992).
stimulation. J. Neurophysiol. 105, 2150–2156 a critical appraisal. Stroke 40, 1926–1931 150. Di Lazzaro, V. et al. Segregating two inhibitory
(2011). (2009). circuits in human motor cortex at the level of
118. Datta, A., Truong, D., Minhas, P., Parra, L. C. 134. Assenza, G. et al. Neuronal functionality GABAA receptor subtypes: a TMS study. Clin.
& Bikson, M. Inter-individual variation during assessed by magnetoencephalography is Neurophys. 118, 2207–2214 (2007).
transcranial direct current stimulation and related to oxidative stress system in acute 151. Ziemann, U., Rothwell, J. C. & Ridding, M. C.
normalization of dose using MRI-derived ischemic stroke. Neuroimage 44, 1267–1273 Interaction between intracortical inhibition and
computational models. Front. Psychiatry 3, 91 (2009). facilitation in human motor cortex. J. Physiol.
(2012). 135. Sheorajpanday, R. V., Nagels, G., Weeren, A. J., 496, 873–881 (1996).
119. Maeda, F., Keenan, J. P., Tormos, J. M., Topka, H. van Putten, M. J. & De Deyn, P. P. Quantitative 152. Di Lazzaro, V. et al. Origin of facilitation of
& Pascual-Leone, A. Interindividual variability of EEG in ischemic stroke: correlation with motor-evoked potentials after paired magnetic
the modulatory effects of repetitive transcranial functional status after 6 months. Clin. stimulation: direct recording of epidural activity
magnetic stimulation on cortical excitability. Exp. Neurophysiol. 122, 874–883 (2011). in conscious humans. J. Neurophysiol. 96,
Brain Res. 133, 425–430 (2000). 136. Finnigan, S., Rose, S. E. & Chalk, J. B. 1765–1771 (2006).
120. Hamada, M., Murase, N., Hasan, A., Contralateral hemisphere delta EEG in acute 153. Siebner, H. R. et al. Consensus paper:
Balaratnam, M. & Rothwell, J. C. The role of stroke precedes worsening of symptoms and combining transcranial stimulation with
interneuron networks in driving human motor death. Clin. Neurophysiol. 119, 1690–1694 neuroimaging. Brain Stimul. 2, 58–80 (2009).
cortical plasticity. Cereb. Cortex 23, 1593–1605 (2008). 154. Ziemann, U. Transcranial magnetic stimulation
(2013). 137. Tecchio, F. et al. Outcome prediction in acute at the interface with other techniques:
121. Cheeran, B. et al. A common polymorphism monohemispheric stroke via a powerful tool for studying the human cortex.
in the brain-derived neurotrophic factor gene magnetoencephalography. J. Neurol. 254, Neuroscientist 17, 368–381 (2011).
(BDNF) modulates human cortical plasticity 296–305 (2007). 155. Ilmoniemi, R. J. & Kicic, D. Methodology for
and the response to rTMS. J. Physiol. 586, 138. Assenza, G., Zappasodi, F., Pasqualetti, P., combined TMS and EEG. Brain Topogr. 22,
5717–5725 (2008). Vernieri, F. & Tecchio, F. A contralesional EEG 233–248 (2010).
122. Nitsche, M. A., Müller-Dahlhaus, F., Paulus, W. power increase mediated by interhemispheric 156. Ziemann, U. et al. Consensus: motor cortex
& Ziemann, U. The pharmacology of disconnection provides negative prognosis in plasticity protocols. Brain Stim. 1, 164–182
neuroplasticity induced by non-invasive brain acute stroke. Restor. Neurol. Neurosci. 31, (2008).
stimulation: building models for the clinical 177–188 (2013). 157. Thickbroom, G. W. Transcranial magnetic
use of CNS active drugs. J. Physiol. 590, 139. Finnigan, S. & van Putten, M. J. EEG in stimulation and synaptic plasticity: experimental
4641–4662 (2012). ischaemic stroke: quantitative EEG can uniquely framework and human models. Exp. Brain Res.
123. Wu, D. et al. Effects on decreasing upper-limb inform (sub‑) acute prognoses and clinical 180, 583–593 (2007).
poststroke muscle tone using transcranial direct management. Clin. Neurophysiol. 124, 10–9 158. Huang, Y. Z., Edwards, M. J., Rounis, E.,
current stimulation: a randomized sham- (2012). Bhatia, K. P. & Rothwell, J. C. Theta burst
controlled study. Arch. Phys. Med. Rehabil. 94, 140. Graziadio, S., Tomasevic, L., Assenza, G., stimulation of the human motor cortex. Neuron
1–8 (2013). Tecchio, F. & Eyre, J. The myth of the ‘unaffected’ 45, 201–206 (2005).
124. Khedr, E. M., Etraby, A. E., Hemeda, M., side after unilateral stroke: is reorganisation of 159. Di Lazzaro, V. et al. Motor cortex plasticity
Nasef, A. M. & Razek, A. A. Long-term effect the non-infarcted corticospinal system to re- predicts recovery in acute stroke. Cereb. Cortex
of repetitive transcranial magnetic stimulation establish balance the price for recovery? Exp. 20, 1523–1528 (2010).
on motor function recovery after acute Neurol. 238, 168–175 (2012). 160. Müller-Dahlhaus, F., Ziemann, U. & Classen, J.
ischemic stroke. Acta Neurol. Scand. 121, 141. Ilmoniemi, R. J. et al. Neuronal responses to Plasticity resembling spike-timing dependent
30–37 (2010). magnetic stimulation reveal cortical reactivity synaptic plasticity: the evidence in human
125. Hesse, S. et al. Combined transcranial direct and connectivity. Neuroreport 8, 3537–3540 cortex. Front. Synaptic Neurosci. 2, 34
current stimulation and robot-assisted arm (1997). (2010).
training in subacute stroke patients: an 142. Ferreri, F. et al. Does an intraneural interface 161. Stefan, K., Kunesch, E., Benecke, R.,
exploratory, randomized multicenter trial. short-term implant for robotic hand control Cohen, L. G. & Classen, J. Mechanisms
Neurorehabil. Neural Repair 25, 838–846 modulate sensorimotor cortical integration? of enhancement of human motor cortex
(2011). An EEG–TMS co-registration study on a human excitability induced by interventional paired
126. Bolognini, N. et al. Neurophysiological and amputee. Restor. Neurol. Neurosci. 32, 281–292 associative stimulation. J. Physiol. 543,
behavioral effects of tDCS combined with (2014). 699–708 (2002).
constraint-induced movement therapy in 143. Premoli, I. et al. TMS-EEG signatures of 162. Wolters, A. et al. Timing-dependent plasticity in
poststroke patients. Neurorehabil. Neural Repair GABAergic neurotransmission in the human human primary somatosensory cortex. J. Physiol.
25, 819–829 (2011). cortex. J. Neurosci. 34, 5603–5612 (2014). 565, 1039–1052 (2005).

NATURE REVIEWS | NEUROLOGY ADVANCE ONLINE PUBLICATION | 11


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

163. Heidegger, T., Krakow, K. & Ziemann, U. Effects mechanisms of transcranial DC-stimulation- 175. Sujith, O. Functional electrical stimulation
of antiepileptic drugs on associative LTP-like induced after-effects of human motor cortex in neurological disorders. Eur. J. Neurol. 15,
plasticity in human motor cortex. Eur. J. Neurosci. excitability. Brain 125, 2238–2247 (2002). 437–444 (2008).
32, 1215–1222 (2010). 170. Powell, J., Pandyan, A. D., Granat, M., 176. Wu, C. W.‑H. & Kaas, J. H. The effects of long-
164. Korchounov, A. & Ziemann, U. Neuromodulatory Cameron, M. & Stott, D. J. Electrical stimulation standing limb loss on anatomical reorganization
neurotransmitters influence LTP-like plasticity in of wrist extensors in poststroke hemiplegia. of the somatosensory afferents in the brainstem
human cortex: a pharmaco-TMS study. Stroke 30, 1384–1389 (1999). and spinal cord. Somatosens. Mot. Res. 19,
Neuropsychopharmacology 36, 1894–1902 171. Conforto, A. B., Kaelin-Lang, A. & Cohen, L. G. 153–163 (2002).
(2011). Increase in hand muscle strength of stroke
165. Nitsche, M. A. et al. Transcranial direct current patients after somatosensory stimulation. Author contributions
stimulation: state of the art 2008. Brain Stim. 1, Ann. Neurol. 51, 122–125 (2002). G.D.P. developed the model and wrote the article.
206–223 (2008). 172. Ng, S. S. & Hui-Chan, C. W. Transcutaneous G.P. searched the literature and wrote the section
166. Ranieri, F. et al. Modulation of LTP at rat electrical nerve stimulation combined with task- on the stimulation protocols and clinical trials. G.A.
hippocampal CA3-CA1 synapses by direct related training improves lower limb functions searched the literature and wrote the section on EEG
current stimulation. J. Neurophysiol. 107, in subjects with chronic stroke. Stroke 38, and repetitive electric stimulation. F.C. searched the
1868–1880 (2012). 2953–2959 (2007). literature and participated in writing the section on
167. Medeiros, L. F. et al. Neurobiological effects of 173. Ada, L. & Foongchomcheay, A. Efficacy of predictors of recovery. F.F. searched the literature
transcranial direct current stimulation: a review. electrical stimulation in preventing or reducing and wrote the section on TMS–EEG in the multimodal
Front. Psychiatry 3, 110 (2012). subluxation of the shoulder after stroke: a meta- diagnostic approach. D.F. was involved in the
168. Nitsche, M. A. et al. Modulating parameters of analysis. Aust. J. Physiother. 48, 257–267 computational development of the model. M.T.
excitability during and after transcranial direct (2002). searched the literature and wrote the section on
current stimulation of the human motor cortex. 174. Price, C. I. & Pandyan, A. Electrical stimulation synaptic dysfunctions following stroke. F.R. provided
J. Physiol. 568, 291–303 (2005). for preventing and treating post-stroke shoulder substantial contributions to discussion of the
169. Liebetanz, D., Nitsche, M. A., Tergau, F. & pain: a systematic Cochrane review. Clin. Rehabil. content. U.Z., J.C.R. and V.D.L. guided the
Paulus, W. Pharmacological approach to the 15, 5–19 (2001). manuscript development and revised the manuscript.

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneurol


View publication stats © 2014 Macmillan Publishers Limited. All rights reserved

You might also like