Sharma, N., Classen, J., & Cohen, L. G. (2013) - Neural Plasticity and Its Contribution To Functional Recovery.

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Handbook of Clinical Neurology, Vol.

110 (3rd series)


Neurological Rehabilitation
M.P. Barnes and D.C. Good, Editors
# 2013 Elsevier B.V. All rights reserved

Chapter 1

Neural plasticity and its contribution to functional recovery


NIKHIL SHARMA1, JOSEPH CLASSEN2, AND LEONARDO G. COHEN 1*
1
Human Cortical Physiology and Stroke Neurorehabilitation Section, National Institute of Neurological
Disorders and Stroke, NIH, Bethesda, MD, USA
2
Department of Neurology, University of Leipzig, Leipzig, Germany

DEFINITION
lesions, one of the focuses of this volume, is already
The idea that the cerebral cortex is dynamically orga- starting to lead to the development of more rational
nized was proposed in 1912, when Brown and Sherring- strategies to facilitate neurorehabilitation (Taub et al.,
ton stimulated the motor cortex of chimpanzees and 2002; Cheeran et al., 2009).
found that “a point which began by yielding primary At a cellular level, neuronal circuits consist of synap-
extension may come to yield primary flexion in the latter tic connections between axons and dendrites. As these
part of the stimulation series” (Brown and Sherrington, circuits extend over the brain there is the potential for
1912). In many investigations since then these phenom- a large number of possible interactive combinations
ena have been referred to as neural plasticity. Neural allowing for great flexibility. Modification of sensory in-
plasticity can be defined as the ability of the central put may induce rapid changes in cortical representations
nervous system (CNS) to adapt in response to changes through various mechanisms including unmasking of
in the environment or lesions. This property of the connections that are silent in the native state (Calford
CNS may involve modifications in overall cognitive and Tweedale, 1991a, b). For example, blocking inhibi-
strategies to successfully cope with new challenges tion pharmacologically within a small region of the
(i.e., attention, behavioral compensation) (Bury and primary motor cortex (M1) immediately unveils new rep-
Jones, 2002), recruitment of new/different neural resentational patterns (Jacobs and Donoghue, 1991),
networks (Johansen-Berg et al., 2002; Fridman et al., through unmasking horizontal excitatory connections
2004; Lotze et al., 2006; Heuninckx et al., 2008), or previously hidden by inhibitory neurons. The strength
changes in strength of such connections or specific brain of these horizontal connections and the balance of
areas in charge of carrying out a particular task (i.e., excitation and inhibition appear to shape cortical repre-
movement, language, vision, hearing) (Cohen et al., sentations. Corticofugal connections make extensive
1997; Grefkes et al., 2008). At the cellular level, changes long-range ( 1 mm) links with other pyramidal tract
in membrane excitability, synaptic plasticity, as well as neurons, and with local inhibitory interneurons
structural changes in dendritic and axonal anatomy as (Landry et al., 1990; McGuire et al., 1991). It is now
measured in vivo and in vitro may be demonstrated in known that long-term potentiation (LTP) can be induced
animals and humans (Clarkson et al., 2010; Li et al., in these horizontal connections of adult M1, contributing
2010). The study of neuroplasticity engages scientists to long-lasting associations among neurons within a mo-
from many different disciplines because of the pro- tor cortical area (Hess and Donoghue, 1994). Moreover,
found implications it has for understanding the func- vertical synaptic pathways in M1 can experience short-
tional underpinnings of action and cognition in the term depression, short-term facilitation, long-term de-
healthy and lesioned brain (Dimyan and Cohen, 2010). pression and, under conditions of disinhibition, also
Mechanistic understanding of neuroplastic changes in LTP (Castro-Alamancos et al., 1995). In addition, slower,
the process of functional recovery following brain progressive plastic changes can be driven by learning

*Correspondence to: Leonardo G. Cohen, M.D., Chief, Human Cortical Physiology Section and Stroke Neurorehabilitation
Clinic, National Institute of Neurological Disorders and Stroke NIH, Building 10, Room 7D54 Bethesda, MD 20892, USA.
Tel: þ1-301-496-9782, Fax:þ1-301-402-7010, E-mail: cohenl1@mail.nih.gov
4 N. SHARMA ET AL.
(Robertson and Irvine, 1989; Chino et al., 1997), following CNS abnormalities during development have
competition with other inputs (Merzenich et al., 1983), been particularly impressive given their ability to re-
and use (Nudo et al., 1996b). establish almost normal behavior (Chen et al., 2002).
Basic science investigations have substantially ad- One such example is the substantial recovery of motor
vanced our understanding of the mechanisms of plasticity function or language in children posthemispherectomy,
and metaplasticity, important in multiple areas of human implemented to ameliorate intractable seizures (Vargha-
cognition such as learning and memory, and in functional Khadem et al., 1997). The potential of neuroplastic
recovery from lesions in the CNS, as in stroke changes to influence behavior and recovery of function
(Buonomano and Merzenich, 1998; Floel and Cohen, was first widely accepted in relation to the developing
2006). The term “metaplasticity” is often, but incorrectly, brain. Only more recently was it understood that neuro-
used interchangeably with “homeostatic” plasticity (see plastic changes of substantial clinical relevance could
below) (Abraham and Bear, 1996; Fischer et al., 1997; occur in the adult CNS and in the elderly (Merzenich
Gentner et al., 2008; Jung and Ziemann, 2009). In the past et al., 1996). It has now been proposed, for example, that
few years it has become evident that these findings have recruitment of wider brain networks in the elderly and
direct implications for the way in which human disease is after stroke may play a beneficial role in maintaining
treated, and new efforts have been invested in research the ability of individuals to carry out specific tasks or
that translates these advances in the basic science domain even in facilitating relearning (Heuninckx et al., 2008;
to the formulation of new, rational strategies for promot- Hummel et al., 2010).
ing recovery of function in humans. To accomplish this
goal, it is important to demonstrate that similar principles FUNCTIONAL RELEVANCE
to those described in animal models apply to the human
Plasticity of cortical representations within and across
cerebral cortex in relevant behavioral settings.
different brain regions is thought to represent the neural
basis underlying sensory substitution, for example in
SITES OF PLASTICITY
blind and deaf humans (Rauschecker, 1995), as well as
In most cases, the cerebral cortex has been the target of in the recovery of motor function after cortical lesions
studies of human plasticity (Wolpaw and Tennissen, like stroke (Nudo et al., 1996a). Although neuroplasti-
2001). However, reorganization requires fine-tuning of city, as defined above, is a ubiquitous phenomenon
activity at cortical as well as subcortical sites. In the (our brain is constantly changing), it may have different
motor domain, for example, spinal processes play a role impact on different behaviors. It may be beneficial (of-
in modulating locomotor learning (Bizzi et al., 2000) and ten referred to as adaptive plasticity, the most common
plasticity after amputations and nerve transections (Wu forms of plasticity studied; Cohen et al., 1997; Lee,
and Kaas, 1999). Plastic changes following deafferenta- 2009), have no influence (representing only epiphenom-
tion can be identified at cortical (Kaas et al., 1983) and ena of the modified behavior), or even result in deleteri-
subcortical (Devor and Wall, 1981) sites. The extent to ous consequences (i.e., maladaptive; Flor et al., 2006) on
which plastic changes detected at cortical levels reflect performance of particular tasks or sensory experiences.
reorganization in subcortical structures is incompletely This concept has been referred to as functional relevance
understood and still underinvestigated (Wu and Kaas, of neuroplasticity. Conceptually, it would not be surpris-
2000). Therefore, it is important to keep in mind that ing that plastic changes in, for example neuronal
the neural substrates of recovery of function are likely networks, may have beneficial implications on a partic-
distributed over multiple sites at different levels of the ular behavior but at a cost to other behavior (Chklovskii
neuroaxis and not restricted to one specific location. It et al., 2004). This concept of cost of neuroplastic changes,
still represents a challenge to understand how these which has been to some extent overlooked, is starting to
different levels interact with one another to accomplish receive attention. Understanding these changes and how
a particular behavioral goal. they can be influenced is pivotal in developing better treat-
ments and therapies for patients (Hodics et al., 2006;
WINDOW OF OPPORTUNITY Hummel and Cohen, 2006; Cramer, 2008).
Neural plasticity occurs throughout the life span (Elias and
PLASTICITY, METAPLASTICITY,
Wagster, 2007). During normal human development the
AND HOMEOSTATIC PLASTICITY
CNS must continue to optimize performance and learn
and adapt in the presence of changes in anatomical con- Plasticity likely depends on multiple mechanisms evolv-
straints (such as, for example, changes in limb length or ing on different temporal scales – minutes to months,
muscle mass or strength) and experience (Gaillard et al., even years. Rapid onset-mechanisms, which may operate
2000). Additionally, neuroplastic changes identified over a limited period of time, are believed to represent
NEURAL PLASTICITY AND ITS CONTRIBUTION TO FUNCTIONAL RECOVERY 5
initial steps of more slowly evolving processes of reorga- decrease the induction threshold, thereby increasing the
nization through which functional gains (or losses) may probability for LTP. Alternatively, a history of enhanced
be sustained (Classen et al., 1998; Kleim and Jones, postsynaptic activity would increase the threshold for
2008). At the level of neuronal synapses, multiple trans- LTP and therefore increase the likelihood for long-term
formations may occur from relatively short-lasting LTP, depression induction (Ragert et al., 2009). Based on this
which appears to be largely independent of protein combination of basic science and human neurophysiolog-
synthesis, to long-lasting LTP, which may persist along ical evidence, it is attractive to speculate that the response
the life span. These synaptic changes are complemented to motor training protocols could depend on the history of
by changes in neuronal excitability and structural activity at the time training is imparted. In other words,
changes, with the latter ones being detectable using activities carried out in the period of time preceding the
light microscopy, for example. From a behavioural per- actual rehabilitative treatment (sleep, caffeine intake,
spective, consolidation refers to a process that results in reading, feeding, etc.) could have substantial influence,
enduring performance improvements (Cohen et al., so far not well characterized, on outcomes and perhaps
2005; Krakauer, 2009) that is underway during training to some extent contribute to well-described interindividual
or learning but typically occurs after. Through consoli- variability in treatment response. Human studies have
dation, newly acquired skills become more robust in indeed provided experimental support for a homeostatic
the face of disruptive experiences or may even improve model of plasticity (Jung and Ziemann, 2009). On
further, a process termed off-line learning. Through the other hand, experimental or therapeutic manipulations
reconsolidation, stored memories may be purposefully applied after the treatment may also provide an
modified in order to strengthen or weaken them opportunity for modulating the ultimate behavioral
(Censor et al., 2010). response (Reis et al., 2009).
Metaplasticity refers to the influence that baseline
neural activity immediately preceding presentation of GENETIC INFLUENCES
a plasticity-inducing protocol (for example in vitro theta
Genetic factors can influence the human brain’s ability
burst stimulation) can substantially influence the ability
to experience neuroplastic changes. For example, a
of neuronal elements to exhibit plasticity. The functional
genetic polymorphism (Val66Met) in brain-derived neu-
significance of metaplasticity may include, but is not
rotrophic factor (BDNF) reduces electrophysiological
limited to, controlling homeostasis of neural network
measurements of training-dependent plasticity of the
excitability, for example, by virtue of modifying synap-
primary motor cortex (Kleim et al., 2006; Cheeran
tic efficacy in an operational range. Metaplasticity may,
et al., 2008). Although the implications for motor
therefore, protect against potentially noxious excitability
learning and recovery of function need to be firmly
increases (Abraham and Bear, 1996). It should be kept in
established, these factors could partially explain interin-
mind that the specific characteristics of the baseline
dividual variability in functional recovery or response to
activity preceding application of the same plasticity-
pharmacological or training-based interventions. BDNF
inducing protocol can then result in opposite effects
is almost certainly one of the first of many possible ge-
on synaptic efficacy (Seol et al., 2007). More attention
netic polymorphisms that affect training-dependent
is now paid to these phenomena in humans because it
plasticity and also the ability to learn (Fritsch et al.,
is thought that they can substantially impact information
2010). Other mechanisms such as polymorphisms in
coding and cortical reorganization (Gentner et al., 2008;
the gene coding for catechol-O-methyltransferase,
Jung and Ziemann, 2009).
serotonin transporter, and other proteins involved in
An example in humans is that the magnitude of the
modulating or regulating neurotransmission are
response to noninvasive brain stimulation protocols
being explored and will likely lead to more individually
applied to the primary motor cortex critically depends
tailored rehabilitative protocols after brain lesions
on the previous history of neural activity (Ziemann
(Pearson-Fuhrhop et al., 2009).
et al., 1998; Iyer et al., 2003; Gentner et al., 2008). As long
as the modulation is confined to the magnitude, but not
NONINVASIVE TECHNIQUES CAPABLE
the sign, of responses (increases vs. decreases in excitabil-
OF EVALUATING NEUROPLASTICITY
ity, for example), most of these findings may be
IN HUMANS
interpreted within the framework of homeostatic plastic-
ity as proposed in the Bienenstock–Cooper–Munro One important factor that contributed to substantial
(BCM) theory of synaptic plasticity (Bienenstock et al., advances in the understanding of neuroplasticity at a
1982; Abraham and Bear, 1996). Fundamental to the systems level in the human brain has been the develop-
BCM theory is a time-variable induction threshold. For ment of techniques that allowed the noninvasive mea-
example, prolonged low levels of postsynaptic activity surement of these changes. Techniques like positron
6 N. SHARMA ET AL.
emission tomography, magnetic resonance imaging and white matter have also been described in the elderly
(MRI), both functional (fMRI) and structural (particu- (Boyke et al., 2008). A word of caution: the cellular
larly diffusion tensor imaging, DTI), magneto (MEG) changes that underpin these changes are not yet clear.
and electro (EEG) encephalography, and transcranial Nevertheless, in healthy volunteers, learning to juggle
magnetic (TMS) and direct current stimulation have produces changes in gray matter density as well as in
all played important roles in the noninvasive evaluation white matter (Scholz et al., 2009) in biologically plausible
of neuroplastic processes associated with recovery of regions. Although there are numerous technical difficul-
function after CNS lesions. These techniques provide in- ties that have prevented the application of these tech-
formation on the possible relation between anatomical niques to the lesioned brain, some interesting studies
connectivity (DTI) or functional activity in specific brain are starting to appear in patient populations. In a recent
areas as well as interactions between neural networks study it was shown that constraint-induced therapy, a
(fMRI) and a particular behavior, recovery process, or treatment proposed to improve motor function after
response to treatment. fMRI has excellent spatial resolu- stroke (Wolf et al., 2006), induced increases in matter
tion but less sharp temporal resolution and alone does not density in the affected hemisphere, which is in keeping
allow firm conclusions on causeeffect links between with functional MRI data (Gauthier et al., 2008). There
these associations (Cohen et al., 1997). were also increases in gray matter density in the nonaf-
Dramatic increases in readily available computational fected hemisphere, which are difficult to predict in our
power have led to the development of novel analytical current framework of understanding. In some cases,
approaches to functional imaging. Some of these apply identification of these changes allows the formulation
economic theories, such as structural equation modeling of predicting algorithms after stroke (Stinear, 2010).
(Simon, 1953) and Granger’s causality (Granger, 1969), In contrast to MRI techniques, MEG and EEG allow a
to model the interactions between (sub)cortical regions millisecond by millisecond analysis of the activity in
(Deshpande et al., 2009; Kim and Horwitz, 2009). functional networks in relation to behavior (Birbaumer
Dynamic causal modeling also explores regional interac- and Cohen, 2007; Pantev et al., 2009). As such, they
tions but does so within a Bayesian framework (Penny can provide information on the timing of neuroplastic
et al., 2004). These approaches explore interactions that changes or serial processing in a way that imaging tech-
are overlooked by conventional activation analysis niques alone still cannot. Additionally, MEG and EEG do
(Rowe et al., 2002). provide important information on activity in neural net-
Model “free” analysis using multivariate statistical works with a very accurate temporal resolution (Fujioka
approaches (such as independent component analysis et al., 2006). Of note is that activity originating in spe-
and principal component analysis) have established the cific brain networks as measured with MEG has been
existence of resting state networks (De Luca et al., successfully used to control a hand orthosis that controls
2006). The study of these networks represents a fertile movements of a completely paralyzed hand after stroke
area of research given their ability to experience neuro- (Buch et al., 2008). Similar results in terms of output
plastic changes, for example in relation to learning control using EEG in completely paralyzed patients
(Albert et al., 2009; Mantini et al., 2009). Changes in rest- have been reported previously (Birbaumer et al., 1999;
ing networks may be lost in classical fMRI study designs Wolpaw and McFarland, 2004).
that involve contrasting one condition (task) with a Noninvasive brain stimulation techniques have
control condition (most commonly rest). Another devel- contributed in different ways to the evaluation of sys-
opment that takes advantage of the increasing sophisti- tems’ neuroplasticity in the healthy and lesioned brain.
cation in these analytical tools is real-time fMRI In particular, TMS allows the evaluation of the behav-
(deCharms, 2008). The excitement of this approach is ioral consequences of disruption of activity (virtual
that it potentially can demonstrate the ability of training lesion) in relatively focal brain regions, for example
in human and nonhuman subjects to recruit specific those shown to be active during a particular behavior
brain regions or neural networks. in fMRI studies (Pascual-Leone et al., 2000). Disruption
Although a lot of work has focused on the evaluation of a specific behavior as a consequence of TMS-induced
of dynamic changes in neural networks, there is mount- disruption of a particular brain region has been inter-
ing evidence that motor training can induce structural preted as indicative of a causeeffect link between
changes as well. These include changes in gray matter the two (Cohen et al., 1997). In this sense, neuroimaging
density (measured using voxel-based morphometry) and TMS virtual lesion experiments are complementary.
(Smith et al., 2007; Wrigley et al., 2009) and in white An example of the way in which these two techniques op-
matter (measured using DTI) (Johansen-Berg, 2007; erate to address fundamental hypotheses in motor control
Ciccarelli et al., 2008; Johansen-Berg and Rushworth, has been the evaluation of the role of the supplementary
2009; Scholz et al., 2009). Structural changes in gray motor area or primary motor cortex in motor learning
NEURAL PLASTICITY AND ITS CONTRIBUTION TO FUNCTIONAL RECOVERY 7
(Muellbacher et al., 2002; Perez et al., 2007; Censor et al.,
2010). In another example of how these tools can be
creatively utilized, TMS has been applied in the
fMRI environment to examine fundamental questions
of interregional connectivity within neural networks, oth-
PMd PMd PMv
erwise impossible to address experimentally (Bestmann *left
et al., 2005). This armamentarium has created an impor-
SMA
tant momentum in human systems neuroscience, making *leftPMd®rightM1

possible the experimental evaluation of hypotheses until M1 M1


recently beyond the scope of investigation. PPC PPC
THAL
CBL
MODULATION OF NEUROPLASTICITY
Given its proposed influence on learning processes and
recovery of function, one goal of present investigations PNS
has been to develop strategies to modulate neuroplasti- Fig. 1.1. The currently described influences of other brain
city: to facilitate it when it plays an adaptive function and areas on the output of the primary motor cortex (M1) are
downregulate it when it is maladaptive (see above). shown. Open arrows denote facilitation, while filled arrows de-
Different approaches have been tried in animal and note inhibition. In many cases the influence shown represents a
human settings. Use of pharmacological agents like net effect of several specific interactions, whose details are dis-
amphetamine, L-dopa, or selective serotonin reuptake in- cussed in the relevant section of the text and are shown in
hibitors (SSRIs) (for review see Floel and Cohen, 2010) in subsequent figures. These influences include projections from
association with motor training protocols may result in be- motor areas in the ipsi- and contralateral hemispheres and the
havioral gains accompanied by cortical reorganization in effects of afferent sensory input. PMd ¼ dorsal premotor cor-
tex; PMv ¼ ventral premotor cortex; SMA ¼ supplementary
humans (Tardy et al., 2006). Clearly, the development of
motor area; PPC ¼ posterior parietal cortex; CBL ¼
better training protocols that take into account advances cerebellum; THAL ¼ thalamus; PNS ¼ peripheral nervous
in basic science is an important area of research (Luft system. (Figure reproduced with permission from Reis
et al., 2004; Wolf et al., 2006; Ramachandran and Altschu- et al., 2008b.)
ler, 2009). Examples of proposed new rehabilitative
paradigms include the combination of customarily used
training protocols with action observation (Stefan et al., advantages of combining different stimulating modali-
2005), motor imagery (Sharma et al., 2006; Page et al., ties have been suggested as well. For example, a combi-
2009; Sharma et al., 2009), and focused attention (Stefan nation of facilitatory stimulation of the ipsilesional and
et al., 2004) thought to ameliorate function by inhibitory stimulation of the contralesional primary motor
facilitating the mirror neuron system (Nelissen et al., 2005). cortices (Vines et al., 2008) or combination of peripheral
In addition to these approaches, recent years have nerve stimulation applied to the paretic hand and facilita-
seen the formulation of interventions based on better tory stimulation of the ipsilesional M1 (Celnik et al., 2009)
understood principles of intracortical interactive func- in association with training have shown potential benefits
tions (see Fig. 1.1 for a summary of the interregional in patients with chronic stroke. Invasive cortical stimula-
influences on M1). For example, the identification tion through epidural electrodes over the primary motor
of persistent interhemispheric inhibitory interactions cortex may have similar effects and has been proposed
between the primary motor cortices after stroke after stroke in humans and in animal models (Brown
(Murase et al., 2004) led to the hypothesis that facilitat- et al., 2008; Plow et al., 2009). While most reports seem
ing excitability in the ipsilesional M1 or downregulating to point to benefits of these techniques, it should be kept
excitability in the contralesional M1 could enhance either in mind that negative results are often underreported and
static excitability or plastic processes leading to im- that results from well-controlled, multicenter clinical
proved function. Both strategies have been tried in trials under way are still not available.
different laboratories leading to promising proof of Manipulation of somatosensory input elicits clear
principle behavioral and physiological results (for effects on somatosensory as well as motor function.
review see Fregni and Pascual-Leone, 2006; Hummel In healthy humans, somatosensory stimulation of me-
and Cohen, 2006; Talelli and Rothwell, 2006). Studies dian, ulnar, and/or radial nerves at the wrist induces clear
in healthy subjects pointed to the importance of increases in fMRI activation and cortical excitability in
synchronous application of cortical stimulation to M1 the stimulated hand motor cortical representations
and motor training protocols (Reis et al., 2008a). Possible (Ridding et al., 2001; Kaelin-Lang et al., 2002; Wu
8 N. SHARMA ET AL.
et al., 2005; Conforto et al., 2010) while hand anesthesia Bestmann S, Baudewig J, Siebner HR et al. (2005). BOLD
induces increases in excitability and improved tactile MRI responses to repetitive TMS over human dorsal
discrimination in the non-anesthesized hand (Werhahn premotor cortex. Neuroimage 28: 22–29.
et al., 2002a, b). In stroke patients, somatosensory Bienenstock EL, Cooper LN, Munro PW (1982). Theory
for the development of neuron selectivity: orientation
stimulation of the paretic limb (Conforto et al., 2002;
specificity and binocular interaction in visual cortex. J
Sheffler et al., 2006; Celnik et al., 2007) and anesthesia
Neurosci 2: 32–48.
of the non-paretic hand (Floel et al., 2004) show compa- Birbaumer N, Cohen LG (2007). Brain-computer interfaces:
rable short-lasting behavioral and electrophysiological communication and restoration of movement in paralysis.
beneficial effects on paretic hand function, consistent J Physiol 579: 621–636.
with the documented correction of abnormalities in Birbaumer N, Ghanayim N, Hinterberger T et al. (1999).
interhemispheric interactions between the primary A spelling device for the paralysed. Nature 398: 297–298.
motor cortices (Murase et al., 2004; Floel et al., 2008). Bizzi E, Tresch MC, Saltiel P et al. (2000). New perspectives
Results from studies using transcutaneous electrical on spinal motor systems. Nat Rev Neurosci 1: 101–108.
stimulation are consistent with those carried out using Boyke J, Driemeyer J, Gaser C et al. (2008). Training-induced
peripheral nerve stimulation and should therefore be brain structure changes in the elderly. J Neurosci 28:
7031–7035.
considered in neurorehabilitation.
Brown JA, Lutsep HL, Weinand M et al. (2008). Motor cortex
More information is becoming available on the neural
stimulation for the enhancement of recovery from stroke: a
mechanisms underlying recovery of motor function and prospective, multicenter safety study. Neurosurgery 62:
neuroplasticity after stroke (see for example Prabhakaran 853–862.
et al., 2008; Swayne et al., 2008; Marshall et al., 2009). Brown TG, Sherrington CS (1912). On the instability of a
An emerging body of evidence is providing new insight cortical point. Proc R Soc Lond (Biol) 85: 250–277.
into the interregional interactions between the premotor Buch E, Weber C, Cohen LG et al. (2008). Think to move: a
and parietal areas and primary motor cortex in healthy in- neuromagnetic brain-computer interface (BCI) system
dividuals, as well as strategies to modulate the strength of for chronic stroke. Stroke 39: 910–917.
these interactions (Koch et al., 2006; Koch and Rothwell, Buonomano DV, Merzenich MM (1998). Cortical plasticity:
2009). It is only reasonable to expect the development of from synapses to maps. Ann Rev Neurosci 21: 149–186.
Bury SD, Jones TA (2002). Unilateral sensorimotor cortex
newer interventional proposals based on the emerging
lesions in adult rats facilitate motor skill learning with
understanding of these mechanisms.
the “unaffected” forelimb and training-induced dendritic
structural plasticity in the motor cortex. J Neurosci 22:
8597–8606.
CONCLUSIONS Calford MB, Tweedale R (1991a). Acute changes in cutaneous
receptive fields in primary somatosensory cortex after
The last decade saw impressive improvements in our
digit denervation in adult flying fox. J Neurophysiol 65:
understanding of the ability of the CNS to reorganize
178–187.
in response to changes in the environment and lesions. Calford MB, Tweedale R (1991b). Immediate expansion
This understanding resulted in parallel gains in our in- of receptive fields of neurons in area 3b of macaque
sight into mechanisms of both action and cognition in monkeys after digit denervation. Somatosens Mot Res 8:
health and disease. Understanding of these neuroplastic 249–260.
principles is evolving into the development of more ra- Castro-Alamancos MA, Donoghue JP, Connors BW (1995).
tional, hypothesis-driven strategies to promote recovery Different forms of synaptic plasticity in somatosensory
of function and will likely result in improvements in and motor areas of the neocortex. J Neurosci 15: 5324–5333.
patient care along the bench to bedside translational Celnik P, Hummel F, Harris-Love M et al. (2007). Somatosen-
pipeline (Cheeran et al., 2009). sory stimulation enhances the effects of training functional
hand tasks in patients with chronic stroke. Arch Phys Med
Rehabil 88: 1369–1376.
ACKNOWLEDGMENTS
Celnik P, Paik NJ, Vandermeeren Y et al. (2009). Effects of
This work was supported by the Intramural Research combined peripheral nerve stimulation and brain polariza-
Program of the NINDS, NIH. tion on performance of a motor sequence task after chronic
stroke. Stroke 40: 1764–1771.
REFERENCES Censor N, Dimyan MA, Cohen LG (2010). Modification of
existing human motor memories is enabled by primary
Abraham WC, Bear MF (1996). Metaplasticity: the plasticity cortical processing during memory reactivation. Curr Biol
of synaptic plasticity. Trends Neurosci 9: 126–130. 20: 1545–1549.
Albert NB, Robertson EM, Miall RC (2009). The resting Cheeran B, Talelli P, Mori F et al. (2008). A common polymor-
human brain and motor learning. Curr Biol 19: 1023–1027. phism in the brain-derived neurotrophic factor gene
NEURAL PLASTICITY AND ITS CONTRIBUTION TO FUNCTIONAL RECOVERY 9
(BDNF) modulates human cortical plasticity and the Fischer TM, Blazis DE, Priver NA et al. (1997). Metaplasticity
response to rTMS. J Physiol 586: 5717–5725. at identified inhibitory synapses in Aplysia [see comments].
Cheeran B, Cohen L, Dobkin B et al. (2009). The future of Nature 389: 860–865.
restorative neurosciences in stroke: driving the transla- Floel A, Cohen LG (2006). Translational studies in neuroreha-
tional research pipeline from basic science to rehabilitation bilitation: from bench to bedside. Cogn Behav Neurol 19:
of people after stroke. Neurorehabil Neural Repair 23: 1–10.
97–107. Floel A, Cohen LG (2010). Recovery of function in humans:
Chen R, Cohen LG, Hallett M (2002). Nervous system reorga- cortical stimulation and pharmacological treatments after
nization following injury. Neuroscience 111: 761–773. stroke. Neurobiol Dis 37: 243–251.
Chino YM, Smith EL, Hatta S et al. (1997). Postnatal develop- Floel A, Nagorsen U, Werhahn KJ et al. (2004). Influence of
ment of binocular disparity sensitivity in neurons of the pri- somatosensory input on motor function in patients with
mate visual cortex. J Neurosci 17: 296–307. chronic stroke. Ann Neurol 56: 206–212.
Chklovskii DB, Mel BW, Svoboda K (2004). Cortical rewiring Floel A, Hummel F, Duque J et al. (2008). Influence of somato-
and information storage. Nature 431: 782–788. sensory input on interhemispheric interactions in patients
Ciccarelli O, Catani M, Johansen-Berg H et al. (2008). with chronic stroke. Neurorehabil Neural Repair 22:
Diffusion-based tractography in neurological disorders: 477–485.
concepts, applications, and future developments. Lancet Flor H, Nikolajsen L, Staehelin Jensen T (2006). Phantom limb
Neurol 7: 715–727. pain: a case of maladaptive CNS plasticity? Nat Rev Neu-
Clarkson AN, Huang BS, Macisaac SE et al. (2010). Reducing rosci 7: 873–881.
excessive GABA-mediated tonic inhibition promotes func- Fregni F, Pascual-Leone A (2006). Hand motor recovery after
tional recovery after stroke. Nature 468: 305–309. stroke: tuning the orchestra to improve hand motor func-
Classen J, Liepert J, Wise SP et al. (1998). Rapid plasticity of tion. Cogn Behav Neurol 19: 21–33.
human cortical movement representation induced by prac- Fridman EA, Hanakawa T, Chung M et al. (2004). Reorgani-
tice. J Neurophysiol 79: 1117–1123. zation of the human ipsilesional premotor cortex after
Cohen DA, Pascual-Leone A, Press DZ et al. (2005). Off-line stroke. Brain 127: 747–758.
learning of motor skill memory: a double dissociation of goal Fritsch B, Reis J, Martinowich K et al. (2010). Direct current
and movement. Proc Natl Acad Sci U S A 102: 18237–18241. stimulation promotes BDNF-dependent synaptic plasticity:
Cohen LG, Celnik P, Pascual-Leone A et al. (1997). Functional potential implications for motor learning. Neuron 66:
relevance of cross-modal plasticity in blind humans. Nature 198–204.
389: 180–183. Fujioka T, Ross B, Kakigi R et al. (2006). One year of musical
Conforto AB, Kaelin-Lang A, Cohen LG (2002). Increase in training affects development of auditory cortical-evoked
hand muscle strength of stroke patients after somatosen- fields in young children. Brain 129: 2593–2608.
sory stimulation. Ann Neurol 51: 122–125. Gaillard WD, Hertz-Pannier L, Mott SH et al. (2000).
Conforto AB, Ferreiro KN, Tomasi C et al. (2010). Effects of Functional anatomy of cognitive development: fMRI of
somatosensory stimulation on motor function after sub- verbal fluency in children and adults. Neurology 54:
acute stroke. Neurorehabil Neural Repair 24: 263–272. 180–185.
Cramer SC (2008). Repairing the human brain after stroke. II. Gauthier LV, Taub E, Perkins C et al. (2008). Remodeling
Restorative therapies. Ann Neurol 63: 549–560. the brain: plastic structural brain changes produced
deCharms RC (2008). Applications of real-time fMRI. Nat by different motor therapies after stroke. Stroke 39: 1520–1525.
Rev Neurosci 9: 720–729. Gentner R, Wankerl K, Reinsberger C et al. (2008). De-
De Luca M, Beckmann CF, De Stefano N et al. (2006). fMRI pression of human corticospinal excitability induced
resting state networks define distinct modes of long- by magnetic theta-burst stimulation: evidence of rapid
distance interactions in the human brain. Neuroimage 29: polarity-reversing metaplasticity. Cereb Cortex 18:
1359–1367. 2046–2053.
Deshpande G, LaConte S, James GA et al. (2009). Multivariate Granger C (1969). Investigating causal relations by economet-
Granger causality analysis of fMRI data. Hum Brain Mapp ric models and cross-spectral methods. Econometrica 37:
30: 1361–1373. 424–438.
Devor M, Wall P (1981). Effect of peripheral nerve injury on Grefkes C, Nowak DA, Eickhoff SB et al. (2008).
receptive fields of cells in the cat spinal cord. J Comp Neu- Cortical connectivity after subcortical stroke assessed with
rol 199: 277–291. functional magnetic resonance imaging. Ann Neurol 63:
Dimyan MA, Cohen LG (2010). Contribution of transcranial 236–246.
magnetic stimulation to the understanding of functional re- Hess G, Donoghue JP (1994). Long-term potentiation of
covery mechanisms after stroke. Neurorehabil Neural horizontal connections provides a mechanism to reorganize
Repair 24: 125–135. cortical motor maps. J Neurophysiol 71: 2543–2547.
Elias JW, Wagster MV (2007). Developing context and back- Heuninckx S, Wenderoth N, Swinnen SP (2008). Systems
ground underlying cognitive intervention/training studies neuroplasticity in the aging brain: recruiting additional
in older populations. J Gerontol B Psychol Sci Soc Sci neural resources for successful motor performance in
62: 5–10. elderly persons. J Neurosci 28: 91–99.
10 N. SHARMA ET AL.
Hodics T, Cohen LG, Cramer SC (2006). Functional imaging Landry P, Labelle A, Deschenes M (1990). Intracortical distri-
of intervention effects in stroke motor rehabilitation. Arch bution of axonal collaterals of pyramidal tract cells in the
Phys Med Rehabil 87: S36–S42. cat motor cortex. Brain Res 191: 327–336.
Hummel FC, Cohen LG (2006). Non-invasive brain stimula- Lee JL (2009). Reconsolidation: maintaining memory rele-
tion: a new strategy to improve neurorehabilitation after vance. Trends Neurosci 32: 413–420.
stroke? Lancet Neurol 5: 708–712. Li S, Overman JJ, Katsman D et al. (2010). An age-related
Hummel FC, Heise K, Celnik P et al. (2010). Facilitating sprouting transcriptome provides molecular control
skilled right hand motor function in older subjects by of axonal sprouting after stroke. Nat Neurosci 13:
anodal polarization over the left primary motor cortex. 1496–1504.
Neurobiol Aging 31: 2160–2168. Lotze M, Markert J, Sauseng P et al. (2006). The role of
Iyer MB, Schleper N, Wassermann EM (2003). Priming stim- multiple contralesional motor areas for complex hand
ulation enhances the depressant effect of low-frequency movements after internal capsular lesion. J Neurosci 26:
repetitive transcranial magnetic stimulation. J Neurosci 6096–6102.
23: 10867–10872. Luft AR, McCombe-Waller S, Whitall J et al. (2004). Repet-
Jacobs KM, Donoghue JP (1991). Reshaping the cortical itive bilateral arm training and motor cortex activation in
motor map by unmasking latent intracortical connections. chronic stroke: a randomized controlled trial. JAMA 292:
Science 251: 944–947. 1853–1861.
Johansen-Berg H (2007). Structural plasticity: rewiring the Mantini D, Corbetta M, Perrucci MG et al. (2009). Large-scale
brain. Curr Biol 17: R141–R144. brain networks account for sustained and transient activity
Johansen-Berg H, Rushworth MF (2009). Using diffusion during target detection. Neuroimage 44: 265–274.
imaging to study human connectional anatomy. Annu Marshall RS, Zarahn E, Alon L et al. (2009). Early imaging
Rev Neurosci 32: 75–94. correlates of subsequent motor recovery after stroke. Ann
Johansen-Berg H, Rushworth MF, Bogdanovic MD et al. Neurol 65: 596–602.
(2002). The role of ipsilateral premotor cortex in hand McGuire BA, Gilbert CD, Rivlin PK et al. (1991). Targets of
movement after stroke. Proc Natl Acad Sci U S A 99: horizontal connections in macaque primary visual cortex.
14518–14523. J Comp Neurol 305: 370–392.
Jung P, Ziemann U (2009). Homeostatic and nonhomeostatic Merzenich MM, Kaas JH, Wall JT et al. (1983). Progression of
modulation of learning in human motor cortex. J Neurosci change following median nerve section in the cortical rep-
29: 5597–5604. resentation of the hand in areas 3b and 1 in adult owl and
Kaas JH, Merzenich MM, Killackey HP (1983). The reorgani- squirrel monkeys. Neuroscience 10: 639–665.
zation of somatosensory cortex following peripheral nerve Merzenich M, Wright B, Jenkins W et al. (1996). Cortical
damage in adult and developing mammals. Annu Rev plasticity underlying perceptual, motor, and cognitive skill
Neurosci 6: 325–356. development: implications for neurorehabilitation. Cold
Kaelin-Lang A, Luft AR, Sawaki L et al. (2002). Modulation Spring Harb Symp Quant Biol 61: 1–8.
of human corticomotor excitability by somatosensory Muellbacher W, Ziemann U, Wissel J et al. (2002). Early con-
input. J Physiol 540: 623–633. solidation in human primary motor cortex. Nature 415:
Kim J, Horwitz B (2009). How well does structural equation 640–644.
modeling reveal abnormal brain anatomical connections? Murase N, Duque J, Mazzocchio R et al. (2004). Influence of
An fMRI simulation study. Neuroimage 45: 1190–1198. interhemispheric interactions on motor function in chronic
Kleim JA, Jones TA (2008). Principles of experience- stroke. Ann Neurol 55: 400–409.
dependent neural plasticity: implications for rehabilita- Nelissen K, Luppino G, Vanduffel W et al. (2005). Observing
tion after brain damage. J Speech Lang Hear Res 51: others: multiple action representation in the frontal lobe.
S225–S239. Science 310: 332–336.
Kleim JA, Chan S, Pringle E et al. (2006). BDNF val66met Nudo RJ, Wise BM, SiFuentes FS et al. (1996a). Neural
polymorphism is associated with modified experience- substrates for the effects of rehabilitative training on
dependent plasticity in human motor cortex. Nat Neurosci motor recovery after ischemic infarct. Science 272:
9: 735–737. 1791–1794.
Koch G, Rothwell JC (2009). TMS investigations into the Nudo RJ, Milliken GW, Jenkins WM et al. (1996b).
task-dependent functional interplay between human Use-dependent alterations of movement representations
posterior parietal and motor cortex. Behav Brain Res 202: in primary motor cortex of adult squirrel monkeys. J Neu-
147–152. rosci 16: 785–807.
Koch G, Franca M, Del Olmo MF et al. (2006). Time course of Page SJ, Szaflarski JP, Eliassen JC et al. (2009). Cortical
functional connectivity between dorsal premotor and plasticity following motor skill learning during
contralateral motor cortex during movement selection. mental practice in stroke. Neurorehabil Neural Repair 23:
J Neurosci 26: 7452–7459. 382–388.
Krakauer JW (2009). Motor learning and consolidation: the Pantev C, Lappe C, Herholz SC et al. (2009). Auditory-
case of visuomotor rotation. Adv Exp Med Biol 629: somatosensory integration and cortical plasticity in musical
405–421. training. Ann N Y Acad Sci 1169: 143–150.
NEURAL PLASTICITY AND ITS CONTRIBUTION TO FUNCTIONAL RECOVERY 11
Pascual-Leone A, Walsh V, Rothwell J (2000). Transcranial Sharma N, Simmons LH, Jones PS et al. (2009). Motor
magnetic stimulation in cognitive neuroscience – virtual imagery after subcortical stroke: a functional magnetic res-
lesion, chronometry, and functional connectivity. Curr onance imaging study. Stroke 40: 1315–1324.
Opin Neurobiol 10: 232–237. Sheffler LR, Hennessey MT, Naples GG et al. (2006). Peroneal
Pearson-Fuhrhop KM, Kleim JA, Cramer SC (2009). Brain nerve stimulation versus an ankle foot orthosis for correction
plasticity and genetic factors. Top Stroke Rehabil 16: of footdrop in stroke: impact on functional ambulation.
282–299. Neurorehabil Neural Repair 20: 355–360.
Penny WD, Stephan KE, Mechelli A et al. (2004). Modelling Simon HA (1953). Causal ordering and identifiability. In:
functional integration: a comparison of structural equa- WC Hood, TC Koopmans (Eds.), Studies in Econometric
tion and dynamic causal models. Neuroimage 23: S264–S274. Method. Wiley, New York, pp. 49–74.
Perez MA, Tanaka S, Wise SP et al. (2007). Neural substrates Smith CD, Chebrolu H, Wekstein DR et al. (2007). Age and
of intermanual transfer of a newly acquired motor skill. gender effects on human brain anatomy: a voxel-based
Curr Biol 17: 1896–1902. morphometric study in healthy elderly. Neurobiol Aging
Plow EB, Carey JR, Nudo RJ et al. (2009). Invasive cortical 28: 1075–1087.
stimulation to promote recovery of function after stroke: Stefan K, Wycislo M, Classen J (2004). Modulation of associa-
a critical appraisal. Stroke 40: 1926–1931. tive human motor cortical plasticity by attention. J
Prabhakaran S, Zarahn E, Riley C et al. (2008). Inter-individual Neurophysiol 92: 66–72.
variability in the capacity for motor recovery after ischemic Stefan K, Cohen LG, Duque J et al. (2005). Formation of a motor
stroke. Neurorehabil Neural Repair 22: 64–71. memory by action observation. J Neurosci 25: 9339–9346.
Ragert P, Camus M, Vandermeeren Y et al. (2009). Modulation Stinear C (2010). Prediction of recovery of motor function
of effects of intermittent theta burst stimulation applied over after stroke. Lancet Neurol 9: 1228–1232.
primary motor cortex (M1) by conditioning stimulation of Swayne OB, Rothwell JC, Ward NS et al. (2008). Stages of
the opposite M1. J Neurophysiol 102: 766–773. motor output reorganization after hemispheric stroke
Ramachandran VS, Altschuler EL (2009). The use of visual suggested by longitudinal studies of cortical physiology.
feedback, in particular mirror visual feedback, in restoring Cereb Cortex 18: 1909–1922.
brain function. Brain 132: 1693–1710. Talelli P, Rothwell J (2006). Does brain stimulation after
Rauschecker JP (1995). Compensatory plasticity and sensory stroke have a future? Curr Opin Neurol 19: 543–550.
substitution in the cerebral cortex. Trends Neurosci 18: 36–43. Tardy J, Pariente J, Leger A et al. (2006). Methylphenidate
Reis J, Robertson E, Krakauer JW et al. (2008a). Consensus: modulates cerebral post-stroke reorganization. Neuro-
“Can tDCS and TMS enhance motor learning and memory image 33: 913–922.
formation?”. Brain Stimul 1 (4): 363–369. Taub E, Uswatte G, Elbert T (2002). New treatments in
Reis J, Swayne OB, Vandermeeren Y et al. (2008b). Contribu- neurorehabilitation founded on basic research. Nat Rev
tion of transcranial magnetic stimulation to the understand- Neurosci 3: 228–236.
ing of cortical mechanisms involved in motor control. Vargha-Khadem F, Carr LJ, Isaacs E et al. (1997). Onset of
J Physiol 586 (2): 325–351. speech after left hemispherectomy in a nine-year-old
Reis J, Schambra HM, Cohen LG et al. (2009). Noninvasive boy. Brain 120: 159–182.
cortical stimulation enhances motor skill acquisition over Vines BW, Cerruti C, Schlaug G (2008). Dual-hemisphere
multiple days through an effect on consolidation. Proc Natl tDCS facilitates greater improvements for healthy subjects’
Acad Sci U S A 106: 1590–1595. non-dominant hand compared to uni-hemisphere stimula-
Ridding MC, McKay DR, Thompson PD et al. (2001). tion. BMC Neurosci 9: 103.
Changes in corticomotor representations induced by pro- Werhahn KJ, Mortensen J, Kaelin-Lang A et al. (2002a).
longed peripheral nerve stimulation in humans. Clin Cortical excitability changes induced by deafferentation
Neurophysiol 112: 1461–1469. of the contralateral hemisphere. Brain 125: 1402–1413.
Robertson D, Irvine DR (1989). Plasticity of frequency orga- Werhahn KJ, Mortensen J, Van Boven RW et al. (2002b).
nization in auditory cortex of guinea pigs with partial uni- Enhanced tactile spatial acuity and cortical process-
lateral deafness. J Comp Neurol 282: 456–471. ing during acute hand deafferentation. Nat Neurosci 5:
Rowe J, Stephan KE, Friston K et al. (2002). Attention 936–938.
to action in Parkinson’s disease: impaired effective connec- Wolf SL, Winstein CJ, Miller JP et al. (2006). Effect of con-
tivity among frontal cortical regions. Brain 125: 276–289. straint-induced movement therapy on upper extremity
Scholz J, Klein MC, Behrens TE et al. (2009). Training induces function 3 to 9 months after stroke: the EXCITE random-
changes in white-matter architecture. Nat Neurosci 12: ized clinical trial. JAMA 296: 2095–2104.
1370–1371. Wolpaw JR, McFarland DJ (2004). Control of a two-
Seol GH, Ziburkus J, Huang S et al. (2007). Neuromodulators dimensional movement signal by a noninvasive brain-
control the polarity of spike-timing-dependent synaptic computer interface in humans. Proc Natl Acad Sci U S A
plasticity. Neuron 55: 919–929. 101: 17849–17854.
Sharma N, Pomeroy VM, Baron JC (2006). Motor imagery: a Wolpaw JR, Tennissen AM (2001). Activity-dependent spinal
backdoor to the motor system after stroke? Stroke 37: cord plasticity in health and disease. Annu Rev Neurosci 24:
1941–1952. 807–843.
12 N. SHARMA ET AL.
Wrigley PJ, Gustin SM, Macey PM et al. (2009). Anatomical Wu CW, van Gelderen P, Hanakawa T et al. (2005). Enduring
changes in human motor cortex and motor pathways fol- representational plasticity after somatosensory stimulation.
lowing complete thoracic spinal cord injury. Cereb Cortex Neuroimage 27: 872–884.
19: 224–232. Ziemann U, Hallett M, Cohen LG (1998). Mechanisms of
Wu CW, Kaas JH (1999). Reorganization in primary motor deafferentation-induced plasticity in human motor cortex.
cortex of primates with long-standing therapeutic amputa- J Neurosci 18: 7000–7007.
tions. J Neurosci 19: 7679–7697.
Wu CW, Kaas JH (2000). Spinal cord atrophy and reorganiza-
tion of motoneuron connections following long-standing
limb loss in primates. Neuron 28: 967–978.

You might also like