You are on page 1of 8

Special Report

Brain Plasticity and Stroke Rehabilitation


The Willis Lecture
Barbro B. Johansson, MD, PhD

Abstract—Neuronal connections and cortical maps are continuously remodeled by our experience. Knowledge of the
potential capabilityof the brain to compensate for lesions is a prerequisite for optimal stroke rehabilitation strategies.
Experimental focal cortical lesions induce changes in adjacent cortex and in the contralateral hemisphere. Neuroimaging
studies in stroke patients indicate altered poststroke activation patterns, which suggest some functional reorganization.
To what extent functional imaging data correspond to outcome data needs to be evaluated. Reorganization may be the
principle process responsible for recovery of function after stroke, but what are the limits, and to what extent can
postischemic intervention facilitate such changes?
Postoperative housing of animals in an enriched environment can significantly enhance functional outcome and can
also interact with other interventions, including neocortical grafting. What role will neuronal progenitor cells play in
future rehabilitation—stimulated in situ or as neural replacement? And what is the future for blocking neural growth
inhibitory factors? Better knowledge of postischemic molecular and neurophysiological events, and close interaction
between basic and applied research, will hopefully enable us to design rehabilitation strategies based on neurobiological
principles in a not-too-distant future. (Stroke. 2000;31:223-230.)
Key Words: neuronal plasticity n recovery n rehabilitation n stroke

S ince regeneration of transectioned central axons has


never been convincingly demonstrated in higher mam-
mals, it seems in most instances that one must resort to the
cortical representation areas, cortical maps, can be modified
by sensory input, experience, and learning (Figure 1), as well
as in response to brain lesions.17–30 The potential relevance
assumption that intact fibers take over for the damaged for stroke rehabilitation of those data was proposed more than
Downloaded from http://ahajournals.org by on August 21, 2021

ones.”1 a decade ago.19 Transient alterations of cortical representation


The above words were written in 1973 by Alf Brodal, a areas may be common in everyday life, as indicated by
Norwegian neuroanatomist, based on his own experience transcranial magnetic stimulation studies during learning
after a stroke. To what extent has his assumption been shown tasks in human volunteers.31 If we regularly have to perform
to be correct? In this review I will present current concepts on a very skilled motor task, the cortical representation for the
brain plasticity in intact and lesioned brain, and evidence that muscles involved will remain enlarged, as seen for the fingers
postischemic interventions can alter molecular events and of the left but not the right hand in string players.32 Similarly,
influence functional recovery after brain lesions. the sensorimotor cortical representation of the reading finger
is expanded in blind Braille readers33 and, furthermore,
Current Concepts on Brain Plasticity
fluctuates with the reading activity pattern.34
That neuronal cortical connections can be remodeled by our
experience was suggested by Hebb half a century ago.2,3
Since then, many studies have demonstrated chemical and Possible Mechanisms Behind Brain Plasticity
anatomic plasticity in the cerebral cortex of adult animals.4 –16 Several mechanisms are likely to be involved in brain
Animals reared or housed as adults in complex environments plasticity.35 Activity-dependent modification of synaptic con-
with access to various toys and activities develop more nections and reorganization of adult cortical areas are thought
dendritic branching and more synapses per neuron and have to involve long-term potentiation (LTP) and long-term de-
higher gene expression for trophic factors than animals pression (LTD), mechanisms by which information is stored
housed individually or in small groups in standard cages.4 –12 in the mammalian central nervous system.36 –37 Synaptic
Similar changes can be induced during learning.13–16 plasticity in cortical horizontal connections has been pro-
Another aspect of brain plasticity, first and most exten- posed to underlie cortical map reorganization.38 – 40 Gluta-
sively demonstrated by Merzenich and coworkers, is that mate, the main excitatory neurotransmitter, plays a crucial

Presented as the Willis Lecture at the 24th American Heart Association International Conference on Stroke and Cerebral Circulation, Nashville, Tenn,
February 4, 1999. The opinions expressed in this article are not necessarily those of the editors or of the American Heart Association.
From the Division for Experimental Neurology, Wallenberg Neuroscience Center, University Hospital, Lund, Sweden.
Correspondence to Barbro B. Johansson, MD, PhD, Division for Experimental Neurology, Wallenberg Neuroscience Center, University Hospital,
S-221 85 Lund, Sweden. E-mail Barbro.Johansson@neurol.lu.se
© 2000 American Heart Association, Inc.
Stroke is available at http://www.strokeaha.org

223
224 Stroke January 2000

There is increasing evidence that astrocytes take an active


part in synaptic plasticity.49,50 Rapid astrocytic changes in
cortex and ultrastructural evidence for increased contact
between astrocytes and synapses in rats reared in a complex
environment suggest a close relationship between astrocytic
plasticity and experience-induced synaptic plasticity.51,52
Nonsynaptic transmission may also play a role in plasticity
processes.53 Plastic changes occur not only in the cortex but
have also been demonstrated in subcortical regions, including
thalamus and brain stem.54,55

Spontaneous Events and Training Effects


After a brain lesion, changes in other brain regions have been
documented at different postlesion times, from minutes to
Figure 1. The area or somatosensory cortex (black) in a mon- months.54,55 Postlesion events may be due to deafferentation,
key before (A) and after (B) controlled tactile stimulation. removal of inhibition, activity-dependent synaptic changes,
Reprinted with permission.21
changes in membrane excitability, growth of new connec-
role. Cortical map reorganization in the primary somatosen- tions, or unmasking of preexisting connections.27 Unmasking
sory cortex can be prevented by blockade of N-methyl-D- has generally been proposed to be responsible for rapid
aspartate (NMDA) receptors.41– 43 g-Aminobutyric acid changes in cortical maps,56 and there is evidence that synaptic
(GABA)-A receptor antagonists can facilitate LTP induction plasticity can be very rapid.48
in neocortical synaptic systems, and the induction can be Cortical mapping by intracellular recordings in primates
blocked by GABA-A receptor agonists.40 Transmitters re- has demonstrated that the tissue surrounding a small lesion in
leased by the diffuse neuromodulatory systems originating in the hand-representation area of primary motor cortex in adult
locus coeruleus (noradrenalin), nucleus basalis (acetylcho- monkeys undergoes a further territorial loss in the functional
line), lateral tegmentum (dopamine), and raphe nuclei (sero- representation of the affected body part, perhaps through
tonin) may modify the process.44,45 Nitric oxide is another nonuse or disruption of local intrinsic cortical circuitry.28 This
candidate for dynamic modulation of cerebral cortex synaptic further tissue loss could be prevented and functional reorga-
function.46 There is evidence that mechanisms involved in nization in the undamaged surrounding motor cortex stimu-
Downloaded from http://ahajournals.org by on August 21, 2021

synaptic plasticity varies between cortical regions.35,47 Local lated by retraining of hand use, starting 5 days after induction
neurotrophin actions, transmitter release, and synaptic protein of the lesion.29 Similarly, reorganization of primary somato-
synthesis are thought to promote synaptic remodeling and sensory cortex occurs in parallel with sensorimotor skill
changes in receptor expression or activation.12 As illustrated recovery in monkeys after restricted lesions. No significant
in Figure 2, dendritic spines, which receive the vast majority changes were recorded in the hand representation area in
of excitatory synaptic contacts in the mammalian brain, are somatosensory cortex in the opposite intact (untrained)
continuously being formed and modified.48 hemisphere.30

Figure 2. Confocal images of dendritic


spines of pyramidal neurons from
somatosensory cortex in an adult rat
housed together with 3 other rats in a
standard cage (left) or during 3 weeks
housed in an enriched environment
(right). Lucifer yellow, a fluorescent dye,
has been microinjected into the individ-
ual neurons (P. Belichenko, MD, PhD,
and B.B. Johansson, MD, PhD, unpub-
lished data, 1998).
Johansson Stroke and Brain Plasticity 225

Morphologic studies in the rat indicate that cortical lesions lin growth factor-1, transforming growth factor b1, and glial
can induce an increase of dendritic branching in the contralat- cell line– derived neurotrophic factor, have been reported to
eral hemisphere, with a maximum 2 to 3 weeks after the be beneficial in the early ischemic period. Whether some of
lesion.57 If the rats were prevented from using the intact these trophic factors can be beneficial in the rehabilitation
forelimb, both the morphologic changes and functional re- phase has not yet been evaluated. Because of the poor
covery were inhibited.58 Although some studies with other penetration of many growth factors into the brain, an inter-
techniques could not verify those findings,59,60 a detailed esting approach is to use substances that induce endogenous
electromicroscopic longitudinal study61 has confirmed the growth factor synthesis in the brain after peripheral adminis-
development of time-dependent morphologic changes with a tration. A b2-adrenoceptor agonist as been shown to reduce
significant increase in dendritic volume in cortical layer of infarct volume and induce an earlier and more pronounced
the contralateral motor cortex 18 days after the lesion, and in increase in mRNA of nerve growth factor (NGF), bFGF, and
the number of synapses per neuron 30 days after the lesion. transforming growth factor-b1 (TGF-b1) than was seen in
After a focal brain infarct, an increased density and untreated rats after permanent focal ischemia. 77
distribution of GAP 43 immunoreactivity has been observed Cholecystokinin-8 increases both NGF protein and NGF
in the ipsilateral cortex 3 to 14 days after the vascular mRNA in mouse cortex and hippocampus when injected
occlusion and of synaptophysin immunoreactivity in the same intraperitoneally at physiological doses and may thus repre-
areas at postoperative days 14 to 60. A larger distribution of sent a potential experimental model for investigating the
synaptophysin immunoreactivity was also noted in the con- effects of endogenous NGF upregulation after brain lesions.78
tralateral hemisphere.62 An increased neuronal labeling of Based on data on the role of BDNF for plasticity in the
MAP-2, GAP-43, and cyclin D1 immunoreactivity from day intact brain,79,80 we have studied the late postischemic gene
2 and up to 28 days has been found in the penumbra zone.63 expression for this protein in rats killed 2 to 30 days after the
ischemic event. Moreover, intraventricular BDNF has been
Enriched Environment and Neurotrophic Factors shown to rescue neurons in acute ischemia.81 Unexpectedly, a
There is substantial evidence that the postoperative environ-
secondary increase in BDNF gene expression observed in
ment can influence the outcome after experimental brain
control rats did not occur in rats housed in an enriched
damage, such as traumatic brain lesions, hippocampal sec-
environment, who had significantly lower BDNF expression
tioning, and cortical ablation.64,65 After an experimental brain
in ipsilateral and contralateral cortex than rats in a standard
infarction, rats housed in an enriched environment with the
environment 2 to 12 days after the insult.82 Similar results
opportunity for various activities and interaction with other
were obtained for expression of NGFI-A mRNA,83 a gene
Downloaded from http://ahajournals.org by on August 21, 2021

rats perform significantly better than rats housed in standard


that earlier had been shown to be activated in an enriched
laboratory environment,66 even when the transfer to an
enriched environment was delayed for 15 days.67 A compar- environment in intact rats.10 However, a late increase of
ison between enriched environment, social interaction, and NGFI-A mRNA expression was observed at 30 days after the
physical activity in the form of wheel-running indicated that lesion, which suggested that it might be important for later
social interaction was superior to wheel-running and that an postischemic events.83
enriched environment which allowed free physical activity The reason for the early dampening of the postischemic
combined with social interaction resulted in the best increase seen in control animals for BDNF is not clear. BDNF
performance.68 is related to synaptic activity. Cortical networks adjacent to a
We hypothesized that the beneficial effect of enriched focal brain ischemia are hyperexcitable because of an imbal-
environment might be caused by increased synthesis of ance between excitatory and inhibitory synaptic func-
neurotrophic factors. Neurotrophic factors are polypeptides tion.84 – 86 Hyperexcitability has been recorded not only
capable of promoting neuronal survival. Local neurotrophin around the infarct but also in the contralateral hemisphere
action may promote synaptic remodeling and changes in measured 1 week after the ischemic event.87– 89 Both a
receptor expression.12 Ischemia is a strong inducer of gene detrimental role (impaired processing of incoming informa-
expression in the brain.69 –71 More than 90 different genes tion) and a beneficial role (adaptation and favorable recovery)
have been shown to be acutely induced, generally with an of this hyperexcitability have been proposed.87 Could it be
early peak within minutes or hours of onset of ischemia and that rats in an enriched environment have a better balance
a rapid return to normal or subnormal levels. Whether the between inhibitory and excitatory transmission and that
early transient increase in gene expression during the initial depression of hyperexcitability might be beneficial in the
postischemic hours is related to outcome is not known. There early stage? Clearly, more studies are needed before any
is some evidence that trophic factors can rescue neurons in conclusions can be drawn. The relationship between growth
the acute stage. In addition to reducing infarct size even when inhibitory and growth promotor factors are likely to be
given hours after the ischemic insult, basic fibroblast growth important, and it should be noted also that growth inhibitory
factor (bFGF) may attenuate the thalamic degeneration fol- factor mRNA is increased during the postischemic period
lowing cortical infarction.72–74 Nerve growth factor (NGF) after focal ischemia in the rat.90 The recent reports on
has been reported to improve memory and motor functions increased corticofugal plasticity after neutralization of a
and reduce dendritic atrophy in the remaining pyramidal myelin-associated neurite growth inhibitor is a reminder that
neurons.75 As reviewed elsewhere,76 several other growth inhibitors of brain plasticity may be just as important as
factors, including brain-derived growth factor (BDNF), insu- stimulating factors for postischemic functional outcome.91,92
226 Stroke January 2000

Pharmacological Interventions Transplantation


The possible role of pharmacological interventions in the As reviewed elsewhere, implantation of fetal neocortical cells
postischemic rehabilitation phase has been extensively re- after cortical lesions has been performed successfully in
viewed by other authors.93–98 I will here restrict myself to a several laboratories.76 Transplanted cells can interact with the
few comments. Considering the many complex events that host tissue by forming connections but also by being a source
occur in postschemic brain, it is likely that the efficacy of a of trophic factors that can influence the surrounding tissue.
drug can vary with the postischemic time, size and type of Although both anatomic and functional integration with the
lesion, and interactions with other therapeutic interventions. host brain have been observed,114 improvement in behavioral
In a recent review, Schallert and Hernandez97 write: “depend- tests have been noticed only when transplantation is com-
ing on the site, extent and nature of the injury and secondary bined with posttransplantation housing in an enriched envi-
degenerative events, and the timing of drug administration, ronment.115,116 If performed 1 week after ligation of the
GABA agonists may have either negative or positive effects. middle cerebral artery, but not if delayed for 3 weeks, the
In many research reports, rather bold suggestions have been combined procedure can improve functional outcome more
offered about the potential clinical efficacy of GABAergic than an enriched environment alone, and it can also reduce
drugs without regards to these important variables”. Interac- the secondary thalamic atrophy that occurs after cortical
tions between drugs and environmental factors is another lesions. Although grafting can successfully be performed at
aspect. If combined with test-specific training, norepineph- later postischemic times, there is so far no evidence for
rine, amphetamine, and other a-adrenergic stimulating drugs functional improvement at later times.
can enhance motor performance after unilateral ablation of Immortalized embryonic neuronal cells lines or cultured
sensorimotor cortex95,99 and have also been shown to enhance neural multipotent progenitor cells (the transition from stem
the immunoreactivity to synaptic proteins after focal brain cell to progenitor cell is not sharp) implanted into a lesioned
ischemia.100 However, amphetamine had no additional effect brain have shown promising results in other experimental
on outcome in rats housed in an enriched environment after models. When implanted into the neocortex of adult mice
focal brain ischemia,101 perhaps due to enhanced noradrener- undergoing targeted apoptosis of neocortical pyramidal neu-
gic release under such housing conditions. rons, they migrate long distances into the regions of cell
death, where they differentiate and make appropriate long-
What Is the Possible Role of Neurogenesis? distance projections.117–119 The results indicate the presence
Neural stem cells, multipotential cells that are precursors to of environmental signals that promote differentiation of the
neurons and glia, have been identified in the adult vertebrate implanted cells. Furthermore, adult mature astrocytes in the
Downloaded from http://ahajournals.org by on August 21, 2021

central nervous system. Although first reported to be present host brain have been shown to retain the capacity to transform
in brains from rodents in the sixties,102,103 it is only during the into developmental radial glia that may help the active
last years that they have been extensively studied.104,105 They migration of transplanted neural precursors.120 Whether trans-
are predominantly found in the periventricular ependymal or plantation of neuronal precursor cells to neocortical infarct
subependymal zone and in the dentate gyrus but may also be cavities would be a good substitute for the currently used fetal
present in small numbers in other regions.106 Stem cells from neocortical tissue block techniques remains to be
the adult brain proliferate and differentiate into neurons and demonstrated.
glia in tissue culture with the same efficiency for neuronal
differentiation as found in fetal stem cells. That stem cells are Clinical Evidence for Reorganization of
present also in human brains was first shown in tissue culture Cortical Networks
from the subependymal zone and periventricular white mat- Studies using PET, functional MRI (fMRI), transcranial
ter.107 Recent studies have shown that they can differentiate stimulation, and magnetoencephalography (MEG) support
to neurons in the adult human dentate gyrus in vivo.108 With the concept of functional reorganization after stroke.121–130
the observation that such cells in experimental studies can be PET studies on blood flow distribution during finger move-
manipulated in vivo by growth factors and by environmental ments in a previously paretic hand have demonstrated com-
enrichment,109 –111 the clinical potential of in vivo manipula- plex patterns of activation, with increased activity with large
tion of stem cells in humans is currently subject to much individual variations.121,122 Until now, studies comparing the
speculation.104 –106 degree and pattern of activation in patients with good and
The aspects of neurogenesis that are relevant for this less-than-good recovery and with specific therapeutic inter-
review are the following: (1) Does neurogenesis increase in ventions are few, and the published data are sometimes
response to brain lesions? (2) If so, is neurogenesis related to contradictory. Because of large individual variations, careful
outcome? (3) Can those events be influenced by postlesion longitudinal studies of individual patients with specific defi-
interventions? and (4) Can stem cells or progenitor cells be cits and well-defined lesions are needed. Individuals may use
used for transplantation after stroke? The first question can be different compensation strategies before and after training,128
answered affirmatively. It has been shown to occur in and the activation pattern can change with time. It has been
excitotoxic and mechanical lesions in the dentate gyrus reported that changes in activation pattern can be induced by
(hippocampus) in the adult rat112 and after transient global forced training of the paretic hand even 4 to 15 years after
ischemia in gerbils.113 The second and third questions remain stroke onset.131 The knowledge that the degree of cortical
to be answered, and the fourth question will be discussed in lateralization and interhemispheric interaction varies for spe-
the transplantation section below. cific language components in the normal human brain may be
Johansson Stroke and Brain Plasticity 227

relevant for the interpretation of data on aphasic function with increased NMDA receptor-mediated excitation
individuals.132 and decreased efficacy of GABAergic inhibition.84 – 86 In the
presence of excitatory and toxic substances from the ischemic
Stroke Units and Early Training tissue, an additional release of excitatory substances induced
It is well established that early mobilization can reduce by motor activity may be harmful in the early postischemic
secondary thromboembolic events, pneumonia, and mortality stage. Consistent with this hypothesis is the observation that
in acute stroke.133–135 It is now recommended that stroke the NMDA receptor antagonist MK-801can prevent second-
patients be admitted to specialized stroke units with specially ary cortical damage in rats forced to use their impaired limb
trained medical and nursing staff, coordinated multidisci- after a lesion in the sensorimotor cortex.143
plinary rehabilitation, and education programs for patients It is important to define the window for a possible
and their families. Stroke unit care has been shown to be increased vulnerability and additional peri-infarct neuronal
associated with a long-term reduction of death and of the loss. However, I do not think these animal data should make
combined poor outcomes of death and dependency, effects us change the policy of early mobilization of stroke patients.
that were independent of patient age, sex, or variations in Housing animals in an enriched environment, which may
stroke unit organization. No study has shown to what extent correspond to early mobilization, has no aggravating effects.
the beneficial effect is due to specific rehabilitation strategies, The most important tasks in the early stage are to prevent
to the daily time spent in physiotherapy and occupational complications and train the patient to regain balance and body
therapy, or is a nonspecific effect of a more stimulating symmetry. In addition, cortical and lacunar infarcts may
environment with competent staff that can encourage and differ.
support the patients and family members. Scientific evidence
demonstrating the values of specific rehabilitation interven- Age and Plasticity
tions after stroke is limited. Comparisons between different Age influences the impact of vascular occlusion in ro-
methods in current use have so far failed to show that any dents,144 –146 but does it also influence functional recovery? In
particular physiotherapy or stroke rehabilitation strategy is a comparison between 3- and 20-month-old rats, upregulation
superior to another. There is some evidence that forced use of of MAP1B and MAP2 was diminished but not abolished in
a paretic arm may improve function in the chronic the aged compared with the young animals.147 It might not be
stage.121,136 –138 fair to compare humans with rats living their entire lives in a
Clinical data are thus strongly in favor of early mobiliza- laboratory with little stimulation. The decrease in synaptic
tion and training. On the other hand, there are some disturb- density in aged laboratory rats can be prevented by rearing in
Downloaded from http://ahajournals.org by on August 21, 2021

ing animal data indicating that overtraining of the lesioned an enriched environment.148 The loss of neurons in aging
forelimb induced by immobilization of the intact forelimb can humans is to some extent compensated for by selective
expand cortical lesions.139,140 Referring to those studies, dendritic growth. A postmortem comparison of dendrites of
Nudo et al29 started training monkeys 5 days after the lesion. layer II pyramidal neurons in the parahippocampal gyrus of
Housing animals in an enriched environment with the oppor- adult (aged 51 years) and neurologically healthy aged (80
tunity to perform various activities but no specific training years) individuals showed that the dendritic trees were longer
significantly improves functional outcome without increasing and more branched in the healthy 80-year-olds.149 In a
tissue loss. However, if combined with more specific training population of persons aged 65 years and older, the level of
from 24 hours after the insult, an increased tissue loss was cognitive function is positively related to the frequency and
observed.141 Despite the larger tissue loss in the early training intensity of cognitive activity.150 However, it has been shown
group, the rats improved more than standard rats, confirming that a head injury in young adulthood, although well com-
earlier data of poor correlation between infarct volume and
pensated for at the time, exacerbates cognitive decline in later
functional outcome in rats housed in an enriched environ-
years.151 This might be relevant for individuals with repeated
ment. The better outcome in the early training group than in
stroke.
standard rats may be related to compensatory adaptation in
the contralateral hemisphere, subcortical region, or Concluding Remarks
cerebellum. The current trend to equate tissue loss with outcome is not
Even if the increased tissue loss did not correspond to relevant when it comes to postischemic interventions, which
unfavorable outcome, it is clearly an unwanted effect, one can improve outcome without a change in infarct volume.
which might make the brain more vulnerable to additional That environmental stimulation improves recovery is not a
insults or aging. What could be the course of this vulnerable new observation,152 but it is worthwhile to point out that it can
early postischemic period? Motor activity stimulates the enhance the effect of other therapies, as shown with neocor-
release of glutamate and catecholamines.142 One possible tical grafting.66 – 68,115 It is not only the number of neurons left,
explanation for the increased tissue loss might be that but how they function and what connections they can make
hyperexcitability of the surrounding tissue in the early post- that will decide functional outcome.
ischemic period makes the surrounding neurons vulnerable to
excitation. As discussed above in the section on enriched Acknowledgments
environment and neurotrophic factors, cortical networks ad- Studies from the author’s laboratory were supported by grants from
jacent to a focal brain ischemia are hyperexcitable because of the Swedish Medical Research Council (Project 14X-4968), the
an imbalance between excitatory and inhibitory synaptic Bank of Sweden Tercentenary Foundation, the Swedish Heart and
228 Stroke January 2000

Lung Foundation, the Swedish Association of Neurologically Dis- 27. Hallett M. Plasticity in the human motor system. Neuroscientist. 1999;5:
abled, and the Swedish Stroke Foundation. 324–332.
28. Nudo RJ, Milliken GW. Reorganization of movement representations in
primary motor cortex following focal ischemic infarcts in adult squirrel
References monkeys. J Neurophysiol. 1996;75:2144–2149.
1. Brodal A. Self-observations and neuro-anatomical considerations after a 29. Nudo RJ, Wise BM, SiFuentes F, Milliken GW. Neural substrates for the
stroke. Brain. 1973;96:675– 694. effects of rehabilitative training on motor recovery after ischemic infarct.
2. Hebb DO. The effects of early experience on problem solving at maturity. Science. 1996;272:1791–1794.
Am Psychol. 1947;2:737–745. 30. Xerri C, Merzenich MM, Peterson BE, Jenkins W. Plasticity of primary
3. Hebbs DG. The Organization of Behavior. New York, NY: John Wiley & somatosensory cortex paralleling sensorimotor skill recovery from stroke in
Sons Inc; 1949. adult monkeys. J Neurophysiol. 1998;79:2119–2148.
4. Bennett EL, Diamond MC, Krech D, Rosenzweig MR. Chemical and 31. Pascual-Leone A, Grafman J, Hallett M. Modulation of cortical motor
anatomical plasticity of brain. Science. 1964;146:610–619. output maps during development of implicit and explicit knowledge.
5. Rosenzweig MR. Environmental complexity, cerebral change, and Science. 1994;263:1287–1289.
behavior. Am Psychol. 1966;21:321–332. 32. Elbert T, Pantev C, Wienbruch C, Rockstroh B, Taub E. Increased cortical
6. Holloway RL. Dendritic branching: some preliminary results of training and representation of the fingers of the left hand in string players. Science.
complexity in rat visual cortex. Brain Res. 1966;2:393–396. 1995;270:305–307.
7. Volkmar FR, Greenough WT. Rearing complexity affects branching of 33. Pascual-Leone A, Torres F. Plasticity of the sensorimotor cortex repre-
dendrites in the visual cortex of the rat. Science. 1972;176:1445–1447. sentation of the reading finger in Braille readers. Brain. 1993;116:39–52.
8. Turner AM, Greenough WT. Differential rearing effect on rat visual cortex 34. Pascual-Leone A, Wassermann EM, Sadato N, Hallett M. The role of
synapses, I: synaptic and neuronal density and synapses per neuron. Brain reading activity on the modulation of motor cortical outputs to the reading
Res. 1985;329:195–203. hand in Braille readers. Ann Neurol. 1995;38:910–915.
9. Torasdotter M, Metsis M, Henriksson BF, Winblad B, Mohammet AH, 35. Shaw CA, Lanius RA, van den Doel K. The origin of synaptic neuroplas-
Donaldson LF. Environmental enrichment results in higher levels of nerve ticity: crucial molecules or a dynamic cascade? Brain Res Rev. 1994;19:
growth factor mRNA in the rat visual cortex and hippocampus. Behav Brain 241–263.
Res. 1992;51:179–183. 36. Bear MF, Malenka RC. Synaptic plasticity: LTP and LTD. Curr Opin
10. Olsson T, Mohammet AH, Donaldson LF, Henriksson BF, Seckl JR. Glu- Neurobiol. 1994;4:389–399.
cocorticoid receptor and NGFI-A gene expression are induced in the hip- 37. Buonomano DV, Merzenich MM. Cortical plasticity: from synapses to
pocampus after environmental enrichment in adult rats. Molec Brain Res. maps. Annu Rev Neurosci. 1998;21:149–186.
1994;23:349–353. 38. Hess G, Donoghue JP. Long-term potentiation of horizontal connections
11. Kolb B. Brain Plasticity and Behaviour. Hillsdale, NJ: Lawrence Erlboum; provides a mechanism to reorganize cortical motor maps. J Neurophysiol.
1995. 1994;71:2543–2547.
12. Klintsova AY, Greenough WT. Synaptic plasticity in cortical systems. Curr 39. Das A, Gilbert CD. Long-range horizontal connections and their role in
Opin Neurobiol. 1999;9:203–208. cortical reorganization revealed by optical recording of cat primary visual
13. Greenough WT, Larson JR, Withers GS. Effect of unilateral and bilateral cortex. Nature. 1995;375:780–784.
training in a reaching task on dendritic branching of neurons in the rat 40. Hess G, Aizenman CD, Donoghue JP. Conditions for the induction of
motor-sensory forelimb cortex. Behav Neurol Biol. 1985;44:301–314. long-term potentiation in Layer II/III horizontal connections of the rat motor
Downloaded from http://ahajournals.org by on August 21, 2021

14. Van Reemts J, Dikova M, Werbrouck L, Clincke G, Borgers M. Synaptic cortex. J Neurophysiol. 1996;75:1765–1777.
plasticity in rat hipppocampus associated with learning. Behav Brain Res. 41. Kano M, Lino K, Kano M. Functional reorganization of adult cat somato-
1992;51:179–183. sensory cortex is dependent on NMDA receptors. Neuroreport. 1991;2:
15. Neeper SA, Gomez-Pinilla F. Choi J, Cotman C. Exercise and brain neu- 77–80.
rotrophins. Nature. 1995;373:109. Letter. 42. Hess G, Jacobs KM, Donoghue JP. N-Methyl-D-aspartate receptor mediated
16. Kleim JA, Lussnig E, Schwarz ER, Comery TA, GreenoughWT. Synapto- component of field potentials evoked in horizontal pathways of rat motor
genesis and Fos expession in the motor cortex of the adult rat after motor cortex. Neuroscience. 1994;61:225–235.
43. Garraghty PE, Muja N. NMDA receptors and plasticity in adult primate
skill learning. J Neurosci. 1996;16:4529–4535.
somatosensory cortex. J Comp Neurol. 1996;367:319–326.
17. Merzenich MM, Kaas JH, Wall JT, Nelson RJ, Sur M, Felleman D. Topo-
44. Kilgard MP, Merzenich MM. Cortical map reorganization enabled by
graphic reorganization of somatosensory cortical areas 3b and 1 in adult
nucleus basalis activity. Science. 1998;279:1714–1718.
monkeys following restricted deafferentation. Neuroscience. 1983;8:33–55.
45. Kirkwood A, Rozas C, Kirkwood J, Perez F, Bear MF. Modulation of
18. Merzenich MM, Nelson RJ, Stryker MP, Cynader MS, Schoppmann A,
long-term synaptic depression in visual cortex by acetylcholine and norepi-
Zook JM. Somatosensory cortical map changes following digit amputation
nephrine. J Neurosci. 1999;19:1599–1609.
in adult monkeys. J Comp Neurol. 1984;224:591–605.
46. Kara P, Friedlander MJ. Dynamic modulation of cerebral cortex synaptic
19. Jenkins WM, Merzenich MM. Reorganization of neocortical representations
function by nitric oxide. Prog Brain Res. 1998;118:183–198.
after brain injury: a neurophysiological model of the bases of recovery from
47. Castro-Alamancos MA, Donoghue JP, Connors BW. Different forms of
stroke. Progr Brain Res. 1987;71:249–266. synaptic plasticity in somatosensory and motor areas of the neocortex.
20. Pons TP, Garraghty PE, Mishkin M. Lesion-induced plasticity in the J Neurosci. 1995;15:5324–5333.
somatosensory cortex of adult macaques. Proc Nat Acad Sci U S A. 1988; 48. Fischer M, Kaech S, Knutti D, Matus A. Rapid actin-based plasticity in
85:5279–5281. dendritic spines. Neuron. 1998;20:847–854.
21. Jenkins WM, Merzenich MM, Ochs MT, Allard R, Guic-Robles E. Func- 49. Vernadakis A. Glia-neuron intercommunications and synaptic plasticity.
tional reorganization of primary somatosensory cortex in adult owl monkeys Prog Neurobiol. 1996;49:185–214.
after behaviorally controlled tactile stimulation. J Neurophysiol. 1990;68: 50. Pfrieger FW, Barres BA. New views on synapse-glia interactions. Curr
82–104. Opin Neurobiol. 1996;6:615–621.
22. Kaas JH. Plasticity of sensory and motor maps in adult mammals. Ann Rev 51. Jones TA, Hawrylak N, Greenough WT. Rapid laminal-dependent changes
Neurosci. 1991;14:137–167. in GFAP immunoreactive astrocytes in the visual cortex of rats reared in a
23. Liepert J, Tegenthoff M, Malin JP. Changes of cortical motor area size complex environment. Psychoneuroendocrinology. 1996;21:189–201.
during immobilization. Electroencephalogr Clin Neurophysiol. 1995;97: 52. Jones TA, Greenough WT. Ultrastructural evidence for increased contact
382–386. between astrocytes and synapses in rats reared in a complex environment.
24. Seitz RJ, Huang Y, Knorr U, Tellmann L, Herzog H, Freund HJ. Neurobiol Learn Mem. 1996;65:48–56.
Large-scale plasticity of the human motor cortex. Neuroreport. 1995;6: 53. Bach-y-Rita P. Nonsynaptic diffusion neurotransmission (NDN) in the
742–744. brain. Neurochem Int. 1993;23:297–318.
25. Schieber MH. Physiological basis for functional recovery. J Neurol Rehabil. 54. Nicolelis MAL. Dynamic and distributed somatosensory representations as
1995;9:65–71. the substance for cortical and subcortical plasticity. Semin Neurosci. 1997;
26. Florence SL, Taub HB, Kaas JH. Large-scale sprouting of cortical con- 9:24–33.
nections after peripheral injury in adult macaque monkeys. Science. 1998; 55. Jones EG, Pons TP. Thalamic and brain stem contributions to large-scale
282:1117–1121. plasticity of primate somatosensory cortex. Science. 1998;282:1121–1125.
Johansson Stroke and Brain Plasticity 229

56. Jacobs KM, Donoghue JP. Reshaping the cortical motor map by unmasking 81. Schäbitz W-R, Schwab S, Spranger M, Hacke W. Intraventricular brain
latent intracortical connections. Science. 1991;251:944–947. derived neurotrophic factor reduces infarct size after focal cerebral ischemia
57. Jones TA, Schallert T. Overgrowth and pruning of dendrites in adult rats in rats. J Cereb Blood Flow Metab. 1997;17:500–506.
recovering from neocortical damage. Brain Res. 1992;581:156–160. 82. Johansson BB, Zhao L-R, Mattsson B. Environmental influence on neuro-
58. Jones TA, Schallert T. Use-dependent growth of pyramidal neurons after trophic gene expression after experimental brain infarction in the rat. In: Ito
neocortical damage. J Neurosci. 1994;14:2140–2152. U, Fieschi C, Orzi F, Kuroiwa T, Klatzo I, eds. Maturation Phenomenon in
59. Forgie ML, Gibb R, Kolb B. Unilateral lesions of the forelimb area of rat Cerebral Ischemia III. Berlin, Germany: Springer-Verlag; 1999:261–266.
motor cortex: lack of evidence for use-dependent neural growth in the 83. Dahlqvist P, Zhao L, Johansson I-M, Mattsson B, Johansson BB, Seckl JR,
undamaged hemisphere. Brain Res. 1996;710:249–259. Olsson T. Environmental enrichment alters NGFI-A and glucocorticoid
60. Prusky G, Whishaw IQ. Morphology of identified corticospinal cells in the receptor mRNA expression after MCA occlusion in rats. Neuroscience.
rat following motor cortex injury: absence of use-dependent change. Brain 1999;93:527–535.
Res. 1996;714:1–8. 84. Schiene K, Bruehl C, Zilles K, Qü MS, Hagemann G, Kraemer M, Witte
61. Jones TA, Kleim JA, Greenough WT. Synaptogenesis and dendritic growth OW. Neuronal hyperexcitability and reduction of GABAA-receptor
in the cortex opposite unilateral sensorimotor cortex damage in adult rats: a expression in the surround of cerebral photothrombosis. J Cereb Blood Flow
quantitative electron microscopic examination. Brain Res. 1996;733: Metab. 1996;16:906–914.
142–148. 85. Mittmann T, Qu M, Zilles K, Luhmann HJ. Long-term cellular dysfunction
62. Stroemer RP, Kent TA, Hulsebosch CR. Neocortical neural sprouting, after focal cerebral ischemia: in vitro analyses. Neuroscience. 1998;85:
synaptogenesis, and behavioral recovery after neocortical infarction in rats. 15–27.
Stroke. 1995;26:2135–2144. 86. Qu M, Mittmann T, Luhmann HJ, Schleicher A. Long-term changes of
63. Li Y, Jiang N, Powers C, Chopp M. Neuronal damage and plasticity ionotropic glutamate and GABA receptors after unilateral permanent focal
identified by microtubule-associated protein 2, growth-associated protein cerebral ischemia in the mouse brain. Neuroscience. 1998;85:29–43.
43, and cyclin D1 immunoreactivity after focal cerebral ischemia in rats. 87. Buchkremer-Ratzmann I, August M, Hagemann G, Witte OW. Electrophys-
Stroke. 1998;29:1972–1981. iological transcortical diaschisis after cortical photothrombosis in rat brain.
64. Will B, Kelche C. Environmental approaches to recovery of function from Stroke. 1996;27:105–1111.
brain damage: a review of animal studies (1981–1991). In: Rose FD, 88. Reinecke S, Lutzenburg M, Hagemann G, Bruehl C, Neumann-Haefelin T,
Johnson DA, eds. Recovery From Brain Damage. New York, NY: Plenum Witte OW. Electrophysiological transcortical diaschisis after middle
Publishing Corp; 1992:79–103. cerebral artery occlusion (MCAO) in rats. Neurosci Lett. 1999;261:85–88.
65. Held JM, Gordon J, Gentile AM. Environmental influences on locomotor 89. Qu M, Buchkremer-Ratzmann I, Schiene K, Schroeter M, Witte OW, Zilles
recovery following cortical lesions in rats. Behav Neurosci. 1985;99: K. Bihemispheric reduction of GABAA receptor binding following focal
678–690. cortical photothrombotic lesions in the rat brain. Brain Res. 1998;813:
66. Ohlsson A-L, Johansson BB. Environment influences functional outcome of 374–380.
cerebral infarction in rats. Stroke. 1995;26:644–649. 90. Yuguchi T, Kohmura E, Sakaki T, Nonaka M, Yamada K, Yaahita T,
67. Johansson BB. Functional outcome in rats transferred to an enriched envi- Yamashita T, Kishiguchi T, Sakaguchi T, Hayakawa T. Expression of
ronment 15 days after focal brain ischemia. Stroke. 1996;27:324–326. growth inhibitory factor mRNA after focal ischemia in rat brain. J Cereb
68. Johansson BB, Ohlsson A-L. Environment, social interaction, and physical Blood Flow Metab. 1997;17:745–752.
activity as determinants of functional outcome after cerebral infarction in the 91. Kartje GL, Schultz MK, Lopez-Yunez A, Schnell L, Schwab ME. Cortico-
Downloaded from http://ahajournals.org by on August 21, 2021

rat. Exp Neurol. 1996;139:322–327. striatal plasticity is restricted by myelin-associated neurite growth inhibitors
69. Kinouchi H, Sharp FR, Chan PH, Koistinaho J, Sagar SM, Yoshimoto T. in the adult rat. Ann Neurol. 1999;45:778–86.
Induction of c-fos, junB, c-jun, and hsp70 mRNA in cortex, thalamus, basal 92. Wenk CA, Thallmair M, Kartje GL, Schwab ME. Increased corticofugal
ganglia, and hippocampus following middle cerebral artery occlusion. plasticity after unilateral cortical lesions combined with neutralization of the
J Cereb Blood Flow Metab. 1994;14:808–817. IN-1 antigen in adult rats. J Comp Neurol. 1999;410:143–157.
70. Akins PT, Liu PK, Hsu CY. Immediate early gene expression in response to 93. Goldstein LB. Basic and clinical studies on pharmacologic effects on
cerebral ischemia: friend or foe? Stroke. 1996;27:1682–1687. recovery from brain injury. J Neurol Transplant Plast. 1993;4:175–192.
71. Koistinaho J, Hökfelt T. Altered gene expression in brain ischemia. Neu- 94. Goldstein LB. Potential effects of common drugs on stroke recovery. Arch
roreport. 1997;8:i-viii. Neurol. 1998;55:454–456.
72. Yamada K, Kinoshita A, Kohmura E, Sakaguchi T, Taguchi J, Kataoka K. 95. Feeney DM. Mechanisms of noradrenergic modulation of physical therapy:
Basic fibroblast growth factor prevents thalamic degeneration after cortical effects on functional recovery after cortical injury. In: Goldstein LB, ed.
infarction. J Cereb Blood Flow Metab. 1991;11:472–478. Restorative Neurology: Advances in Pharmacotherapy for Recovery After
73. Lin DA, Finklestein SP. Basic fibroblast growth factor: a treatment for Stroke. Armonk, NY: Futura Publishing Co; 1998:35–78.
stroke? Neuroscientist. 1997;3:247–250. 96. Saponjic RM, Hoane MR, Barth TM. Acetylcholine and recovery of
74. Kawamata T, Dietrich WD, Schallert T, Gotts JE, Cocke RR, Beowitz LI, function following brain injury. In: Goldstein LB, ed. Restorative Neu-
Finkelstein SP. Intracisternal basic fibroblast growth factor enhanced func- rology: Advances in Pharmacotherapy for Recovery After Stroke. Armonk,
tional recovery and upregulates the expression of a molecular marker of NY: Futura Publishing Co; 1998:79–89.
neuronal sprouting following focal cerebral infarction. Proc Natl Acad Sci 97. Schallert T, Hernandez TD. GABAergic drugs and neuroplasticity after
U S A. 1997;94:8179– 8184. brain injury: impact on functional recovery. In: Goldstein LB, ed.
75. Kolb B, Cote S, Ribeiro da Silva A, Cuello AC. Nerve growth factor Restorative Neurology: Advances in Pharmacotherapy for Recovery After
treatment prevents dendritic atrophy and promotes recovery of function after Stroke. Armonk, NY: Futura Publishing Co; 1998:91–120.
cortical injury. Neuroscience. 1997;76:1139–1151. 98. Barth TM, Hoane MR, Barbay S. Effect of glutamate antagonists on the
76. Johansson BB. Neurotrophic factors and transplants. In: Goldstein LB, ed. recovery and maintenance of behavioral functions afterbrain injury. In:
Restorative Neurology: Advances in Pharmacotherapy for Recovery After Goldstein LB, ed. Restorative Neurology: Advances in Pharmacotherapy
Stroke. Armonk, NY: Futura Publishing Co; 1998:141–166. for Recovery After Stroke. Armonk, NY: Futura Publishing Co; 1998:
77. Culmsee C, Stumm RK, Schafer MK, Weihe E, Krieglstein J. Clenbuterol 91–120.
induces growth factor mRNA, activates astrocytes, and protects rat brain 99. Feeney DM, Gonzalez A, Law WA. Amphetamine, haloperidol and expe-
tissue against ischemic damage. Eur J Pharmacol. 1999;379:33–45. rience interact to affect the rate of recovery after motor cortex injuries.
78. Tirassa P, Aloe L, Stenfors C, Turrini P, Lundeberg T. Cholecystokinin-8 Science. 1982;217:855–857.
protects central cholinergic neurons against fimbria-fornix lesions through 100. Stroemer RP, Kent TA, Hulsebosch CR. Enhanced neocortical neural
the upregulation of nerve growth factor synthesis. Proc Natl Acad Sci U S A. sprouting, synaptogenesis, and behavioral recovery with d-amphetamine
1999;96:6473–6477. therapy after neocortical infarction in rats. Stroke. 1998;29:2381–2395.
79. Castrén E, Zafra F, Thoenen H, Lindholm D. Light regulated expression of 101. Johansson BB, Mattsson B, Ohlsson A-L. Functional outcome after brain
brain-derived neurotrophic factor mRNA in rat visual cortex. Proc Natl infarction: effect of enriched environment and amphetamine. In: Ito U,
Acad Sci U S A. 1992;89:9444–9448. Kirino T, Kuroiwa T, Klatzo I, eds. Maturation Phenomenon in Cerebral
80. Rocamora N, Welker E, Pascual M, Soriano E. Upregulation of BDNF Ischemia II. Berlin, Germany: Springer-Verlag; 1997:159–167.
mRNA expression in the barrel cortex of adult mice after sensory stimu- 102. Altman J, Das GD. Autoradiographic and histological evidence of postnatal
lation. J Neurosci. 1996;16:4411–4419. hippocampal neurogenesis in rats. J Comp Neurol. 1965;124:319–335.
230 Stroke January 2000

103. Altman J, Das GD. Postnatal neurogenesis in the guinea-pig. Nature. 1967; 128. Small SL, Flores DK, Noll DC. Different neural circuits subserve reading
214:1098–1101. before and after therapy for acquired dyslexia. Brain Lang. 1998;62:
104. McKay R. Stem cells in the central nervus system. Science. 1997;276: 298–308.
66–71. 129. Heiss WD, Kessler J, Thiel A, Ghaemi M, Karbe H. Differential capacity of
105. Snyder EY. Neural stem-like cells: developmental lessons with therapeutic left and right hemispheric areas for compensation of poststroke aphasia. Ann
potential. The Neuroscientist. 1998;4:408–425. Neurol. 1999;45:430–438.
106. Palmer TD, Markakis EA, Willhoite AR, Safar F, Gage FH. Fibroblast 130. Musso M, Weiller C, Kiebel S, Muller SP, Bulau P, Rinjtjes M. Training-
growth factor-2 activates a latent neurogenic program in neural stem cells induced brain plasticity. Brain. 1999;122:1781–1790.
from diverse regions of the adult CNS. J Neurosci. 1999;19:8487–8497. 131. Kopp B, Kunkel A, Muhlnickel W, Villringer K, Taub E, Flow H. Plasticity
107. Kirschenbaum B. Nedergaard M, Preuss A, Barami I, Fraser RA, Goldman in the motor system related to therapy-induced improvement of movement
SA. In vitro neuronal production and differentiation by precursor cells after stroke. Neuroreport. 1999;10:807–810.
derived from the adult human forebrain. Cereb Cortex. 1994;6:576–589. 132. Pulvermüller F, Mohr B. The concept of transcortical cell assemblies: a key
108. Eriksson PS, Perfilieva E, Björk-Eriksson T, Alborn A-M, Nordborg C, to the understanding of cortical lateralization and interhemispheric inter-
Peterson DA, Gage FH. Neurogenesis in the adult human hippocampus. Nat action. Neurosci Biobehav Rev. 1996;20:557–566.
Med. 1998;4:1313–1317. 133. Stroke Unit Trialists’ Collaboration. Collaborative systematic review of the
109. Kempermann G, Kuhn HG, Gage FH. More hippocampal neurons in adult randomised trials of organised in-patient (stroke unit) care after stroke. BMJ.
mice living in an enriched environment. Nature. 1997;386:493–495. 1997;314:1151–1159.
110. Kempermann G, Kuhn HG, Gage FH. Experience-dependent neurogenesis 134. Indredavik B, Slørdahl SA, Bakke F, Rokseth R, Håheim LL. Stroke unit
in the senescent dentate gyrus. J Neurosci. 1998;18:3206–3212. treatment: long-term effects. Stroke. 1997;28:1861–1866.
111. Nilsson M, Perfilieva E, Johansson U, Orwar O, Eriksson PS. Enriched 135. Stroke Unit Trialists’ Collaboration. How do stroke units improve patient
environment increases neurogenesis in the adult rat dentate gyrus and outcomes? A collaborative systematic review of the randomized trials.
improves spatial memory. J Neurobiol. 1999;39:569–578. Stroke. 1997;28:2139–2144.
112. Gould E, Tanapat P. Lesion-induced proliferation of neuronal progenitors in 136. Wolf SL, Lecraw DE, Barton LA, Jann BB. Forced use of hemiplegic upper
the dentate gyrus of the adult rat. Neuroscience. 1997;80:427–436. extremities to reverse the effect of learned nonuse among chronic stroke and
113. Liu J, Solway K, Messing RO, Sharp FR. Increased neurogenesis in the head-injured patients. Exp Neurol. 1989;104:125–132.
dentate gyrus after transient global ischemia in gerbils. J Neurosci. 1998; 137. Taub E, Miller NE, Novack TA, Cool EW, Fleming WC, Nepomuceno CS,
18:7768–7778. Connell JS, Crago JE. Technique to improve chronic motor deficit after
114. Grabowski M, Brundin P, Johansson BB. Functional integration of cortical stroke. Arch Phys Med Rehabil. 1993;74:347–354.
138. Kunkel A, Kopp B, Muller G, Villringer K, Villringer A, Taub E, Flor H.
grafts placed in brain infarcts of rats. Ann Neurol. 1993;34:362–368.
Constraint-induced movement therapy for motor recovery in chronic stroke
115. Grabowski M, Sørensen JC, Mattsson B, Zimmer J, Johansson BB.
patients. Arch Phys Med Rehabil. 1999;80:624–628.
Influence of an enriched environment and cortical grafting in functional
139. Kozlowski DA, James DC, Schallert T. Use-dependent exaggeration of
outcome in brain infarcts of adult rats. Exp Neurol. 1995;133:96–102.
neuronal injury after unilateral sensorimotor cortex lesions. J Neurosci.
116. Mattsson B, Sørensen JC, Zimmer J, Johansson BB. Neural grafting to
1996;16:4776–4786.
experimental neocortical infarcts improves behavioral outcome and reduces
140. Humm JL, Kozlowski DA, James DC, Gotts JE, Schallert T. Use-dependent
thalamic atrophy in rats housed in enriched but not in standard envi-
exacerbation of brain damage occurs during an early post-lesion vulnerable
ronments. Stroke. 1997;28:1225–1232.
period. Brain Res. 1998;783:286–292.
117. Macklis JD. Transplanted neocortical neurons migrate selectively into
141. Risedal A, Zeng J, Johansson BB. Early training may exacerbate brain
Downloaded from http://ahajournals.org by on August 21, 2021

regions of neuronal degeneration produced by chromophore-targeted laser


damage after focal brain ischemia in the rat. J Cereb Blood Flow Metab.
photolysis. J Neurosci. 1993;13:3848–3863.
1999;19:997–1003.
118. Hernit-Grant CS, Macklis JD. Embryonic neurons transplanted to regions of 142. Vanderwolf CH, Cain DP. The behavioral neurobiology of learning and
targeted photolytic cell death in adult mouse somatosensory cortex re-form memory: a conceptual reorientation. Brain Res Brain Res Rev. 1994;19:
specific callosal projections. Exp Neurol. 1996;139:131–142. 264–297.
119. Snyder EY, Yoon C, Flax JD, Macklis JD. Multipotent neural progenetors 143. Humm JL, Kozlowski DA, Bland ST, James DC, Schallert T. Use-
can differentiate toward replacement of neurons undergoing targeted apo- dependent exaggeration of brain injury: is glutamate involved? Exp Neurol.
ptotic degeneration in adult mouse neocortex. Proc Natl Acad Sci U S A. 1999;157:349–358.
1997;94:11645–11650. 144. Futrell N, Garcia JH, Peterson E, Millikan C. Embolic stroke in aged rats.
120. Leavitt BR, Hernit-Grant CS, Macklis JD. Mature astrocytes transform into Stroke. 1991;22:1582–1591.
transitional radial glia within adult mouse neocortex that supports directed 145. Davis M, Mendelow AD, Perry RH, Chambers IR, James OF. Experimental
migration of transplanted immature neurons. Exp Neurol. 1999;157:43–57. stroke and neuroprotection in the aging rat brain. Stroke. 1995;26:
121. Weiller C, Chollet KJ, Friston KJ, Wise RJS, Frackowiak RSJ. Functional 1072–1078.
reorganization of the brain in recovery from striatocapsular infarction in 146. Sutherland GR, Dix GA, Auer RN. Effect of age in rodent models of focal
man. Ann Neurol. 1992;31:463–472. and forebrain ischemia. Stroke. 1996;27:1663–1668.
122. Weiller C, Ramsay SC, Wise RJS, Friston KJ, Frackowiak RSJ. Individual 147. Popa-Wagner A, Schroder E, Schmoll H, Walker LC, Kessler C. Upregu-
patterns of functional reorganization in the human cerebral cortex after lation of MAP1B and MAP2 in the rat brain after middle cerebral artery
capsular infarction. Ann Neurol. 1993;33:181–189. occlusion: effect of age. J Cereb Blood Flow Metab. 1999;19:425–434.
123. Weiller C. Imaging recovery from stroke. Exp Brain Res. 1998;123: 148. Saito S, Kobayashi S, Ohashi Y, Igarashi M, Komiya Y, Ando S. Decreased
13–1718. synaptic density in aged brains and its prevention by rearing under enriched
124. Cramer SC, Nelles G, Benson RR, Kaplan JD, Parker RA, Kwong KK, environment as revealed by synpatophysin contents. J Neurosci Res. 1994;
Kennedy DN, Finklestein SP, Rosen BR. A functional MRI study of 39:57–62.
subjects recovered from hemiparetic stroke. Stroke. 1997;28:2518–2527. 149. Buell SJ, Coleman PD. Quantitative evidence for selective dendritic growth
125. Rossini PM, Tecchio F, Pizzella V, Lupoi D, Cassetta E, Pasqualetti P, in normal human aging but not in senile dementia. Brain Res. 1981;214:
Romani GL, Orlacchio A. On the reorganization of sensory hand areas after 23–41.
mono-hemispheric lesion: a functional(MEG)/anatomical (MRI) integrative 150. Wilson RS, Bennett DA, Beckett LA, Morris MC, Gilley DW, Bienias JL,
study. Brain Res. 1998;782:153–166. Scherr PA, Evans DA. Cognitive activity in older persons from a geograph-
126. Pizzamiglio L, Perani D, Cappa SF, Vallar G, Paolucci S, Grassi F, Paulesu ically defined population. J Gerontol B Psychol Sci Soc Sci. 1999;54:
E, Fazio F. Recovery of neglect after right hemispheric damage. Arch P155–P160.
Neurol. 1999;55:561–568. 151. Corkin S. Penetrating head injury in young adulthood exacerbates cognitive
127. Warburton E, Price CJ, Swinburn K, Wise RJS. Mechanisms of recovery decline in later years. J Neurosci. 1989;9:3876–3883.
from aphasia: evidence from positron emission tomography studies. 152. Ulrich R. View through a window may influence recovery from surgery.
J Neurol Neurosurg Psychiatry. 1999;66:155–161. Science. 1984;224:420–421.

You might also like