ArticlePDF Available

Stimulation of the mesencephalic locomotor region for gait recovery after stroke: DBS for Gait Recovery

Authors:

Abstract and Figures

Objective: One-third of all stroke survivors are unable to walk, even after intensive physiotherapy. Thus, other concepts to restore walking are needed. Since electrical stimulation of the mesencephalic locomotor region (MLR) is known to elicit gait movements, this area might be a promising target for restorative neurostimulation in stroke patients with gait disability. The present study aims to delineate the effect of high-frequency stimulation of the MLR (MLR-HFS) on gait impairment in a rodent stroke model. Methods: Male Wistar rats underwent photothrombotic stroke of the right sensorimotor cortex and chronic implantation of a stimulating electrode into the right MLR. Gait was assessed using clinical scoring of the beam walking test and videokinematic analysis (CatWalk™) at baseline and on days 3 and 4 after experimental stroke with and without MLR-HFS. Results: Kinematic analysis revealed significant changes in several dynamic and static gait parameters resulting in overall reduced gait velocity. All rats exhibited major coordination deficits during the beam walking challenge and were unable to cross the beam. Simultaneous to the onset of MLR-HFS, a significantly higher walking speed and improvements in several dynamic gait parameters were detected by the Catwalk™-system. Rats regained the ability to cross the beam unassisted showing a reduced number of paw slips and misses. Interpretation: MLR-HFS can improve disordered locomotor function in a rodent stroke model. It may act by shielding brainstem and spinal locomotor centers from abnormal cortical input after stroke, thus allowing for compensatory and independent action of these circuits. This article is protected by copyright. All rights reserved.
Visualization of the photothrombotic stroke and the electrode tip. (A) Representative coronal T2-weighted (T2w) magnetic resonance scans revealing the photothrombotic stroke (hyperintense area) in the right sensorimotor cortex of a rat brain. (B) Macroscopic view of a rat brain after removal from the skull. The black arrows indicate the photothrombotically induced lesion. Primary motor cortex (M1), blue framed; secondary motor cortex (M2), green framed; primary somatosensory cortex, forelimb, yellow framed; primary somatosensory cortex, hindlimb, brown framed (according to Paxinos and Watson's rat brain atlas 23 ). (C) Brain sections in 3 planes (coronal [top]; sagittal [middle]; horizontal [bottom]) of an averaged brain generated from T2w scans of the rats used in this study. The overlapping size and site of the photothrombotic lesion is displayed by a heatmap on the right hemisphere. The sections are superimposed on the corresponding atlas template. 23 The heatmap color red represents a low overlapping of 10%, whereas yellow indicates an overlapping of 100%. Within the M1, overlapping of the lesions is almost 100%. Cg1 5 cingulate cortex, area 1; Cg2 5 cingulate cortex, area 2; FrA 5 frontal association cortex; MPtA 5 medial parietal association cortex; PrL 5 prelimbic cortex; S1BF 5 primary somatosensory cortex, barrel field; S1DZ 5 primary somatosensory cortex, dysgranular zone; S1FL 5 primary somatosensory cortex, forelimb region; S1HL 5 primary somatosensory cortex, hindlimb region; S1J 5 primary somatosensory cortex, jaw region; S1Tr 5 primary somatosensory cortex, trunk region; S1ULp 5 primary somatosensory cortex, upper lip region; V2ML 5 secondary visual cortex, mediolateral.
… 
Assessment of locomotor behavior before and after photothrombotic stroke without and during highfrequency stimulation of the mesencephalic locomotor region (MLR-HFS; day 4 after intervention). (A) Whereas no locomotion was seen after photothrombotic stroke (PT) without MLR-HFS, gait velocity changed after stroke almost to the baseline values when animals were stimulated in the MLR-HFS. *p < 0.001 (95% confidence interval [CI] 5 20.3124.66), #p < 0.001 (95% CI 5 222.82 to 216.48); ns 5 not significant (p > 0.05, 95% CI 5 20.72 to 6.40); error bars indicate standard deviation; 2-tailed paired t test. (B) Beamwalking score, assessed according to a 7-point scale (see Materials and Methods section). After photothrombotic stroke, MLR-HFS restored gait coordination and balance significantly compared to the test condition without MLR-HFS. *p < 0.001 (95% CI 5 5.25-6.36); #p < 0.001 (95% CI 5 26.38 to 23.49); ns 5 not significant (p > 0.05, 95% CI 5 20.33 to 22.06); error bars indicate standard deviation; 2-tailed paired t test. (C) Effect of MLR-HFS on affected fore-and hindpaw. Whereas there was no difference between forepaw and hindpaw regarding faults before photothrombotic stroke, rats made significantly more faults with the hindpaw than with the forepaw after photothrombotic stroke even during MLR-HFS. *p 5 0.037 (95% CI 5 22.54 to 20.13); ns, not significant (p > 0.05, 95% CI 5 20.29 to 0.29); error bars indicate standard deviation; 2-tailed paired t test. FL 5 forelimb; HL 5 hindlimb; non-stim 5 assessment after PT, no MLR-HFS; stim 5 assessment after PT, with MLR-HFS.
… 
Content may be subject to copyright.
RESEARCH ARTICLE
Stimulation of the Mesencephalic
Locomotor Region for Gait
Recovery After Stroke
Felix Fluri, MD,
1
Uwe Malzahn, PhD,
2
Gy
orgy A. Homola, PhD,
3
Michael K. Schuhmann, PhD,
1
Christoph Kleinschnitz, MD,
1
* and
Jens Volkmann, MD, PhD
1
Objective: One-third of all stroke survivors are unable to walk, even after intensive physiotherapy. Thus, other con-
cepts to restore walking are needed. Because electrical stimulation of the mesencephalic locomotor region (MLR) is
known to elicit gait movements, this area might be a promising target for restorative neurostimulation in stroke
patients with gait disability. The present study aims to delineate the effect of high-frequency stimulation of the MLR
(MLR-HFS) on gait impairment in a rodent stroke model.
Methods: Male Wistar rats underwent photothrombotic stroke of the right sensorimotor cortex and chronic implanta-
tion of a stimulating electrode into the right MLR. Gait was assessed using clinical scoring of the beam-walking test
and video-kinematic analysis (CatWalk) at baseline and on days 3 and 4 after experimental stroke with and without
MLR-HFS.
Results: Kinematic analysis revealed significant changes in several dynamic and static gait parameters resulting in
overall reduced gait velocity. All rats exhibited major coordination deficits during the beam-walking challenge and
were unable to cross the beam. Simultaneous to the onset of MLR-HFS, a significantly higher walking speed and
improvements in several dynamic gait parameters were detected by the CatWalk system. Rats regained the ability to
cross the beam unassisted, showing a reduced number of paw slips and misses.
Interpretation: MLR-HFS can improve disordered locomotor function in a rodent stroke model. It may act by shield-
ing brainstem and spinal locomotor centers from abnormal cortical input after stroke, thus allowing for compensatory
and independent action of these circuits.
ANN NEUROL 2017;00:000–000
Motor deficits are the most common symptoms after
ischemic stroke. Approximately one-third of stroke
survivors suffer from impaired mobility and cannot walk
1 year after an ischemic cerebrovascular event.
1
Gait after
stroke is characterized by a slow and asymmetrical walk-
ing pattern, with reduced stride length and a prolonged
swing phase of the affected limb.
2
There are different
rehabilitation strategies after stroke, such as physical ther-
apy,
3
neurodevelopmental training, and motor relearning
programs.
4
However, the overall effect of physiotherapy
in chronic stroke survivors is modest,
5
and there are no
pharmacological or interventional alternatives. This is
even more remarkable as locomotion relies on phyloge-
netically well-conserved brainstem and spinal circuits,
which themselves remain unaffected by a hemispheric
lesion. These mesencephalic and spinal central pattern
generators (CPGs) produce the alternating rhythmic neu-
ral signals necessary for stepping behavior
6
and the anti-
gravitational postural adjustments required for walking.
7
In animals, CPGs are capable of sustaining coordinated
View this article online at wileyonlinelibrary.com. DOI: 10.1002/ana.25086
Received Sep 5, 2016, and in revised form Dec 29, 2016, Jun 20, 2017, and Oct 19, 2017. Accepted for publication Oct 20, 2017.
Address correspondence to Dr Fluri, Department of Neurology, University Hospital W
urzburg, Josef-Schneider Strasse 11, 97080 W
urzburg, Germany.
E-mail: felix.fluri@gmx.ch
From the
1
Department of Neurology, University Hospital W
urzburg;
2
Institute of Clinical Epidemiology and Biometry, University of W
urzburg; and
3
Department of Diagnostic and Interventional Neuroradiology, University Hospital W
urzburg, W
urzburg, Germany.
*Current address for Dr Kleinschnitz: Department of Neurology, University Hospital Essen, Essen, Germany.
Additional supporting information can be found in the online version of this article.
V
C2017 American Neurological Association 1
gait even after complete deafferentation from cortical
input.
8,9
Hence, the interesting question arises whether
gait impairment after stroke could reflect a dysfunc-
tional descending input to brainstem and spinal central
pattern generators,
10
rather than a loss of function of
these pathways. This would open the possibility of neu-
romodulatory therapy such as deep brain stimulation
(DBS), which is highly effective in treating dysfunc-
tional motor circuit activity in Parkinson disease
11
or
dystonia.
12
Here, we explore the restorative potential of DBS
of the mesencephalic locomotor region (MLR) in a rat
model of hemiplegic stroke. We induced gait deficits in
Wistar rats by inflicting a photothrombotic lesion to the
right sensorimotor cortex,
13
and evaluated stimulation
effects of the MLR by video-kinematic gait analysis and
behavioral assessment during a gait challenge. Photo-
thrombosis is a widely used stroke model in rodents with
the advantage of lesioning a defined brain region, thus
allowing probing the response of the disabled brain site
and interconnections to a targeted intervention. The
MLR was chosen as a target for neuromodulation
because it is involved in the initiation and control of
gait.
14
A recently published study suggests that the MLR
of rats consists of noncholinergic, predominantly gluta-
matergic cells localized in the lateral pontine tegmentum,
which form a strip extending from the ventrolateral peri-
aqueductal gray matter to the region ventromedial to the
pedunculopontine tegmental nucleus (PTg) with ipsilat-
eral predominace.
15
Electrical or pharmacological stimu-
lation of this region elicits locomotor behavior even in
decerebrated animals such as cats,
8
rats,
16
salamanders,
17
and nonhuman primates.
18
Moreover, there is clinical
experience with MLR stimulation in patients with Par-
kinson disease,
19–21
in principle making it feasible to
translate results from rodents to humans.
Materials and Methods
Animals
All experiments were performed in adult (250–275g, 6–8 weeks
old) male Wistar rats (n 520; Charles River, Sulzfeld,
Germany). Rats were acclimatized for 1 week in an animal facil-
ity and housed in a room with con trolled temperature (22 6
0.5 8C) under a 12/12-hour light/dark cycle. They were allowed
free access to food and water. All animal experiments were
approved by the institutional review board of Julius Maximilian
University, Wurzburg and by the local authorities of lower
Frankonia (Regierung von Unterfranken, Wurzburg, Germany).
Induction of Photothrombotic Stroke
Animals were divided into 2 groups: the first underwent photo-
thrombotic stroke only (n 510), whereas the second was sub-
jected to both photothrombosis and electrode insertion into the
MLR (n 510, see below), to exclude any behavioral effect of
electrode implantation into the MLR.
Rats were anesthetized with 2.5% isoflurane during the
surgery. Body temperature was maintained at 37 60.5 8Cbya
feedback-controlled heating system. A photothrombotic cerebral
stroke was induced in all rats as follows.
22
A template with an
aperture (10 35mm) for the light source was put on the
exposed skull 5mm anterior to 5mm posterior and 0.5mm to
5.5mm lateral to bregma, an area which corresponds to the sen-
sorimotor cortex (Fig 1).
23
A light guide was placed over the
aperture. Rose Bengal (0.5ml; Sigma, St Louis, MO) in NaCl
0.9% (10mg/ml) was administered intravenously, and the brain
was illuminated (KL1500LCD; Olympus, Tokyo, Japan)
through the intact skull for 15 minutes. Immediately after this
procedure, a microelectrode was implanted in half of the ani-
mals (see section below).
Microelectrode Implantation
For high-frequency stimulation of the MLR (MLR-HFS),
monopolar microelectrodes (catalogue # UE-PSEGSECN1M;
FHC, Bowdoin, ME) were used in this study. To avoid electro-
chemical neurotoxicity, electrodes made of platinum/iridium
with a mean impedance of 0.93MX(range 50.8–1.2MX) were
used. Electrodes were implanted close to the dorsal part of the
MLR ipsilateral to the lesion (coordinates: 7.8mm posterior,
2.0mm lateral, and 5.8mm ventral to the bregma) as described
in detail elsewhere.
24
By using the aforementioned coordinates,
the tip of the electrode was placed slightly above the dorsal part
of the MLR (Fig 2A), which avoids the destruction of this
small structure but still ensures an effect of stimulation on the
MLR. The electrode was implanted ipsilateral to the photo-
thrombotic stroke for the following reason. In a recent study by
Bachmann et al,
25
unilateral injection of FastBlue into the left
rostral medulla oblongata resulted in predominant retrograde
labeling of the left MLR and left motor cortical areas and to a
lesser extent of the contralateral cortex, indicating a largely
uncrossed organization of the corticomesencephalic–spinal loco-
motor circuit (as corroborated by Matsumura et al
26
in
macaque monkey).
Five stainless steel screws (M1.6; length, 3mm; Hummer
& Rieß, Nuremberg, Germany) were inserted in boreholes
without penetrating the dura overlaying the brain surface. A
custom-made plug (GT-Labortechnik, Arnstein, Germany) was
put on the pin of the electrode, and the ground wire of the
plug was connected with one of the screws. To fix the elec-
trode/plug with the bone screws, dental cement was applied on
the skull and molded around the electrode/plug by forming a
small cap. Wound edges were closed with a suture at the front
and behind the cap. Thereafter, animals were allowed to wake
up.
Behavioral Testing
Rats were trained for 7 days to traverse a horizontal wooden
beam (90cm long, 9mm wide, 70cm above ground). At the
same time, they learned to cross the runway of the CatWalk
system (Noldus, Wageningen, the Netherlands), a video-based
ANNALS of Neurology
2 Volume 00, No. 0
analysis system to assess static and dynamic gait parameters (for
a complete description of this method, see Hamers et al
27
). On
the last day of training, traversing the wooden beam and cross-
ing the CatWalk system (3 runs per animal) were recorded;
these measurements were used as baseline values. Induction of
photothrombotic stroke and implantation of the electrode were
performed 1 day later (ie, on day 8 after the beginning of train-
ing). Three days after intervention, locomotor behavior was first
investigated without stimulation using the CatWalk system.
Three hours later, the same experiment was carried out with
FIGURE 1: Visualization of the photothrombotic stroke and the electrode tip. (A) Representative coronal T2-weighted (T2w)
magnetic resonance scans revealing the photothrombotic stroke (hyperintense area) in the right sensorimotor cortex of a rat
brain. (B) Macroscopic view of a rat brain after removal from the skull. The black arrows indicate the photothrombotically
induced lesion. Primary motor cortex (M1), blue framed; secondary motor cortex (M2), green framed; primary somatosensory
cortex, forelimb, yellow framed; primary somatosensory cortex, hindlimb, brown framed (according to Paxinos and Watson’s
rat brain atlas
23
). (C) Brain sections in 3 planes (coronal [top]; sagittal [middle]; horizontal [bottom]) of an averaged brain gener-
ated from T2w scans of the rats used in this study. The overlapping size and site of the photothrombotic lesion is displayed by
a heatmap on the right hemisphere. The sections are superimposed on the corresponding atlas template.
23
The heatmap color
red represents a low overlapping of 10%, whereas yellow indicates an overlapping of 100%. Within the M1, overlapping of
the lesions is almost 100%. Cg1 5cingulate cortex, area 1; Cg2 5cingulate cortex, area 2; FrA 5frontal association cortex;
MPtA 5medial parietal association cortex; PrL5prelimbic cortex; S1BF 5primary somatosensory cortex, barrel field;
S1DZ 5primary somatosensory cortex, dysgranular zone; S1FL 5primary somatosensory cortex, forelimb region; S1HL 5pri-
mary somatosensory cortex, hindlimb region; S1J 5primary somatosensory cortex, jaw region; S1Tr 5primary somatosensory
cortex, trunk region; S1ULp 5primary somatosensory cortex, upper lip region; V2ML 5secondary visual cortex, mediolateral.
Fluri et al: DBS for Gait Recovery
Month 2017 3
HFS (frequency 5130Hz, pulse length 560 microseconds,
pulse shape 5monophasic square wave pulses) using the stimu-
lus generator STG 4002 (Multichannel Systems, Reutlingen,
Germany). This device includes a large voltage compliance
range of 120V, as well as constant current stimulation as a par-
ticular feature. In the current mode, the device is able to adjust
the voltage to changes in tissue impedance, and thus provides a
constant current output at the electrode. Before starting gait
analyses, a threshold current intensity was determined for each
animal by observing spontaneous locomotor behavior as
described recently.
25
The current threshold for stimulation-
evoked locomotion was determined by beginning at 20 mA and
then increasing the intensity in 10 mA steps until the maximal
locomotion was seen. Switching on and off the device tested
reproducibility of the stimulus-induced locomotion. Thereafter,
the lowest current-evoking locomotion was chosen for further
testing. In the present study, the lowest current intensity result-
ing in increased locomotor activity was 40 mA in all tested ani-
mals. Three crossings of the CatWalk runway without
interruption/hesitation were required for a valid kinematic gait
analysis in each animal. Data were analyzed using the CatWalk
XT 10 software.
The 5 most widely used gait parameters in recently pub-
lished studies on locomotion after stroke
28,29
were analyzed,
namely step cycle, swing speed, and duty cycle (ie, dynamic
paw parameters) as well as stride length and contact area (ie,
static paw parameters, Supplementary Table 1).
The beam-walking task was performed 4 days after pho-
tothrombosis using the same parameters for HFS as during
CatWalk testing. Three traverses per animal were performed
and video was recorded. The time passing between the first and
the last step on the beam was taken to calculate the gait speed.
Fine motor coordination and balance were further determined
using a 7-point nonparametric scale and animals were scored as
follows
30
:15unable to traverse or falls off the beam;
25unable to traverse the beam but able to maintain balance
on the beam; 3 5able to traverse the beam by dragging the
affected limb; 4 5able to traverse the beam and—at least
once—to place the affected limb on the horizontal surface of
the beam; 5 5the affected limbs are used in <50% of its steps
FIGURE 2: Verification of electrode placements. Consecutive brain sections encompassing the electrode site were used for
fluorescent in situ hybridization to visualize choline acetyl-transferase (ChAT)
1
neurons of the pedunculopontine tegmental
area (PTg) and c-Fos
1
neurons indicating the stimulation site. Both ChAT
1
and c-Fos
1
cell groups as well as the electrode site
(dots) were mapped onto atlas drawings of the rat brain. The relationship of electrode sites to cholinergic neurons of the PTg
(A) and to the c-Fos
1
neurons (B) are visualized by cloud diagrams. It is of note that ChAT
1
neurons rarely expressed c-Fos in
our study. The numbers below the drawings indicate the anterior–posterior distance to the bregma. The light gray areas indi-
cate the PTg, the dark gray areas the cuneiform nucleus (Cn).
ANNALS of Neurology
4 Volume 00, No. 0
on the beam; 6 5able to traverse the beam by using the
affected limbs (contralateral to the lesion and implanted elec-
trode) for >50% of its steps along the beam; 7 5able to tra-
verse the beam normally with no more than 2 foot slips. To
examine whether the use of the left fore- and hindlimb changes
during MLR-HFS after photothrombotic stroke, paw slips and
misses off the beam (1 point per fault) were counted before
photothrombotic stroke and thereafter under HFS conditions.
Both the CatWalk analysis and the beam-walking test
were also carried out in rats subjected to photothrombotic
stroke alone, to investigate whether electrode implantation into
the MLR influences locomotor behavior.
Measurement of Lesion Volume
Lesion size was visualized using T2-weighted (T2w) magnetic
resonance imaging (MRI) on a 3.0T scanner (MAGNETOM
Trio; Siemens, Erlangen Germany). T2w scans were acquired
with turbo spin-echo sequences (echo time 5105 milliseconds,
repetition time 52,100 milliseconds) and infarct volume was
determined using ImageJ Analysis Software 1.45s (National
Institutes of Health, Bethesda, MD; http://rsb.info.nih.gov/ij/);
the hyperintense lesion on each scan (1mm thick) was traced
manually and the areas were then summed and multiplied by
the slice thickness.
To compare the location and size of the photothrombotic
lesion among all animals, an average brain of these animals was
generated and all lesions were overlapped in a color-coded heat-
map on this brain as follows. T2w images were brain-extracted
with the brain extraction tool of FSL (FMRIB, Oxford, UK)
optimized for rodent brains and additionally corrected manu-
ally. All lesions were segmented manually in original data. Rat
brains were registered with FLIRT (FMRIB). The lesion masks
were transformed according to the individual brain registra-
tions. The sum of all lesions was overlaid on the normalized
average brain data using a color lookup table. Thereafter, 3
brain sections representing each plane were superimposed on
the corresponding atlas template.
23
Immunohistochemistry
After deep anesthesia, rats were killed by decapitation and the
brains were harvested rapidly and immediately frozen at
220 8C. Coronal sections (12 mm thick) were cut using a cryo-
stat (Leica 3050; Leica Microsystems, Wetzlar, Germany). Sec-
tions encompassing the MLR were stained with hematoxylin
and eosin to visualize the anatomic locations of the electrode
tip. The localization of the stimulation sites was assessed by
choosing 2 approaches. First, the relationship of the electrode
tip location was mapped out with respect to the cholinergic
neurons of the PTg; second, c-Fos expression sites were com-
pared to the localization of cholinergic neurons.
To identify choline acetyl-transferase (ChAT)-positive
neurons of the PTg and to visualize the expression of c-Fos,
fluorescent in situ hybridization of sections encompassing the
MLR and lesion due to the electrode tip were performed using
the RNAscope Multiplex Fluorescent v2 Assay according to the
manufacturer’s instructions (Advanced Cell Diagnostics, Milan,
Italy; catalogue # 323100). Target probes for c-Fos (RNAscope
probe Rn-Fos, catalogue # 403591) and ChAT (RNAscope
probe Rn-Chat-C2, catalogue # 430111-C2) were designed by
Advanced Cell Diagnostics. After amplification and label appli-
cation, sections were counterstained with 4,6-diamidino-2-phe-
nylindole (Sigma-Aldrich, St Louis, MO; catalogue # D9542).
Images were acquired with a Leica MDi8 microscope (magnifi-
cation 5403). Finally, ChAT
1
cells of the PTg as well as c-
Fos
1
cells around the stimulation site of each animal were
delineated as a cloud onto atlas drawings of consecutive (corre-
sponding) brain sections. These cell groups were then related to
the distal end of the electrode trajectory.
Statistical Analysis
For gait speed and number of step cycles measured by the Cat-
Walk system, individual averages of each rat were calculated
over 3 runs for each time point (ie, measurements before and
after photothrombotic stroke, as well as during MLR-HFS) and
used to get group means and standard deviations (SDs). Gait
speed and number of step cycles were further analyzed using
repeated measures analysis of variance (ANOVA) with Green-
house–Geisser corrections as appropriate for sphericity viola-
tions. Post hoc analyses were performed with Tukey multiple
comparison test.
Additional gait parameters (ie, step cycle, swing speed,
and duty cycle as well as stride length and contact area) mea-
sured by the CatWalk system were analyzed as raw values in
relation to instantaneous body velocity.
31
This was necessary,
because most gait parameters change as a function of speed
32
and photothrombotic stroke reduces gait velocity, such that the
intervention itself would act as a confounder. In a first step, we
plotted scattergraphs of each parameter against body velocity
and compared the mostly nonlinear distributions under the 3
different treatment conditions visually. We then conducted
global and velocity-restricted group comparisons (repeated mea-
sures ANOVA and post hoc ttests for paired samples with
Bonferroni correction) at slow (16–30cm/s), medium (30–
65cm/s), and fast (65–150cm/s) body speed.
For the beam-walking test, statistical differences of gait
velocity and scores before photothrombosis and 4 days after
intervention under MLR-HFS were calculated using the 2-
tailed paired ttest. All values are presented as mean 6SD with
95% confidence intervals (CIs). Probability values <0.05 were
considered to indicate statistical significance. Statistical Package
for the Social Sciences (SPSS 17.0; IBM, Armonk, NY) soft-
ware was used for statistical analysis.
Results
Baseline Characteristics
One animal died in each group during the experiment
and thus had to be excluded from the analyses. T2w
scans revealed a photothrombotic lesion in all animals
encompassing the right sensorimotor cortex (see Fig 1A,
B). On T2w scans, lesion size did not differ significantly
between both groups (mean lesion volume: first group,
Fluri et al: DBS for Gait Recovery
Month 2017 5
72.8 66.4mm
3
vs second group, 84.9 69.6mm
3
;
p50.32). To further determine size and site of the pho-
tothrombotic stroke, all scanned brains were coregistered
and a template was calculated; thereafter, the degree of
overlapping of the photothrombotic lesions was visual-
ized by a color-coded heatmap on the template. Whereas
the primary motor cortex (M1) was affected to almost
100% in all animals, the degree of lesional overlapping
was decreased within the secondary motor cortex (M2)
and the primary somatosensory cortex representing the
hindlimb (estimated 75%) and was even less within the
primary somatosensory cortex representing the forelimb
(estimated 50%; see Fig 1C).
Immediately after intervention, as well as before
kinematic gait testing (ie, 3 days after the intervention),
all animals exhibited normal cage mobility and no coor-
dination deficits were observed in the use of the affected
left forepaw during food uptake.
Hematoxylin and eosin staining revealed some vari-
ability of electrode placement (see Fig 2A). In 6 animals,
the electrodes were placed close to or within the cunei-
form nucleus (Cn; ie, 27.80mm from the bregma),
whereas 2 animals exhibited a deviation of the tip posi-
tion into the anterior direction 120 mm from the Cn (ie,
27.68mm from bregma). The electrode tip of a third
animal was found at the lower right outer border of the
Cn, 27.92mm from bregma. It was decided to keep all
animals in the analysis using an intention-to-treat
approach, because the optimal mesencephalic site of
stimulation was not yet defined.
Histological Analysis of the Neurostimulation
Effects
Fluorescent in situ hybridization of consecutive brain sec-
tions were performed to visualize the lesion due to the
electrode tip, the ChAT
1
neurons of the PTg, and c-
Fos
1
neurons around the stimulation site of each animal.
Then, brain sections were mapped onto atlas drawings of
the rat brain to show the relationship of electrode sites to
cholinergic neurons of the PTg (see Fig 2A) as well as to
FIGURE 3: Assessment of locomotor behavior before and
after photothrombotic stroke without and during high-
frequency stimulation of the mesencephalic locomotor
region (MLR-HFS; day 4 after intervention). (A) Whereas no
locomotion was seen after photothrombotic stroke (PT)
without MLR-HFS, gait velocity changed after stroke almost
to the baseline values when animals were stimulated in the
MLR-HFS. *p<0.001 (95% confidence interval [CI] 520.31–
24.66), #p<0.001 (95% CI 5222.82 to 216.48); ns 5not
significant (p>0.05, 95% CI 520.72 to 6.40); error bars
indicate standard deviation; 2-tailed paired ttest. (B) Beam-
walking score, assessed according to a 7-point scale (see
Materials and Methods section). After photothrombotic
stroke, MLR-HFS restored gait coordination and balance sig-
nificantly compared to the test condition without MLR-HFS.
*p<0.001 (95% CI 55.25–6.36); #p<0.001 (95% CI 526.38
to 23.49); ns 5not significant (p>0.05, 95% CI 520.33 to
22.06); error bars indicate standard deviation; 2-tailed
paired ttest. (C) Effect of MLR-HFS on affected fore- and
hindpaw. Whereas there was no difference between fore-
paw and hindpaw regarding faults before photothrombotic
stroke, rats made significantly more faults with the hindpaw
than with the forepaw after photothrombotic stroke even
during MLR-HFS. *p50.037 (95% CI 522.54 to 20.13); ns,
not significant (p>0.05, 95% CI 520.29 to 0.29); error bars
indicate standard deviation; 2-tailed paired ttest. FL 5fore-
limb; HL 5hindlimb; non-stim 5assessment after PT, no
MLR-HFS; stim 5assessment after PT, with MLR-HFS.
ANNALS of Neurology
6 Volume 00, No. 0
the c-Fos
1
neurons (see Fig 2B). The more rostrally
localized electrodes (ie, 27.68mm anterior–posterior to
the bregma) had less effect on gait speed and explain
why velocities of 25.4cm/s and 25.6cm/s were mea-
sured in these 2 animals. Different elements such as
cell somata (ie, glutamatergic as well as cholinergic
neurons) as well as axons and dendrites might be acti-
vated by applying HFS.
Beam-Walking Test
The behavioral outcome after stroke with and without
MLR-HFS was evaluated using the beam-walking test.
This test allows evaluation of coordination and
integration of paw movements after skilled gait training.
On day 7 of training (ie, before photothrombotic stroke),
the average speed of the beam traversing was 22.5 6
0.8cm/s (Supplementary Video 1). Four days after photo-
thrombotic stroke, all animals demonstrated paw
coordination deficits and were unable to traverse the
beam without assistance (Supplementary Video 2). When
MLR-HFS was applied, coordinated locomotion was
restored instantaneously and an average speed of 19.7 6
1.2cm/s for unassisted beam traversing was recorded
(Supplementary Video 3, Fig 3A), which was similar to
the gait velocity measured before photothrombotic stroke
(p50.096).
TABLE 1. Comparison of Gait Parameters Using the CatWalk System: Changes in Locomotor Variables Mea-
sured Before and After Photothrombotic Stroke
LH LF RH RF
Parameter Overall
Mean
Constrained
Velocities
a
Overall
Mean
Constrained
Velocities
a
Overall
Mean
Constrained
Velocities
a
Overall
Mean
Constrained
Velocities
a
Body speed ## ns ## ns ## " ## ns
ns ns ## ##
ns ns ns ns
Step cycle, s "ns ns ns ns ns ns "
"" ns "" "
ns ns ns ns
Duty cycle ## # ns ns #ns ns ns
## ns ## #
## ns ns ns
Stride
length, cm
#ns ns ns ## ns ## ns
ns ns ns ns
ns ## ns ##
Swing speed,
cm/s
## ns ## ## ## ns ## ns
## ## ## ##
## ns ns ns
Contact
area, cm
2
## ## ## ## ## ns ## ns
## ## ## ##
## ## ns ##
A significant increase/decrease of a gait parameter is indicated by "/#(p<0.05) and ""/## (p<0.01). Individual numeric values for each parame-
ter and paw are outlined in Supplementary Table 2.
a
Slow (top), medium (middle), and fast (bottom).
LF 5left forelimb; LH 5left hindlimb; ns 5not significant; RF 5right forelimb; RH 5right hindlimb.
Fluri et al: DBS for Gait Recovery
Month 2017 7
Changes in locomotor skills and balance were fur-
ther determined using a 7-point scale. On day 7 of train-
ing, mean score was 7; only 1 animal slipped with the
forepaw and another with the hindpaw when crossing
the beam. Four days after photothrombotic stroke, skilled
walking on the beam was first tested without HFS in all
animals. Only 1 animal was able to maintain balance on
the beam; all others fell off the beam. After applying
MLR-HFS, skilled locomotion improved significantly in
all animals; one of them returned even to a score of 7
(mean), whereas the animal with least effect of MLR-
HFS regained a mean score of 4.6 (see Fig 3B).
To determine whether MLR-HFS exerts a more pow-
erful effect on the fore- or hindlimb, we assessed paw slips
and misses off the beam before photothrombotic stroke
and thereafter under MLR-HFS conditions. The number
of slips measured for forepaw and hindpaw did not differ
before intervention. Whereas no locomotion was visible
and thus this parameter was not evaluable after photo-
thrombotic stroke without MLR-HFS, significantly fewer
paw slips and misses off the beam were observed for the
forepaw compared to the hindpaw during MLR-HFS (see
Fig 3C).
No difference in locomotor behavior was found
between animals with photothrombotic stroke alone and
those subjected to both photothrombosis and electrode
implantation (ie, both groups were no longer able to tra-
verse the beam on day 4 after the intervention), exclud-
ing a clinically relevant impact of MLR microlesioning
by electrode implantation.
TABLE 2. Comparison of Gait Parameters Using the CatWalk System: After Photothrombosis without and
with Mesencephalic Locomotor Region High-Frequency Stimulation
LH LF RH RF
Parameter Overall
Mean
Constrained
Velocities
a
Overall
Mean
Constrained
Velocities
a
Overall
Mean
Constrained
Velocities
a
Overall
Mean
Constrained
Velocities
a
Body speed "" ns "" ns "" ns "" ns
ns "" ns ""
""""""
Step cycle, s ## ns ## ns #ns ns ns
#ns ns ns
## ns #ns
Duty cycle ## ns ns ns ns ns #ns
ns ns ns ns
ns ns ns ns
Stride length, cm "ns ns ns "" ns "" ns
ns ns "" ns
"" "" ns ns
Swing speed, cm/s "" ns "" # "" # "" ns
ns ns ns ns
"" ns ns ns
Contact area, cm
2
ns ## # ## ns ## ns ##
ns ns ns ns
ns ns ns ns
A significant increase/decrease of a gait parameter is indicated by "/#(p<0.05) and ""/## (p<0.01). Individual numeric values for each parame-
ter and paw are outlined in Supplementary Table 2.
a
Slow (top), medium (middle), and fast (bottom).
LF 5left forelimb; LH 5left hindlimb; ns 5not significant; RF 5right forelimb; RH 5right hindlimb.
ANNALS of Neurology
8 Volume 00, No. 0
CatWalk Analyses
Locomotor impairments of fore- and hindpaws after stroke
and MLR-HFS–related changes of gait were quantified
using the CatWalk system. A similar total number of step
cycles before and after photothrombotic stroke was mea-
sured (3.9 60.4 vs 3.9 60.3; p50.86, 95% CI 520.43
to 0.51). After induction of stroke, MLR-HFS did not
change significantly the number of step cycles compared to
the nonstimulated state (3.9 60.3 vs 3.7 60.6; p50.49,
95% CI 520.40 to 0.77). Gait velocity (mean) was 43.26
6.6cm/s in “healthy” animals, which was significantly
reduced after photothrombosis (31.7 69.0cm/s; p50.007,
95% CI 54.11–18.9). When MLR-HFS was applied after
photothrombotic stroke, a significant increase in gait veloc-
ity was observed (43.8 612.6cm/s vs 31.7 69.0cm/s;
p50.04, 95% CI 5223.5 to 20.74).
When comparing gait parameters before and after
photothrombosis (without HFS), mean step cycle of the
right paws and the left forepaw did not change signifi-
cantly (Table 1), whereas mean stride length, swing
speed, and contact area of all paws (except for the stride
length of the left forepaw) were significantly decreased.
With respect to stride length, the right paws exhibited
the largest deficits. The velocity constrained analysis
revealed that these deficits were largely observed at
medium gait velocity, but not at slow velocity.
TABLE 3. Comparison of Gait Parameters Using the CatWalk System: Before Photothrombosis and Thereafter,
When Mesencephalic Locomotor Region High-Frequency Stimulation Was Applied
LH LF RH RF
Parameter Overall
Mean
Constrained
Velocities
a
Overall
Mean
Constrained
Velocities
a
Overall
Mean
Constrained
Velocities
a
Overall
Mean
Constrained
Velocities
a
Body speed ns ns ns ns ns "" ns ns
ns ns ##
"" " ns ""
Step cycle, s ns ns ns ns ns ns ns ns
ns ns "" ns
## # ns ns
Duty cycle ## ns ns ###ns ns ns
## ns ## ##
## ns ns ns
Stride
length, cm
ns ns ns ns ns ns ns ns
ns ns ns ns
"" ns ns ns
Swing
speed, cm/s
## ns ns ## ## ns ns ns
## ## ## ##
#ns ns ns
Contact
area, cm
2
## ## ## ## ## ## ## ##
## ## ## ##
## ns ns #
A significant increase/decrease of a gait parameter is indicated by "/#(p<0.05) and ""/## (p<0.01). Individual numeric values for each parame-
ter and paw are outlined in Supplementary Table 2.
a
Slow (top), medium (middle), and fast (bottom).
LF 5left forelimb; LH 5left hindlimb; ns 5not significant; RF 5right forelimb; RH 5right hindlimb.
Fluri et al: DBS for Gait Recovery
Month 2017 9
When MLR-HFS was applied after stroke, the over-
all mean value of step cycle decreased significantly for all
paws except for the right forepaw, whereas mean values
of swing speed increased significantly (Table 2). Mean
values for stride length increased significantly for the
right paws, whereas the value of the left hindpaw
remained unchanged. Mean values of the contact area—a
static paw parameter—did not change significantly dur-
ing MLR-HFS.
Next, we compared gait parameters before photo-
thrombosis and thereafter when MLR-HFS was applied
(Table 3). There was no significant difference with
respect to step cycle and stride length between the base-
line and stimulated stroke condition and duty cycle and
swing speed of the forepaws. In contrast, contact area of
all 4 paws, as well as duty cycle and swing speed of the
hindpaws, remained significantly reduced after photo-
thrombotic stroke despite MLR-HFS.
Discussion
In the present study, we examined the effect of MLR-
HFS on stroke-related locomotor deficits in rats, which
underwent photothrombotic lesioning of the right senso-
rimotor cortex and implantation of a stimulation elec-
trode into the MLR ipsilateral to the infarction. We
verified that the MLR is a site of action of HFS by dem-
onstrating immunohistochemically that glutamatergic as
well as cholinergic cells of the dorsal part of the MLR
but also cells in adjacent areas expressed c-Fos after
MLR-HFS. It is difficult, however, to assign the stimula-
tion effect to particular nuclei or neural elements within
this region, because monopolar stimulation with a cur-
rent intensity of 40 mA, as used in this experiment, may
excite neural elements (ie, myelinated axons) within a
radius of 500 to 700 mm from the electrode tip.
33
Which
of these elements alone or in combinations contribute to
the observed behavioral responses can only be answered
in future investigations using cell-type–specific stimula-
tion techniques (eg, opto- or pharmacogenetics).
Gait analysis on day 3 after photothrombosis
revealed impairment of dynamic gait parameters caused
by paresis and—to a lesser extent—by coordination defi-
cits of the contralateral fore- and hindlimb during video-
kinematic assessment. The deficits were subtle and barely
visible during spontaneous cage locomotion. Only chal-
lenging tests such as the beam-walking task, requiring
nonparetic paws and unimpaired interlimb coordination
for maintaining body balance on a narrow path, revealed
a clinically relevant locomotor deficit in cortically
lesioned animals. Implantation of the stimulating elec-
trode alone had no impact on poststroke gait symptoms,
whereas acute HFS of the MLR through the chronically
implanted electrode resulted in an immediate restoration
of the ability to cross the test beam without assistance.
The most prominent behavioral changes induced
by MLR-HFS were an increased gait velocity (127.6%)
as revealed by kinematic analysis as well as a significant
amelioration of the skilled walking on the beam. Interest-
ingly, when investigating the left fore- and hindpaw
regarding slips off the wooden beam, a significantly
higher recovery of the forepaw compared to the hindpaw
was seen when cortically lesioned animals traversed the
beam under MLR-HFS. This might be explained by less
lesioning of the forelimb than hindlimb representation
within sensorimotor cortex as shown by the heatmaps.
However, the differences of lesion size and site between
the somatosensory hind- and forelimb representation
were small, and cortical representations vary widely
among individual rats. Alternatively, one might argue
that the microelectrode was implanted in a section of the
MLR representing the forepaw. Again, this seems
unlikely, because previous studies suggest a nonsomato-
topic and rather mixed body representation in the
MLR.
26
We would therefore like to forward the follow-
ing alternative hypothesis. Descending projections of the
MLR target the medullary and pontine reticular forma-
tion.
34
Recently, Esposito and coworkers have shown that
a distinct brainstem nucleus in the ventral part of the
medullary reticular formation plays a crucial role in con-
trolling motor activity of forepaws by demonstrating that
this brainstem area is connected to a subset of forelimb-
innervating spinal motor neurons.
35
Furthermore, their
experiments showed that a larger number of brainstem
nuclei are connected with forelimb than with hindlimb
motor neurons,
35
which might explain—in part—the
better response of the left forepaw to MLR-HFS.
Although the MLR has no direct axonal projections to
the somatomotor cortex, it is nevertheless indirectly con-
nected to the cortex via a relay in the thalamus,
36
which
might contribute to the modulation of the motor func-
tion of the forelimb.
During MLR-HFS, gait velocity increased signifi-
cantly in cortically lesioned animals. This finding might
be of clinical relevance, because independent community
ambulation of stroke patients has been shown to require
a certain degree of gait velocity (ie, 0.80m/s).
37
Addition-
ally, an increase of gait velocity augments the degree of
ambulatory activity, which is low in stroke survivors.
38
However, gait velocity induced by MLR-HFS varied
greatly among animals, as indicated by the large SD. It is
of note that stimulation parameters were always the same
in each animal, such that we suspect electrode placement
in relation to the individual MLR as an important con-
tributing factor. The anatomical extent of the MLR
ANNALS of Neurology
10 Volume 00, No. 0
overlaps with a region ventromedial to the PTg,
15
the
mesencephalic reticular nucleus, PTg, and Cn in
rodents.
39
Most of the electrodes were placed in the cen-
ter or toward the ventral margin of the Cn. Electrical
stimulation of the ventral margin of the Cn has been
shown to evoke alternating hindlimb movements in pre-
collicular–postmamillary decerebrated cats or a change in
locomotor behavior from fast walking to gallop in pre-
collicular–premamillary decerebrated cats.
40
In the pre-
sent study, 2 of the electrodes were detected at the far
rostral pole of both the Cn/PTg and the MLR. The cur-
rent–distance relation
33
may result in less intense stimu-
lation of the MLR cell populations controlling
locomotor speed (especially the population in the region
ventromedial to the PTg) with such an electrode loca-
tion, which—in turn—would explain a more modest
increase of speed after HFS. Additionally, repetitive elec-
trical stimulation of the dorsal part of the PTg in decere-
brated cats elicited stepping movements of the hindlimb;
however, these repetitive stimuli subsequently attenuated
locomotion along with a decrease in muscle tone.
40
Alto-
gether, the variability of placement of electrodes in the
present study indicates that many sites in the midbrain
may have some effect on locomotion, but not necessarily
the same, which is in line with the study published by
Takakusaki et al.
40
The presented findings are also ham-
pered by the small sample size of 9 animals.
Altogether, this study demonstrates that MLR-HFS
can ameliorate gait disability in a rat model of hemiple-
gic stroke and that a unilateral stimulation of the MLR
(ie, ipsilateral to the photothrombotic stroke) is sufficient
to improve quadrupedal walking. This emphasizes the
restorative potential of mesencephalic and spinal motor
circuits supporting locomotion, which may be unlocked
by neuromodulation therapy. We propose that MLR-
HFS shields the mesencephalic and downstream locomo-
tor systems from aberrant cortical input after stroke, and
allows for autonomous function of these circuits. The
nature and origin of the dysfunctional input activity
remains enigmatic. In Parkinson disease and dystonia, a
proposed mechanism of DBS is the suppression of
abnormal neuronal oscillations binding the basal ganglia–
thalamocortical network into a pathological functional
state.
41
Whether similar dysfunctional activity arises from
the perilesional area after stroke, as a result of maladap-
tive compensatory changes within the cortical motor net-
work or due to cortical deafferentation of the tonic
inhibitory basal ganglia input to the MLR, remains to be
elucidated in future studies.
Another aspect requiring additional research is the
optimal stimulation site within the MLR. The MLR is
primarily a functionally defined region at the
mesopontine junction; its anatomical substrate is not
fully characterized and still remains a matter of debate.
42
The MLR has been suggested to comprise noncholinergic
(ie, glutamatergic) cells that have been identified within
the lateral pontine tegmentum, confined medially by the
ventrolateral periaqueductal gray matter and laterally by
the PTg.
15
Electrical stimulation at various sites within
this region has elicited different forms of locomotor
behavior in various species, depending on the stimulation
amplitude. Because electrical pulses preferentially activate
myelinated fibers ortho- and antidromically, before small-
diameter fibers and cell bodies, it is difficult to discern
the anatomical substrate of a neurostimulation effect that
is often not local, but remote through modulation of
pathways rather than nuclei, even if the precise anatomi-
cal location of the electrode tip and the electrical field
distribution were known.
33
Moreover, a recent study of
pedunculopontine neurostimulation in a rat model of
Parkinson disease has cast doubt on a prominent locomo-
tor function of the PTg.
43,44
The overall effects of
pedunculopontine stimulation in patients have been dis-
appointing, apart from a group in which—by error—a
more lateral target site in the mesencephalon was cho-
sen,
45,46
possibly corresponding to the cuneiform nucleus
in man.
We are aware of several limitations in our study.
First, the number of animals (n 59 per group) is rela-
tively small and histological evaluation revealed a varia-
tion of placement of electrodes, which might explain the
variable stimulation outcome in this study. Second, the
precise location and extent of cortical motor representa-
tions for the left fore- and hindpaw have not been evalu-
ated electrophysiologically, which might contribute to a
relatively high variability of some postlesional gait param-
eters. However, such electrophysiological studies require a
fenestration of the skull, which—on the other hand—
results in a higher burden on animals and thus might
influence the locomotor behavior in the acute phase after
stroke. Third, the chosen coordinates for the photo-
thrombotic infarction also encompass somatosensory cor-
tical areas. Very small lesions result in gait impairments
that are too mild to be measured even with kinematic
analysis (CatWalk). Fourth, all MLR-HFS experiments
were conducted using wired neurostimulation by con-
necting the implanted electrode to an external stimulator.
However, because all experiments (especially the CatWalk
analysis) were performed by 2 investigators, the wire did
not substantially impact the MLR-HFS experiment; ani-
mals subjected to photothrombosis only (ie, not tethered
to a neurostimulator) showed no significant differences
in locomotor parameters when compared to rats connected
to the stimulator via electrical wire (sham-stimulation
Fluri et al: DBS for Gait Recovery
Month 2017 11
experiment, data not shown). It is of note that the resistance
of cerebral tissue varies widely in the phase shortly after
electrode implantation and induction of photothrombosis.
Thus, a stimulator with a large voltage compliance range is
needed to keep a constant current intensity to compare
experiments performed at different time points. The stimu-
lator used in this study is one of the few commercially avail-
able stimulator systems with this property and—to our
knowledge—there is no portable microstimulator for rats
having this feature.
In summary, and despite these uncertainties, this
study provides a novel framework for understanding
stroke-related gait disorder as a network dysfunction
amendable by neuromodulation, and thereby addresses
an as yet unmet clinical need in chronic stroke survivors.
Acknowledgment
This study was supported by the Deutsche Forschungsge-
meinschaft (Sonderforschungsbereich SFB 688) and by
the Interdisziplinares Zentrum fur Klinische Forschung,
University Hospital Wurzburg, Wurzburg, Germany.
We thank L. Frieß and A. Sauer for excellent technical
assistance; and D. Nock for critically proofreading the
manuscript.
Author Contributions
Conception and design of the experiments: F.F., J.V. Per-
formed the experiments: F.F. Analysis of data: all authors.
Processing of MRI data: G.A.H. Wrote the paper: F.F.,
J.V. Critical revision of the manuscript for important
intellectual content: all authors.
Potential Conflicts of Interest
Nothing to report.
References
1. van de Port IGL, Kwakkel G, Schepers VPM, Lindeman E. Predict-
ing mobility outcome one year after stroke: a prospective cohort
study. J Rehabil Med 2006;38:218–223.
2. Titianova EB, Pitk
anen K, P
a
akk
onen A, et al. Gait characteristics
and functional ambulation profile in patients with chronic unilateral
stroke. Am J Phys Med Rehabil 2003;82:778–786.
3. Partridge C, Edwards S. The bases of practice—neurological phys-
iotherapy. Physiother Res Int 1996;1:205–208.
4. Bobath B. Adult hemiplegia: evaluation and treatment. 3rd ed.
Oxford, UK: Oxford Butterworth-Heinemann, 1990.
5. Langhorne P, Coupar F, Pollock A. Motor recovery after stroke: a
systematic review. Lancet Neurol 2009;8:741–754.
6. Duysens J, Van de Crommert HWAA. Neural control of locomo-
tion; the central pattern generator from cats to humans. Gait Pos-
ture 1998;7:131–141.
7. Garcia-Rill E, Hyde J, Kezunovic N, et al. The physiology of the
pedunculopontine nucleus: implications for deep brain stimula-
tion. J Neural Transm 2014;122:225–235.
8. Shik ML, Severin FV, Orlovski K% GN. Control of walking and run-
ning by means of electric stimulation of the midbrain. Biofizika
1966;11:659–666.
9. Wenger N, Moraud EM, Gandar J, et al. Spatiotemporal neuro-
modulation therapies engaging muscle synergies improve motor
control after spinal cord injury. Nat Med 2016;22:138–145.
10. Guzzetta A, Pizzardi A, Belmonti V, et al. Hand movements at 3
months predict later hemiplegia in term infants with neonatal
cerebral infarction. Dev Med Child Neurol 2010;52:767–772.
11. Volkmann J, Allert N, Voges J, et al. Safety and efficacy of pallidal
or subthalamic nucleus stimulation in advanced PD. Neurology
2001;56:548–551.
12. Volkmann J, Wolters A, Kupsch A, et al. Pallidal deep brain stimulation
in patients with primary generalised or segmental dystonia: 5-year fol-
low-up of a randomisedtrial. Lancet Neurol 2012;11:1029–1038.
13. Fluri F, Schuhmann MK, Kleinschnitz C. Animal models of ischemic
stroke and their application in clinical research. Drug Des Devel
Ther 2015;9:3445–3454.
14. Jahn K, Deutschl
ander A, Stephan T, et al. Supraspinal locomotor con-
trol in quadrupeds and humans. Prog Brain Res 2008;171:353–362.
15. Sherman D, Fuller PM, Marcus J, et al. Anatomical location of the
mesencephalic locomotor region and its possible role in locomotion,
posture, cataplexy,and parkinsonism. Front Neurol 2015;6:140.
16. Skinner RD, Garcia-Rill E. The mesencephalic locomotor region
(MLR) in the rat. Brain Res 1984;323:385–389.
17. Cabelguen J-M, Bourcier-Lucas C, Dubuc R. Bimodal locomotion
elicited by electrical stimulation of the midbrain in the salamander
Notophthalmus viridescens. J Neurosci 2003;23:2434–2439.
18. Eidelberg E, Walden JG, Nguyen LH. Locomotor control in
macaque monkeys. Brain 1981;104:647–663.
19. Golestanirad L, Elahi B, Graham SJ, et al. Efficacy and safety of
pedunculopontine nuclei (PPN) deep brain stimulation in the treat-
ment of gait disorders: a meta-analysis of clinical studies. Can J
Neurol Sci 2016;43:120–126.
20. Nosko D, Ferraye MU, Fraix V, et al. Low-frequency versus high-
frequency stimulation of the pedunculopontine nucleus area in
Parkinson’s disease: a randomised controlled trial. J Neurol Neu-
rosurg Psychiatry 2015;86:674–679.
21. Welter M-L, Demain A, Ewenczyk C, et al. PPNa-DBS for gait and
balance disorders in Parkinson’s disease: a double-blind, rando-
mised study. J Neurol 2015;262:1515–1525.
22. Lindau NT, B
anninger BJ, Gullo M, et al. Rewiring of the cortico-
spinal tract in the adult rat after unilateral stroke and anti-Nogo-A
therapy. Brain 2014;137(pt 3):739–756.
23. Paxinos G, Watson C. The rat brain in stereotaxic coordinates. 6th
ed. Amsterdam, the Netherlands: Academic Press Elsevier, 2007.
24. Fluri F, Bieber M, Volkmann J, Kleinschnitz C. Microelectrode
guided implantation of electrodes into the subthalamic nucleus of
rats for long-term deep brain stimulation. J Vis Exp 2015;(104).
25. Bachmann LC, Matis A, Lindau NT, et al. Deep brain stimulation of
the midbrain locomotor region improves paretic hindlimb function
after spinal cord injury in rats. Sci Transl Med 2013;5:208ra146.
26. Matsumura M, Nambu A, Yamaji Y, et al. Organization of somatic
motor inputs from the frontal lobe to the pedunculopontine teg-
mental nucleus in the macaque monkey. Neuroscience 2000;98:
97–110.
27. Hamers FPT, Lankhorst AJ, van Laar TJ, et al. Automated quanti-
tative gait analysis during overground locomotion in the rat: its
application to spinal cord contusion and transection injuries.
J Neurotrauma 2001;18:187–201.
28. Encarnacion A, Horie N, Keren-Gill H, et al. Long-term behavioral
assessment of function in an experimental model for ischemic
stroke. J Neurosci Methods 2011;196:247–257.
ANNALS of Neurology
12 Volume 00, No. 0
29. Parkkinen S, Ortega FJ, Kuptsova K, et al. Gait impairment in a
rat model of focal cerebral ischemia. Stroke Res Treat 2013;2013:
410972.
30. Feeney DM, Gonzalez A, Law WA. Amphetamine, haloperidol,
and experience interact to affect rate of recovery after motor cor-
tex injury. Science 1982;217:855–857.
31. Neckel ND, Dai H, Bregman BS. Quantifying changes following
spinal cord injury with velocity dependent locomotor measures.
J Neurosci Methods 2013;214:27–36.
32. Koopmans GC, Brans M, G
omez-Pinilla F, et al. Circulating
insulin-like growth factor I and functional recovery from spinal
cord injury under enriched housing conditions. Eur J Neurosci
2006;23:1035–1046.
33. Ranck JB Jr. Which elements are excited in electrical stimulation
of mammalian central nervous system: a review. Brain Res 1975;
98:417–440.
34. la Fouge` re C, Zwergal A, Rominger A, et al. Real versus imagined
locomotion: a [18F]-FDG PET-fMRI comparison. Neuroimage
2010;50:1589–1598.
35. Esposito MS, Capelli P, Arber S. Brainstem nucleus MdV mediates
skilled forelimb motor tasks. Nature 2014;508:351–356.
36. Martinez-Gonzalez C, Bolam JP, Mena-Segovia J. Topographical
organization of the pedunculopontine nucleus. Front Neuroanat
2011;5:22.
37. Perry J, Garrett M, Gronley JK, Mulroy SJ. Classification of
walking handicap in the stroke population. Stroke 1995;26:982–
989.
38. Michael KM, Allen JK, Macko RF. Reduced ambulatory activity
after stroke: the role of balance, gait, and cardiovascular fitness.
Arch Phys Med Rehabil 2005;86:1552–1556.
39. Roseberry TK, Lee AM, Lalive AL, et al. Cell-type-specific control
of brainstem locomotor circuits by basal ganglia. Cell 2016;164:
526–537.
40. Takakusaki K, Habaguchi T, Ohtinata-Sugimoto J, et al. Basal gan-
glia efferents to the brainstem centers controlling postural muscle
tone and locomotion: a new concept for understanding motor dis-
orders in basal ganglia dysfunction. Neuroscience 2003;119:293–
308.
41. Wichmann T, DeLong MR. Deep brain stimulation for movement
disorders of basal ganglia origin: restoring function or functional-
ity? Neurotherapeutics 2016;13:264–283.
42. Ryczko D, Dubuc R. The Multifunctional Mesencephalic Locomotor
Region. Curr Pharm Des 2013;19:4448–4470.
43. Gut NK, Winn P. The pedunculopontine tegmental nucleus—a
functional hypothesis from the comparative literature. Mov Disord
2016;31:615–624.
44. Gut NK, Winn P. Deep brain stimulation of different pedunculo-
pontine targets in a novel rodent model of parkinsonism.
J Neurosci 2015;35:4792–4803.
45. Stefani A, Lozano AM, Peppe A, et al. Bilateral deep brain stimu-
lation of the pedunculopontine and subthalamic nuclei in severe
Parkinson’s disease. Brain 2007;130:1596–1607.
46. Zrinzo L, Zrinzo LV, Hariz M. The pedunculopontine and peripe-
duncular nuclei: a tale of two structures. Brain 2007;130:e73.
Fluri et al: DBS for Gait Recovery
Month 2017 13
... In healthy subjects, skilled hand movements are related to coordinated neural networks, which are disturbed after stroke [14]. A promising tool for retuning brain networks is electrical stimulation of distinct brain areas which has been shown to enhance recovery in animal stroke models [60][61][62]. Here we sought to determine the impact of STN-HFS on skilled forelimb reaching and its modulating effect on brain networks in rats undergoing photothrombotic stroke. ...
... Induction of photothrombotic stroke was carried out in rats as described elsewhere [60]. In brief, under deep anesthesia (isoflurane 2.5%), the head of the rat was fixed in a stereotactic frame. ...
... Lesion size was visualized using T2-weighted (T2w) magnetic resonance imaging (MRI) on a 3.0T scanner (MAGNETOM Trio; Siemens, Erlangen Germany) in rats undergoing a 7-day STN-HFS or sham stimulation as reported elsewhere [60]. T2w scans were acquired with turbo spin-echo sequences (echo time, 105 ms, repetition time, 2100 ms) and infarct volume was determined using ImageJ Analysis Software 1.45s (National Institutes of Health, Bethesda, MD, USA. ...
Article
Full-text available
Recovery of upper limb (UL) impairment after stroke is limited in stroke survivors. Since stroke can be considered as a network disorder, neuromodulation may be an approach to improve UL motor dysfunction. Here, we evaluated the effect of high-frequency stimulation (HFS) of the subthalamic nucleus (STN) in rats on forelimb grasping using the single-pellet reaching (SPR) test after stroke and determined costimulated brain regions during STN-HFS using 2-[18F]Fluoro-2-deoxyglucose-([18F]FDG)-positron emission tomography (PET). After a 4-week training of SPR, photothrombotic stroke was induced in the sensorimotor cortex of the dominant hemisphere. Thereafter, an electrode was implanted in the STN ipsilateral to the infarction, followed by a continuous STN-HFS or sham stimulation for 7 days. On postinterventional day 2 and 7, an SPR test was performed during STN-HFS. Success rate of grasping was compared between these two time points. [18F]FDG-PET was conducted on day 2 and 3 after stroke, without and with STN-HFS, respectively. STN-HFS resulted in a significant improvement of SPR compared to sham stimulation. During STN-HFS, a significantly higher [18F]FDG-uptake was observed in the corticosubthalamic/pallidosubthalamic circuit, particularly ipsilateral to the stimulated side. Additionally, STN-HFS led to an increased glucose metabolism within the brainstem. These data demonstrate that STN-HFS supports rehabilitation of skilled forelimb movements, probably by retuning dysfunctional motor centers within the cerebral network.
... [24][25][26][27] On the other hand, the CNF is known to be a main control region for locomotion initiation, maintenance and speed regulation. 23 28 29 Recently, the MLR has gained scientific and clinical interest as target for DBS to improve deficient gait after SCI 16 and stroke 30 with the CNF being proposed as main therapeutic target in recent rodent studies. 23 28 29 Acute electrical activation of the rat MLR has been shown to enable close to physiological hindlimb movements during walking and swimming in a rodent model of chronic incomplete SCI resembling an American Spinal Injury Association (ASIA) Impairment Scale (AIS) D score in humans. ...
... In an acute rodent stroke model, MLR-DBS was able to improve walking speed and limb coordination. 30 DBS in humans is considered safe, reversible and minimally invasive, and is being routinely and successfully applied in the treatment of various movement disorders [31][32][33][34][35][36] with great technical progress in recent years. [37][38][39] While DBS of the PPN in Parkinson's disease has not only yielded clearly positive therapeutic effects, 40 the CNF might be a promising target for locomotion initiation. ...
Article
Full-text available
Introduction Spinal cord injury (SCI) is a devastating condition with immediate impact on the individual’s health and quality of life. Major functional recovery reaches a plateau 3–4 months after injury despite intensive rehabilitative training. To enhance training efficacy and improve long-term outcomes, the combination of rehabilitation with electrical modulation of the spinal cord and brain has recently aroused scientific interest with encouraging results. The mesencephalic locomotor region (MLR), an evolutionarily conserved brainstem locomotor command and control centre, is considered a promising target for deep brain stimulation (DBS) in patients with SCI. Experiments showed that MLR-DBS can induce locomotion in rats with spinal white matter destructions of >85%. Methods and analysis In this prospective one-armed multi-centre study, we investigate the safety, feasibility, and therapeutic efficacy of MLR-DBS to enable and enhance locomotor training in severely affected, subchronic and chronic American Spinal Injury Association Impairment Scale C patients in order to improve functional recovery. Patients undergo an intensive training programme with MLR-DBS while being regularly followed up until 6 months post-implantation. The acquired data of each timepoint are compared with baseline while the primary endpoint is performance in the 6-minute walking test. The clinical trial protocol was written in accordance with the Standard Protocol Items: Recommendations for Interventional Trials checklist. Ethics and dissemination This first in-man study investigates the therapeutic potential of MLR-DBS in SCI patients. One patient has already been implanted with electrodes and underwent MLR stimulation during locomotion. Based on the preliminary results which promise safety and feasibility, recruitment of further patients is currently ongoing. Ethical approval has been obtained from the Ethical Committee of the Canton of Zurich (case number BASEC 2016-01104) and Swissmedic (10000316). Results will be published in peer-reviewed journals and presented at conferences. Trial registration number NCT03053791 .
... In our data, we observed that stereotactic hematoma puncture and drainage not only significantly increased the survival rate of PPH patients but also led to a notable increase in the proportion of patients with moderate to severe disability, thereby improving functional outcomes to some extent. Furthermore, with the development of various rehabilitation techniques, such as neurostimulation therapy showing tremendous potential in enhancing functional outcomes for stroke patients, surviving PPH patients, even those with moderate to severe or profound disability, may benefit from rehabilitation therapy, thereby improving the ultimate clinical outcome [21][22][23]. However, the selection of surgical treatment for PPH remains subjective, lacking an objective scale to predict the outcome of patients after stereotactic hematoma puncture and drainage [24].To address this, we formulated a nomogram to predict the probability of death Moderately severe disability, unable to walk without assistance and unable to attend to own bodily needs without assistance. ...
... There is also evidence for the proposed use of new technologies and techniques, such as deep brain stimulation of midbrain motor centers (mesencephalic motor cortex), as therapeutic strategies to restore motor function after SCIs or stroke provoking plastic changes. Very promising results have been obtained in rodent models with a >80% spinal cord transection, and this technique resulted in acutely functional hindlimb walking and swimming movements [130,131]. The use of stem cellbased regenerative therapy is an innovative approach to inducing a new type of plasticity, which can be produced through SCIs. ...
Article
Full-text available
A spinal cord injury (SCI) causes changes in brain structure and brain function due to the direct effects of nerve damage, secondary mechanisms, and long-term effects of the injury, such as paralysis and neuropathic pain (NP). Recovery takes place over weeks to months, which is a time frame well beyond the duration of spinal shock and is the phase in which the spinal cord remains unstimulated below the level of injury and is associated with adaptations occurring throughout the nervous system, often referred to as neuronal plasticity. Such changes occur at different anatomical sites and also at different physiological and molecular biological levels. This review aims to investigate brain plasticity in patients with SCIs and its influence on the rehabilitation process. Studies were identified from an online search of the PubMed, Web of Science, and Scopus databases. Studies published between 2013 and 2023 were selected. This review has been registered on OSF under (n) 9QP45. We found that neuroplasticity can affect the sensory-motor network, and different protocols or rehabilitation interventions can activate this process in different ways. Exercise rehabilitation training in humans with SCIs can elicit white matter plasticity in the form of increased myelin water content. This review has demonstrated that SCI patients may experience plastic changes either spontaneously or as a result of specific neurorehabilitation training, which may lead to positive outcomes in functional recovery. Clinical and experimental evidence convincingly displays that plasticity occurs in the adult CNS through a variety of events following traumatic or non-traumatic SCI. Furthermore, efficacy-based, pharmacological, and genetic approaches, alone or in combination, are increasingly effective in promoting plasticity.
... Äquivalent zu Versuch A war die Präparation des Tier-Modells kein Bestandteil dieser Doktorarbeit. Für tiefergehende Informationen hinsichtlich der Erzeugung des Schlaganfalls sowie der Implantation der Elektrode wird auf die Literaturangaben[54,58] verwiesen. ...
Thesis
Full-text available
Die präklinische Forschung stellt den ersten wichtigen Meilenstein in der Klärung und Untersuchung klinisch-relevanter Erkrankungen dar. Darüber hinaus unterstützt die präklinische Forschung erheblich die Entwicklung von Therapien. Die Kleintier-Positronenemissionstomographie (µ-PET) spielt dabei eine wichtige Rolle, da sie in der Lage ist, funktionelle, physiologische und biochemische Prozesse in vivo darzustellen und zu quantifizieren. Trotz diverser etablierter PET-Datenauswertungs-Programme bleibt die Analyse von in vivo akquirierten Bilddaten aufgrund der Vielzahl an medizinischen Fragestellungen, der Komplexität der Krankheitsbilder, sowie der Etablierung neuer Radiotracer weiterhin eine große Herausforderung in der Medizin. Ziel dieser Doktorarbeit ist es daher, ein geeignetes, brauchbares Auswertungstool für eine einfache und effiziente Analyse von akquirierten µ-PET-Daten zu entwickeln und zu etablieren, welches das Spektrum bereits vorhandener Programme erweitert. Das entwickelte nuklearmedizinische Datenverarbeitungs-Analyseprogramm (engl. nuclear medicine data processing analysis tool, NU_DPA) wurde in Matlab implementiert und anhand dreier präklinischer Versuchs- bzw. Testreihen erprobt und etabliert. Bei den Datenreihen handelt es sich um µ-PET-Datensätze verschiedener Schlaganfall-Rattenhirnmodelle unter Verwendung folgender Radiotracer. Zum einen die im Gehirn homogen akkumulierende 2-[18F]Fluor-2-desoxy-glukose ([18F]FDG) zum anderen das spezifisch an P-Selektin anreichernde [68Ga]Fucoidan. Das NU_DPA umfasst die automatische Selektion des Zielvolumens (volume-of-interest, VOI) aus dem vollständigen PET-Bild und die anschließende Ausrichtung des VOI mit Hilfe eines PET-Templates (gemittelter PET-Datensatz). Dieses PET Template wird aus den eigenen akquirierten PET-Daten erstellt. Durch das Einbinden eines geeigneten anatomischen MRT-Atlas‘ (anpassbar) können die ausgerichteten PET-Daten einzelnen, Atlas-spezifischen Teilregionen zugeordnet werden. Eine solche Subklassifikation des VOI erlaubt eine genauere Betrachtung und Auswertung der Radiotracer-Akkumulation. Des Weiteren bietet NU_DPA die Möglichkeit einer semiquantitativen Auswertung der PET-Bilddaten anhand von drei unterschiedlichen Parametern, der normalisierten Aktivität, dem Standardized Uptake Value und der Uptake Ratio. Durch die Matlab-integrierten Statistik-Algorithmen ist zusätzlich eine Möglichkeit der statistischen Auswertung der zuvor berechneten Parameter gegeben. Das NU_DPA-Programm stellt somit ein semi-automatisiertes Datenauswertungs-Programm dar, das sowohl die Registrierung als auch die semiquantitative Auswertung von PET-Bilddaten innerhalb einer Versuchsreihe ermöglicht und bereits erfolgreich für die Radiotracer [18F]FDG und [68Ga]Fucoidan in Tiermodellen getestet wurde. Nach derzeitigem Kenntnisstand ist kein Datenauswertungs-Programm bekannt, das PET-Bilddaten unter Verwendung des hinzugefügten Atlas‘ semi-automatisiert analysieren kann und potenziell für homogene und Target-spezifisch akkumulierende Radiotracer geeignet ist.
Article
Full-text available
Ischemic stroke survivors often suffer from severe disability and impaired quality of life, and the current treatments are inadequate. Deep brain stimulation (DBS) is a promising strategy to enhance recovery and alleviate symptoms, as it can modulate the electrical activity of neural circuits and facilitate neuroprotection and regeneration. In this review, we conducted a comprehensive literature search and summarized the chronic sequelae and mechanisms of ischemic stroke. Then we discuss the common targets and outcomes of DBS in preclinical and clinical studies, as well as the challenges and opportunities of DBS for ischemic stroke treatment.
Chapter
Full-text available
Spinal cord injury (SCI) is a debilitating condition that affects millions of people worldwide and results in a remarkable health economic burden imposed on patients and the healthcare system annually. The most common causes of SCI are the trauma caused by falls, traffic accidents, or violence. The course of SCI is associated with several complications that severely impair the patient’s quality of life, including sensory and motor dysfunction, pain, neurogenic bladder and bowel, autonomic dysreflexia, cardiovascular and pulmonary dysfunction, spasticity, urinary tract infection, and sexual dysfunction. Despite great strides that have been made in the field of regenerative medicine and neural repair, the treatment of SCI still mostly revolves around rehabilitative strategies to improve patients’ quality of life and function. Rehabilitation following the SCI is a multidisciplinary process that requires the involvement of multiple disciplines. Moreover, recent advances in the field of neurorehabilitation following SCI, are changing the face of this field. Therefore, we decided to review various aspects of rehabilitation following the SCI, including the goals and different modalities whereby we could achieve them.
Article
The mesencephalic locomotor region (MLR) controls locomotion in vertebrates. In humans with Parkinson disease, locomotor deficits are increasingly associated with decreased activity in the MLR. This brainstem region, commonly considered to include the cuneiform and pedunculopontine nuclei, has been explored as a target for deep brain stimulation to improve locomotor function, but the results are variable, from modest to promising. However, the MLR is a heterogeneous structure, and identification of the best cell type to target is only beginning. Here, I review the studies that uncovered the role of genetically defined MLR cell types, and I highlight the cells whose activation improves locomotor function in animal models of Parkinson disease. The promising cell types to activate comprise some glutamatergic neurons in the cuneiform and caudal pedunculopontine nuclei, as well as some cholinergic neurons of the pedunculopontine nucleus. Activation of MLR GABAergic neurons should be avoided, since they stop locomotion or evoke bouts flanked with numerous stops. The MLR is also considered a potential target in spinal cord injury, supranuclear palsy, primary progressive freezing of gait, or stroke. Better targeting of the MLR cell types should be achieved through optimized deep brain stimulation protocols, pharmacotherapy, or the development of optogenetics for human use.
Article
As a primary nonpharmacological tool, exercise training is neuroprotective after experimental ischemic stroke by relieving neuroinflammation. However, the specific mechanism of which and anti-inflammatory effect of exercise at different intensities require in-depth investigations. To explore the issue, middle cerebral artery occlusion-reperfusion (MCAO-r) in mice were utilized, with subsequent exercise training at different intensities (high-intensity interval training versus moderate-intensity continuous training, i.e. HIIT vs. MICT) during an early phase post-modeling. The neurobehavioral assessment showed that MICT improved the performance of neurological deficit scores and rotarod test earlier, while HIIT appeared to be more efficacious to meliorate locomotor impairments and aerobic fitness at the end of intervention. Both exercise regimens inhibited the expressions of NLRP3 inflammasome components (NLRP3, ASC, and Cl.caspase-1) and pyroptosis-associated proteins (GSDMD, Cl.IL-1β, and Cl.IL-18) as indicated by western blot and immunofluorescence co-staining. Multiplex assay panel revealed that both exercise regimens reduced the levels of pro-inflammatory cytokines and upregulated anti-inflammatory cytokine. Furthermore, an increased proportion of M2-like microglia and a diminished proportion of M1-like microglia in the peri-infarct zone were observed by colocalization analysis, which was jointly validated by western blot. Here, for the first time, our study demonstrated that HIIT elicited better improvements at functional and cardiovascular levels than MICT after ischemic stroke, and anti-inflammatory effect of exercise might result from suppression in inflammasome-mediated pyroptosis by shifting microglial polarization toward neuroprotective M2 phenotype.
Article
Motor deficits after stroke reflect both, focal lesion and network alterations in brain regions distant from infarction. This remote network dysfunction may be caused by aberrant signals from cortical motor regions travelling via mesencephalic locomotor region (MLR) to other locomotor circuits. A method for modulating disturbed network activity is deep brain stimulation. Recently, we have shown that high frequency stimulation (HFS) of the MLR in rats has restored gait impairment after photothrombotic stroke (PTS). However, it remains elusive which cerebral regions are involved by MLR-stimulation and contribute to the improvement of locomotion. Seventeen male Wistar rats underwent photothrombotic stroke of the right sensorimotor cortex and implantation of a microelectrode into the right MLR. 2-[¹⁸F]Fluoro-2-deoxyglucose ([¹⁸F]FDG)-positron emission tomography (PET) was conducted before stroke and thereafter, on day 2 and 3 after stroke, without and with MLR-HFS, respectively. [¹⁸F]FDG-PET imaging analyses yielded a reduced glucose metabolism in the right cortico-striatal thalamic loop after PTS compared to the state before intervention. When MLR-HFS was applied after PTS, animals exhibited a significantly higher uptake of [¹⁸F]FDG in the right but not in the left cortico-striatal thalamic loop. Furthermore, MLR-HFS resulted in an elevated glucose metabolism of right-sided association cortices related to the ipsilateral sensorimotor cortex. These data support the concept of diaschisis i.e., of dysfunctional brain areas distant to a focal lesion and suggests that MLR-HFS can reverse remote network effects following PTS in rats which otherwise may result in chronic motor symptoms.
Article
Full-text available
Background: Pedunculopontine nucleus (PPN) has complex reciprocal connections with basal ganglia, especially with internal globus pallidus and substantia nigra, and it has been postulated that PPN stimulation may improve gait instability and freezing of gait. In this meta-analysis, we will assess the evidence for PPN deep brain stimulation in treatment of gait and motor abnormalities especially focusing on Parkinson disease patients. Methods: PubMed and Scopus electronic databases were searched for related studies published before February 2014. Medline (1966-2014), Embase (1974-2010), CINAHL, Web of Science, Scopus bibliographic, and Google Scholar databases (1960-2014) were also searched for studies investigating effect of PPN deep brain stimulation in treatment of postural and postural instability and total of ten studies met the inclusion criteria for this analysis. Results: Our findings showed a significant improvement in postural instability (p<0.001) and motor symptoms of Parkinson disease on and off medications (p<0.05), but failed to show improvement in freezing of gait. Conclusions: Despite significant improvement in postural instability observed in included studies, evidence from current literature is not sufficient to generalize these findings to the majority of patients.
Article
Full-text available
Deep brain stimulation (DBS) is a widely used and effective therapy for several neurologic disorders, such as idiopathic Parkinson's disease, dystonia or tremor. DBS is based on the delivery of electrical stimuli to specific deep anatomic structures of the central nervous system. However, the mechanisms underlying the effect of DBS remain enigmatic. This has led to an interest in investigating the impact of DBS in animal models, especially in rats. As DBS is a long-term therapy, research should be focused on molecular-genetic changes of neural circuits that occur several weeks after DBS. Long-term DBS in rats is challenging because the rats move around in their cage, which causes problems in keeping in place the wire leading from the head of the animal to the stimulator. Furthermore, target structures for stimulation in the rat brain are small and therefore electrodes cannot easily be placed at the required position. Thus, a set-up for long-lasting stimulation of rats using platinum/iridium electrodes with an impedance of about 1 MΩ was developed for this study. An electrode with these specifications allows for not only adequate stimulation but also recording of deep brain structures to identify the target area for DBS. In our set-up, an electrode with a plug for the wire was embedded in dental cement with four anchoring screws secured onto the skull. The wire from the plug to the stimulator was protected by a stainless-steel spring. A swivel was connected to the circuit to prevent the wire from becoming tangled. Overall, this stimulation set-up offers a high degree of free mobility for the rat and enables the head plug, as well as the wire connection between the plug and the stimulator, to retain long-lasting strength.
Article
Full-text available
This review outlines the most frequently used rodent stroke models and discusses their strengths and shortcomings. Mimicking all aspects of human stroke in one animal model is not feasible because ischemic stroke in humans is a heterogeneous disorder with a complex pathophysiology. The transient or permanent middle cerebral artery occlusion (MCAo) model is one of the models that most closely simulate human ischemic stroke. Furthermore, this model is characterized by reliable and well-reproducible infarcts. Therefore, the MCAo model has been involved in the majority of studies that address pathophysiological processes or neuroprotective agents. Another model uses thromboembolic clots and thus is more convenient for investigating thrombolytic agents and pathophysiological processes after thrombolysis. However, for many reasons, preclinical stroke research has a low translational success rate. One factor might be the choice of stroke model. Whereas the therapeutic responsiveness of permanent focal stroke in humans declines significantly within 3 hours after stroke onset, the therapeutic window in animal models with prompt reperfusion is up to 12 hours, resulting in a much longer action time of the investigated agent. Another major problem of animal stroke models is that studies are mostly conducted in young animals without any comorbidity. These models differ from human stroke, which particularly affects elderly people who have various cerebrovascular risk factors. Choosing the most appropriate stroke model and optimizing the study design of preclinical trials might increase the translational potential of animal stroke models.
Article
Full-text available
The mesencephalic (or midbrain) locomotor region (MLR) was first described in 1966 by Shik and colleagues, who demonstrated that electrical stimulation of this region induced locomotion in decerebrate (intercollicular transection) cats. The pedunculopontine tegmental nucleus (PPT) cholinergic neurons and midbrain extrapyramidal area (MEA) have been suggested to form the neuroanatomical basis for the MLR, but direct evidence for the role of these structures in locomotor behavior has been lacking. Here, we tested the hypothesis that the MLR is composed of non-cholinergic spinally projecting cells in the lateral pontine tegmentum. Our results showed that putative MLR neurons medial to the PPT and MEA in rats were non-cholinergic, glutamatergic, and express the orexin (hypocretin) type 2 receptors. Fos mapping correlated with motor behaviors revealed that the dorsal and ventral MLR are activated, respectively, in association with locomotion and an erect posture. Consistent with these findings, chemical stimulation of the dorsal MLR produced locomotion, whereas stimulation of the ventral MLR caused standing. Lesions of the MLR (dorsal and ventral regions together) resulted in cataplexy and episodic immobility of gait. Finally, trans-neuronal tracing with pseudorabies virus demonstrated disynaptic input to the MLR from the substantia nigra via the MEA. These findings offer a new perspective on the neuroanatomic basis of the MLR, and suggest that MLR dysfunction may contribute to the postural and gait abnormalities in Parkinsonism.
Article
Deep brain stimulation (DBS) is highly effective for both hypo- and hyperkinetic movement disorders of basal ganglia origin. The clinical use of DBS is, in part, empiric, based on the experience with prior surgical ablative therapies for these disorders, and, in part, driven by scientific discoveries made decades ago. In this review, we consider anatomical and functional concepts of the basal ganglia relevant to our understanding of DBS mechanisms, as well as our current understanding of the pathophysiology of two of the most commonly DBS-treated conditions, Parkinson's disease and dystonia. Finally, we discuss the proposed mechanism(s) of action of DBS in restoring function in patients with movement disorders. The signs and symptoms of the various disorders appear to result from signature disordered activity in the basal ganglia output, which disrupts the activity in thalamocortical and brainstem networks. The available evidence suggests that the effects of DBS are strongly dependent on targeting sensorimotor portions of specific nodes of the basal ganglia-thalamocortical motor circuit, that is, the subthalamic nucleus and the internal segment of the globus pallidus. There is little evidence to suggest that DBS in patients with movement disorders restores normal basal ganglia functions (e.g., their role in movement or reinforcement learning). Instead, it appears that high-frequency DBS replaces the abnormal basal ganglia output with a more tolerable pattern, which helps to restore the functionality of downstream networks.
Article
We present data from animal studies showing that the pedunculopontine tegmental nucleus-conserved through evolution, compartmentalized, and with a complex pattern of inputs and outputs-has functions that involve formation and updates of action-outcome associations, attention, and rapid decision making. This is in contrast to previous hypotheses about pedunculopontine function, which has served as a basis for clinical interest in the pedunculopontine in movement disorders. Current animal literature points to it being neither a specifically motor structure nor a master switch for sleep regulation. The pedunculopontine is connected to basal ganglia circuitry but also has primary sensory input across modalities and descending connections to pontomedullary, cerebellar, and spinal motor and autonomic control systems. Functional and anatomical studies in animals suggest strongly that, in addition to the pedunculopontine being an input and output station for the basal ganglia and key regulator of thalamic (and consequently cortical) activity, an additional major function is participation in the generation of actions on the basis of a first-pass analysis of incoming sensory data. Such a function-rapid decision making-has very high adaptive value for any vertebrate. We argue that in developing clinical strategies for treating basal ganglia disorders, it is necessary to take an account of the normal functions of the pedunculopontine. We believe that it is possible to use our hypothesis to explain why pedunculopontine deep brain stimulation used clinically has had variable outcomes in the treatment of parkinsonism motor symptoms and effects on cognitive processing. © 2016 International Parkinson and Movement Disorder Society.
Article
The basal ganglia (BG) are critical for adaptive motor control, but the circuit principles underlying their pathway-specific modulation of target regions are not well understood. Here, we dissect the mechanisms underlying BG direct and indirect pathway-mediated control of the mesencephalic locomotor region (MLR), a brainstem target of BG that is critical for locomotion. We optogenetically dissect the locomotor function of the three neurochemically distinct cell types within the MLR: glutamatergic, GABAergic, and cholinergic neurons. We find that the glutamatergic subpopulation encodes locomotor state and speed, is necessary and sufficient for locomotion, and is selectively innervated by BG. We further show activation and suppression, respectively, of MLR glutamatergic neurons by direct and indirect pathways, which is required for bidirectional control of locomotion by BG circuits. These findings provide a fundamental understanding of how BG can initiate or suppress a motor program through cell-type-specific regulation of neurons linked to specific actions.
Article
Electrical neuromodulation of lumbar segments improves motor control after spinal cord injury in animal models and humans. However, the physiological principles underlying the effect of this intervention remain poorly understood, which has limited the therapeutic approach to continuous stimulation applied to restricted spinal cord locations. Here we developed stimulation protocols that reproduce the natural dynamics of motoneuron activation during locomotion. For this, we computed the spatiotemporal activation pattern of muscle synergies during locomotion in healthy rats. Computer simulations identified optimal electrode locations to target each synergy through the recruitment of proprioceptive feedback circuits. This framework steered the design of spatially selective spinal implants and real-time control software that modulate extensor and flexor synergies with precise temporal resolution. Spatiotemporal neuromodulation therapies improved gait quality, weight-bearing capacity, endurance and skilled locomotion in several rodent models of spinal cord injury. These new concepts are directly translatable to strategies to improve motor control in humans.