Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

brain research ] (]]]]) ]]]–]]]

Available online at www.sciencedirect.com

www.elsevier.com/locate/brainres

Research Report

Therapeutic effects of repetitive transcranial


magnetic stimulation in an animal model of
Parkinson's disease

Ji Yong Leea,b,c,1, Sung Hoon Kimd,1, Ah-Ra Koe, Jin Suk Leeb, Ji Hea Yua,f,
Jung Hwa Seoa,g, Byung Pil Chob,h,n, Sung-Rae Choa,f,g,i,j,nn
a
Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea
b
Department of Anatomy, Yonsei University Wonju College of Medicine, Wonju, South Korea
c
Graduate School of Medicine, Yonsei University, Seoul, South Korea
d
Department of Rehabilitation Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea
e
Life and Health Science, UL, South Korea
f
Brain Korea 21 plus Project for Medical Science, Yonsei University, Seoul, South Korea
g
Graduate Program of Nano Science and Technology, Yonsei University, Seoul, South Korea
h
Institute of Lifestyle Medicine, Yonsei University Wonju College of Medicine, Wonju South Korea
i
Yonsei Stem Cell Research Center, Avison Biomedical Research Center, Seoul, South Korea
j
Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, South Korea

art i cle i nfo ab st rac t

Article history: Repetitive transcranial magnetic stimulation (rTMS) is used to treat neurological diseases
Accepted 26 August 2013 such as stroke and Parkinson's disease (PD). Although rTMS has been used clinically, its
underlying therapeutic mechanism remains unclear. The objective of the present study was

Keywords: to clarify the neuroprotective effect and therapeutic mechanism of rTMS in an animal model

Repetitive transcranial magnetic of PD. Adult Sprague-Dawley rats were unilaterally injected with 6-hydroxydopamine

stimulation (6-OHDA) into the right striatum. Rats with PD were then treated with rTMS (circular coil,

Parkinson's disease 10 Hz, 20 min/day) daily for 4 weeks. Behavioral assessments such as amphetamine-induced

Dopaminergic neuron rotational test and treadmill locomotion test were performed, and the dopaminergic (DA)

Neurotrophic/growth factor neurons of substantia nigra pas compacta (SNc) and striatum were histologically examined.
Expression of neurotrophic/growth factors was also investigated by multiplex ELISA, western
blotting analysis and immunohistochemistry 4 weeks after rTMS application. Among the
results, the number of amphetamine-induced rotations was significantly lower in the rTMS
group than in the control group at 4 weeks post-treatment. Treadmill locomotion was also

n
Corresponding author at: Institute of Lifestyle Medicine, Department of Anatomy, Yonsei University Wonju College of Medicine, 172,
Ilsan-dong, Wonju 220-702, South Korea.
nn
Corresponding author at: Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, 134
Shinchon-dong, Seodaemun-gu, Seoul 120-752, South Korea. Fax: þ82 2 363 2795.
E-mail addresses: bpcho@yonsei.ac.kr (B.P. Cho), srcho918@yuhs.ac (S.-R. Cho).
1
Both authors contributed equally to this work.

0006-8993/$ - see front matter & 2013 Published by Elsevier B.V.


http://dx.doi.org/10.1016/j.brainres.2013.08.051

Please cite this article as: Lee, J.Y., et al., Therapeutic effects of repetitive transcranial magnetic stimulation in an animal
model of Parkinson's disease. Brain Research (2013), http://dx.doi.org/10.1016/j.brainres.2013.08.051
2 brain research ] (]]]]) ]]]–]]]

significantly improved in the rTMS-treated rats. Tyrosine hydroxylase-positive DA neurons


and DA fibers in rTMS group rats were greater than those in untreated group in both
ipsilateral SNc and striatum, respectively. The expression levels of brain-derived neuro-
trophic factor, glial cell line-derived neurotrophic factor, platelet-derived growth factor, and
vascular endothelial growth factor were elevated in both the 6-OHDA-injected hemisphere
and the SNc of the rTMS-treated rats. In conclusion, rTMS treatment improved motor
functions and survival of DA neurons, suggesting that the neuroprotective effect of rTMS
treatment might be induced by upregulation of neurotrophic/growth factors in the PD
animal model.
& 2013 Published by Elsevier B.V.

1. Introduction rTMS treatment, the present study investigated motor func-


tions and tyrosine hydroxylase (TH)-immunoreactive DA
Parkinson's disease (PD) occurs as a result of degenerating neurons and DA nerve fibers in the SNc and striatum. In
cell death of dopaminergic (DA) neurons in the substantia addition, the expression of neurotrophic/growth factors was
nigra pars compacta (SNc). PD is characterized by certain examined after rTMS application in an animal model of PD.
motor disturbances such as resting tremor, bradykinesia, and
gait disturbance. Dopamine replacement therapy is an effec-
tive medical treatment for the symptomatic improvement of
PD. In spite of such effects, fluctuations in abnormal invo-
2. Results
luntary movement eventually occur in most patients after
long-term dopaminergic treatment. Consequently, several
2.1. Effects of rTMS on amphetamine-induced rotation test
studies have investigated various methods other than dopa-
minergic drugs for PD treatment (Wu et al., 2008).
Adult Sprague-Dawley rats were unilaterally injected with 6-
Repetitive transcranial magnetic stimulation (rTMS) is a
hydroxydopamine (6-OHDA) into the right striatum. Two
non-invasive therapeutic device that can alter the excitability
weeks after 6-OHDA injection, rats with PD were treated with
of the cerebral cortex or neural network. rTMS has been used
rTMS (circular coil, 10 Hz, 20 min/day) daily for 4 weeks
to treat neuropsychiatric diseases (Zwanzger et al., 2002;
(Fig. 1A–C). Functional benefit in rTMS-treated rats on
Bentwich et al., 2011; Fitzgerald and Daskalakis, 2011) and
amphetamine-induced rotation behavior was evaluated up
stroke (Theilig et al., 2011; Corti et al., 2012). Recently, several
to 4 weeks after treatment (n¼ 12 each). Whereas untreated
clinical trials have revealed that rTMS offers therapeutic
group showed continuous augmentation in rotational beha-
benefit for functional recovery in PD (Siebner et al., 1999;
vior, the increasing rate of amphetamine-induced rotations
Arias-Carrión, 2008; Wu et al., 2008; Yang et al., 2013). rTMS
was significantly lowered in rTMS-treated rats at 4 weeks
treatment has shown to improve motor function and cogni-
after treatment (Fig. 2A). The number of rotations was not
tion in patients with PD (Zamir et al., 2012). Additionally,
significantly different between the rTMS group (387.3743.4)
increased DA levels in serum and subcortical areas have been
and the untreated group (319.2741.3) at 2 weeks post-
observed following rTMS in both PD patients and experimen-
treatment. However, the number of rotations in the rTMS
tal animals (Strafella et al., 2001, 2003; Khedr et al., 2007;
group (386.9743.8) was significantly lower than that in the
Choand Strafella, 2009).
untreated group (549.9733.6) at 4 weeks post-treatment
On the other hand, the underlying mechanism of rTMS (F ¼5.52, p¼ 0.03).
still remains to be elucidated. Recently, several experiments
have shown that rTMS has the ability to mediate neuroplas-
ticity by enhancing the expressions of glutamate neurotrans-
mitters and brain-derived neurotrophic factor (BDNF) in rat 2.2. rTMS improved locomotor function in a PD animal
brains (Müller et al., 2000; Keck et al., 2000; Yue et al., 2009). model
rTMS did not only activate brain regions in terms of immedi-
ate early gene expression, but also increased the expression Treadmill locomotion test was assessed to determine the
of BDNF-TrkB signaling in rats and humans (Ji et al., 1998; effects of the DA lesion on motor functions (n¼ 12 each).
Hausmann et al., 2000; Doi et al., 2001; Wang et al., 2011). The rTMS-treated rats showed improvement in locomotor
Additionally, rTMS modulated neurotrophic factors such as function 2 weeks after treatment (280725.0 mm), compared
BDNF, cholecystokinin and neuropetide tyrosine in healthy with the untreated group (225.1734.1 mm) (Fig. 2B). The
humans and patients with depression and amyotrophic running distance of rTMS-treated rats continuously increased
lateral sclerosis (Angelucci et al., 2004; Yukimasa et al., to a final score of 320.2723.1 mm throughout the treatment
2006; Gedge et al., 2012). However, there are few rTMS studies period, while the distance of untreated groups maintained to
on the effects of various neurotrophic/growth factors in PD. the score of 230735.5 mm at 4 weeks post-treatment (F¼ 3.56,
To clarify the neuroprotective effect of rTMS on SNc DA p ¼0.04), implying that rTMS induced motor improvements in
neurons and the therapeutic mechanism in overall brain after the PD rats.

Please cite this article as: Lee, J.Y., et al., Therapeutic effects of repetitive transcranial magnetic stimulation in an animal
model of Parkinson's disease. Brain Research (2013), http://dx.doi.org/10.1016/j.brainres.2013.08.051
brain research ] (]]]]) ]]]–]]] 3

6-OHDA
8 weeks 14 weeks
old rTMS old

sacrifice

SNc
ST
MFB
10 days 14 days 2 weeks 4 weeks
Behavioral Behavioral
test test
Amphetamine
6-OHDA -induced
injection rotation test

rTMS treatment

SNc
ST
MFB

Fig. 1 – Experimental design. (A) Schematic diagram of the experimental schedule. PD in rats was confirmed by amphetamine-
induced rotation test 10 days after 6-OHDA injection. In rats with PD, rTMS was treated 2 weeks after 6-OHDA injection, and
behavioral tests were then performed at 2 week intervals. (B) Schematic figure illustrating injection of 6-OHDA into the striatum
using a stereotaxic device. (C) Schematic figure of rTMS treatment on the rat brain. rTMS—repetitive transcranial magnetic
stimulation, 6-OHDA—6-hydroxydopamine, ST—striatum, MFB—medial forebrain bundle and SNc—substantia nigra pars compacta.

Amphetamine-induced Treadmill locomotion


rotation test test
800 400 *
Number of rotations (turns)

Untreated Untreated
Nose tip position (mm)

rTMS * rTMS
600 300

400 200

200 100

0 0
Pretreatment 2 weeks 4 weeks Pretreatment 2 weeks 4 weeks
Post-treatment Post-treatment

Fig. 2 – Behavioral assessment after rTMS treatment in PD rats. (A) Amphetamine-induced rotation test. Rotational behavior in
response to amphetamine (5 mg/kg) was tested before stimulation (pretreatment, 10 days after 6-OHDA injection), 2 weeks
and 4 weeks after rTMS stimulation (n ¼12 each). The increasing rate of amphetamine-induced rotations was significantly
lowered in rTMS-treated rats compared to that in the untreated rats 4 weeks after treatment (*po0.05 compared with
untreated group). (B) Treadmill locomotion test. Data show the distance between the position of rat's nose tip and the rear wall
of treadmill chamber during 20-s treadmill walking periods. The average distance markedly increased after rTMS stimulation,
indicating increased locomotion ability (*po0.05 compared with untreated group). Untreated—PD animals without rTMS
treatments, rTMS—PD animals treated with rTMS.

2.3. Neuroprotective effect of rTMS on DA neurons in the unilateral 6-OHDA injection, the number of THþ neurons
SNc decreased compared to the corresponding contralateral
(non-lesioned) SNc (Table 1). Four weeks after treatment of
The therapeutic effect of rTMS on DA neurons in SNc was rTMS, the number of THþ neurons (210710.0) in the ipsilat-
assessed by immunohistochemistry (n¼ 5 each). Following eral (lesioned) SNc was significantly higher than the number

Please cite this article as: Lee, J.Y., et al., Therapeutic effects of repetitive transcranial magnetic stimulation in an animal
model of Parkinson's disease. Brain Research (2013), http://dx.doi.org/10.1016/j.brainres.2013.08.051
4 brain research ] (]]]]) ]]]–]]]

in untreated group (14477.5) (t¼ 5.34, p ¼0.01) (Fig. 3A, B), 2.4. Neuroprotective effect of rTMS on DA efferents in the
whereas the number of THþ neurons in the contralateral SNc striatum
was not different between the untreated group and rTMS-
treated group (Table 1). In addition, western blotting analysis The therapeutic effect of rTMS on the striatal DA efferent fibers
demonstrated that the expression of TH increased in the was evaluated by striatal TH immunohistochemistry. Repre-
ipsilateral hemisphere 4 weeks after rTMS treatment (Fig. 3C). sentative findings of striatal TH staining in the ipsilateral
The results suggest that rTMS treatment might preserve or hemispheres were observed compared with those in the con-
slow DA neuronal degeneration from 6-OHDA administration. tralateral hemispheres (Fig. 3D). Expressing volume values as

Table 1 – THþ dopaminergic neurons in the SNc at 4 weeks after rTMS treatment.

Contralateral side Ipsilateral side Survival rate (%)

Untreated (n ¼ 5) 1163.4743.5 144.077.5 12.3870.4


rTMS (n ¼ 5) 1205.6741.6 210.879.9* 17.4471.4*

Note: Values are mean7SEM. Survival rate (%) is the percentage of THþ neurons in the ipsilateral SNc relative to those in the contralateral SNc.
TH ¼tyrosine hydroxylase; SNc ¼substantia nigra pars compacta; Untreated ¼ PD animals without rTMS treatments; rTMS ¼ PD animals treated
with rTMS.
n
po0.05 compared with untreated group.

Contralateral Ipsilateral Ipsilateral SNc


250
Untreated rTMS

TH + neurons (number)
200
*
150
Rostral

100

50

0
Untreated rTMS
Caudal

Normal PD
SNc Untreated rTMS
VTA
TH (60 kD)

GAPDH (37 kD)


Contralateral Ipsilateral
Untreated

Ipsilateral Striatum
ST 30
**
TH+ fibers (%)

20

10
rTMS

ST
0
Untreated rTMS

Fig. 3 – THþ neurons after rTMS treatment in PD rats. (A) Representative findings of TH staining in the coronal sections passing
through the rostral and caudal parts of the SNc 4 weeks after rTMS treatment. (B) The number of THþ neurons in the ipsilateral
SNc (n ¼5 each). The total number of THþ neurons of the ipsilateral SNc was significantly greater in the rTMS treatment group
(npo0.05). (C) TH expressions of a normal control, untreated group, and rTMS group in the ipsilateral hemisphere by western
blot analysis. (D) Representative findings of striatal TH staining in the ipsilateral striatum at 4 weeks after rTMS treatment.
(E) The volume of striatal THþ fibers (%) relative to contralateral striatum. Dopaminergic THþ fibers were significantly higher in
the rTMS treatment group than in the untreated group (nnpo0.01). Untreated—PD animals without rTMS treatments, rTMS—
PD animals treated with rTMS, SNc—substantia nigra pars compacta, VTA—ventral tegmental area, TH—tyrosine
hydroxylase, Normal—naïve animal without rTMS treatment, ST—striatum.

Please cite this article as: Lee, J.Y., et al., Therapeutic effects of repetitive transcranial magnetic stimulation in an animal
model of Parkinson's disease. Brain Research (2013), http://dx.doi.org/10.1016/j.brainres.2013.08.051
brain research ] (]]]]) ]]]–]]] 5

the percentage of THþ fibers in the ipsilateral striatum relative neurotrophic factor (GDNF), NGF, PDGF, and VEGF (n¼5 each)
to those in the contralateral striatum, the volume of dopami- were investigated in both hemispheres by western blotting
nergic THþ fibers was significantly higher in the rTMS group analysis. Expressions of neurotrophic/growth factors BDNF
(2277.3%) than in the untreated group (2.070.5%) at 4 weeks (t¼ 3.14, p¼0.013), GDNF (t¼ 2.28, p¼0.02), NGF (t¼ 3.79,
post-treatment (t¼4.73, p¼ 0.003) (Fig. 3E). The results suggest p¼0.04), PDGF (t¼ 6.15, p¼0.008) and VEGF (t¼3.02, p¼ 0.02)
that more striatal THþ efferent fibers in concurrence with more significantly increased in the rTMS-treated rats compared with
THþ neurons in the SNc, relative to the untreated group, could those in untreated group (Fig. 5A–E). The expressions of
improve motor performance following rTMS treatment. neurotrophic/growth factors exhibited a similar pattern to
the multiplex ELISA results in the ipsilateral hemispheres.
On the other hand, the expressions of neurotrophic/growth
2.5. Effects of rTMS on the expression of neurotrophic/
factors in the contralateral hemispheres were not significantly
growth factors
increased at 4 weeks after treatment (Fig. 5A–E).

To identify the growth factors associated with the repair


process of functional recovery and the neuroprotective and/ 2.6. Effects of rTMS on the expression of neurotrophic/
or neurorestorative effects induced by rTMS, the expressions growth factors in the SNc
of various neurotrophic/growth factors including nerve
growth factor (NGF), ciliary neurotrophic factor (CNTF), To identify the effect of rTMS on the expression of neuro-
platelet-derived growth factor AA (PDGF-AA), and vascular trophic/growth factors in the SNc, the expression levels of
endothelial growth factor (VEGF) were investigated in ipsilat- BDNF, GDNF, NGF, PDGF and VEGF were performed using
eral hemispheres by multiplex ELISA assay (n ¼3 each). immunohistochemistry in the ipsilateral SNc at 4 weeks after
Among the factors, NGF (t ¼3.79, p ¼0.03), PDGF (t¼3.12, treatment (n¼ 4 each). Representative findings of BDNF,
p¼ 0.009) and VEGF (t¼ 2.59, p¼ 0.02) levels were significantly GDNF, PDGF, and VEGF staining in the ipsilateral SNc after
elevated in the 6-OHDA-injected hemispheres of rTMS group rTMS treatment were shown in comparison with untreated
compared with those of the untreated groups at 4 weeks after group (Fig. 6A). According to the density of neurotrophic/
treatment, although the change in CNTF expression was not growth factorsþ cells (%), calculated as the immunoreactive
statistically significant (Fig. 4A–D). cell area (/mm2) divided by the SNc area (/mm2), the expres-
To confirm the expressions of neurotrophic/growth factors, sions of BDNF (t¼ 4.54, p¼ 0.04), GDNF (t ¼3.03, p¼ 0.03), PDGF
brain derived neurotrophic factor (BDNF), glial cell line-derived (t¼2.81, p¼ 0.02), and VEGF (t¼2.34, p¼ 0.042, n ¼5) were

80
NGF PDGF
400
* **
PDGF-AA (ρg/ml)
β-NGF (ρg/ml)

60 300

40 200

20 100

0 0
Untreated rTMS Untreated rTMS

VEGF CNTF
4 400

*
VEGF (ρg/ml)

CNTF (ρg/ml)

3 300

2 200

1 100

0 0
Untreated rTMS Untreated rTMS

Fig. 4 – Neurotrophic/growth factors by multiplex ELISA assay. (A–D) The expression levels of neurotrophic/growth factors
were increased in the rTMS treatment group compared with the untreated group (n ¼3 each). The levels of NGF (A), PDGF-AA
(B), and VEGF (C) were significantly higher in the rTMS group than in the untreated group at 4 weeks post-treatment. Change
in CNTF expression (D) was not statistically significant (npo0.05, nnpo0.01). Untreated—PD animals without rTMS treatments,
rTMS—PD animals treated with rTMS, NGF—nerve growth factor, PDGF—platelet-derived growth factor, VEGF—vascular
endothelial growth factor, CNTF—ciliary neurotrophic factor.

Please cite this article as: Lee, J.Y., et al., Therapeutic effects of repetitive transcranial magnetic stimulation in an animal
model of Parkinson's disease. Brain Research (2013), http://dx.doi.org/10.1016/j.brainres.2013.08.051
6 brain research ] (]]]]) ]]]–]]]

BDNF GDNF
3 2.5
Untreated Untreated
rTMS * 2.0 rTMS *

Relative Index
Relative Index
Contralateral Ipsilateral 2
1.5
Untreated rTMS Untreated rTMS
1.0
1
BDNF(28kD) 0.5

0 0.0
Contralateral Ipsilateral Contralateral Ipsilateral

GDNF(21kD)
NGF PDGF
2.0 5
Untreated Untreated **
rTMS rTMS
NGF(27kD) 4

Relative Index

Relative Index
1.5 *
3
1.0
2
PDGF(23kD) 0.5
1

0.0 0
Contralateral Ipsilateral Contralateral Ipsilateral
VEGF(43kD)

VEGF
4
Untreated
GAPDH(37kD) rTMS *
Relative Index

0
Contralateral Ipsilateral

Fig. 5 – Neurotrophic/growth factors by western blotting analysis. (A) Representative expressions of BDNF, GDNF, NGF, PDGF
and VEGF in both contralateral and ipsilateral hemispheres. (B–F) The relative amounts of neurotrophic/growth factors (n ¼5
each). The expression levels of BDNF (B), GDNF (C), NGF (D), PDGF (E) and VEGF (F) were significantly increased in the rTMS
group (npo0.05, nnpo0.01). Untreated—PD animals without rTMS treatments, rTMS—PD animals treated with rTMS, BDNF—
brain-derived neurotrophic factor, GDNF—glial cell-derived neurotrophic factor, NGF—nerve growth factor, PDGF—platelet-
derived growth factor, VEGF—vascular endothelial growth factor.

significantly increased in the rTMS group compared with the modulation of various trophic factors by rTMS treatment in
untreated group (Fig. 6B). On the other hand, NGFþ cell PD rats.
density was not increased after rTMS treatment (data not We demonstrated that rTMS elicited neuroprotective
shown). Taken together, these results suggest that the upre- effects on functional outcomes in the amphetamine rota-
gulation of various neurotrophic/growth factors in rTMS- tional test and treadmill locomotion test. Namely, rTMS
treated hemispheres and SNc might preserve DA neuronal treatment alleviated unilateral 6-OHDA induced rotation 4
damage, consequently inducing functional recovery in rTMS- weeks after treatment. This result was consistent with a
treated PD rats. previous study that demonstrated that the number of rota-
tions was reduced by low-frequency (0.5 Hz) rTMS treatment
(Yang et al., 2010). Treadmill locomotion test also revealed
that rTMS has the capability to improve reciprocal locomo-
3. Discussion tion in unilateral DA-lesioned rats (Pellis et al., 1987; Johnson
et al., 1999; Lee et al., 2012). Treadmill locomotion test
rTMS has been shown to modulate cortical network and nonspecifically reflects cortical dysfunction as well as dys-
corticomotor excitability, and to improve motor function in function of basal ganglia loops including striatal dopamine
patients with PD (Fisher et al., 2008; Yang et al., 2013). depletion (Chang et al., 2003), and can immediately control
However, this study focused on the alterations of neuro- not only basal ganglia loops but also cortical networks (Arias-
trophic/growth factors after rTMS in a PD animal model Carrion et al., 2011). On the other hand, amphetamine-
rather than modulation of the cortical network. This study induced rotation results from a persistent and consistent
showed the therapeutic potential of rTMS to improve motor one side preference, and is thought to be mediated by
functions and to preserve DA neuronal survival by upregula- lateralized functioning of the dopaminergic nigrostriatal
tion of neurotrophic/growth factors in a 6-OHDA-lesioned PD pathways in brain. Amphetamine rotation preferences of
condition. This might be the first report to demonstrate unilateral PD rats have been shown to be related to not only

Please cite this article as: Lee, J.Y., et al., Therapeutic effects of repetitive transcranial magnetic stimulation in an animal
model of Parkinson's disease. Brain Research (2013), http://dx.doi.org/10.1016/j.brainres.2013.08.051
brain research ] (]]]]) ]]]–]]] 7

BDNF GDNF PDGF VEGF


Untreated
rTMS

BDNF GDNF PDGF VEGF


40 10
30 80
* *
*

Density in SNc (%)


* 8
Density in SNc (%)

Density in SNc (%)

Density in SNc (%)

30 60
20 6
20 40
4
10
10 20 2

0 0 0 0
Untreated rTMS Untreated rTMS Untreated rTMS Untreated rTMS

Fig. 6 – Neurotrophic/growth factors in the ipsilateral SNc. (A) Representative findings of immunoreactivity with BDNF, GDNF,
PDGF, and VEGF in ipsilateral SNc at 4 weeks after rTMS treatment. (B) The density of neurotrophic/growth factors-
immunoreactive cells (%) (n ¼ 4 each). Expressions of BDNF, GDNF and PDGF were significantly increased in the rTMS
treatment group compared with the untreated group (npo0.05). Untreated—PD animals without rTMS treatments, rTMS—PD
animals treated with rTMS, BDNF—brain-derived neurotrophic factor, GDNF—glial cell-derived neurotrophic factor, NGF—
nerve growth factor, PDGF—platelet-derived growth factor, VEGF—vascular endothelial growth factor.

endogenous asymmetries in striatal dopamine content but also Our results showed that THþ fibers were primarily increased
survived DA neurons on SNc or DA nerve terminals in striatum in the ventromedial striatum rather than dorsolateral striatum
in a dopamine-dependent manner (Glick et al., 1986). The PD which plays a key role in motor function. However, previous
animal model of this study, which was made by the 6-OHDA studies have shown that ventral striatum was also related with
injected into striatum, was characterized by slowly evolving motor functions such as amphetamine or apomophine rotation
and progressive nigrostriatal degeneration until 8 weeks after 6- and forepaw motor functions (Grealish et al., 2010; Heuer et al.,
OHDA administration and resembles the progressive nature of 2012; Nozaki et al., 2013). Taken together, our data suggested
the neurodegenerative process of human PD (Sauer and Oertel, that rTMS treatment might preserve or slow neuronal degen-
1994; Blandini et al., 2007). We observed that improvement in eration by 6-OHDA administration, and consequently improve
treadmill locomotion was observed at 2 weeks after rTMS motor performances in an animal model of PD.
treatment (4 weeks after 6-OHDA administration), but amphe- Finally, we evaluated endogenous neuroprotective and/or
tamine turning improvements was not shown at the time neurorestorative proteins from the 6-OHDA-lesioned brains
point. A possible explanation for the difference between the after rTMS treatment. We demonstrated that rTMS promoted
amphetamine-induced rotation and treadmill locomotion out- increases in expression levels of neurotrophic/growth factors
comes is that the discrepancy might be related to different such as BDNF, GDNF, NGF, PDGF, and VEGF in ipsilateral
mechanism of each test at the time when degeneration of DA hemispheres and SNc. The present study confirmed that
neurons was ongoing in the SNc. these factors increased in the rTMS group rather than in
The histological results of this study revealed that rTMS- the untreated PD group at 4 weeks after treatment. rTMS can
treated rats had more striatal THþ efferent fibers as well as change signaling pathways and gene transcription, immedi-
more THþ neurons in the SNc than those of the untreated ate early gene expression, and initiate the biosynthesis of
group, demonstrating that rTMS has a neuroprotective effects new molecules which persist in the tissue beyond the period
on survival of DA neurons and DA nerve terminals in the of stimulation, consequently increasing the synthesis of
ipsilateral SNc and striatum of 6-OHDA-induced PD rats. growth factors (Arias-Carrion et al., 2011).

Please cite this article as: Lee, J.Y., et al., Therapeutic effects of repetitive transcranial magnetic stimulation in an animal
model of Parkinson's disease. Brain Research (2013), http://dx.doi.org/10.1016/j.brainres.2013.08.051
8 brain research ] (]]]]) ]]]–]]]

Among these factors, BDNF is expressed by DA neurons in In addition, there are evidences that a variety of neuro-
both the SN and the ventral tegmental area (Baquet et al., chemical factors induced by physical exercise lead to motor
2005; Peterson and Nutt, 2008). In postmortem samples of PD functional improvement as well as neuroprotection in animal
patients, BDNF as well as GDNF was lower in the SNc than in models of PD. (Tuon et al., 2012; Lau et al., 2011). Previous
controls (Mogi et al., 1999; Howells et al., 2000; Peterson and reports also showed that neurotrophic factors such as BDNF
Nutt, 2008). BDNF protected DA neurons in vitro from the and NT-3 augment locomotor activity and rotational beha-
neurotoxic effects of 1-methyl-4-phenylpyridinium ion and vior, and striatal DA turnover, indicating that BDNF enhances
6-OHDA. Intrastriatal injection of BDNF in a rat model prior to the nigrostriatal DA system function (Shen et al., 1994;
lesioning reduced destruction of DA neurons in the SNc and Martin-Iverson et al., 1994; Altar et al., 1994). Therefore, we
decreased the apomorphine-induced rotation (Spina et al., considered that neurotrophic factors induced by rTMS might
1992; Shults et al., 1995; Mogi et al., 1999). rTMS is well known modulate nigrostriatal DA system, leading to functional
for changing the expression of BDNF. Previous studies have recovery.
reported that high-frequency rTMS increased the level of As a limitation of this study, we did not elucidate mechan-
BDNF expression in vivo and in patients with neuropsychia- isms of rTMS that involve modulation of the cortical network.
tric disorders (Kole et al., 1999; Müller et al., 2000; Angelucci Instead of functional alterations of the corticospinal pathway
et al., 2004; Yukimasa et al., 2006; Yue et al., 2009). However, involving short interval intracortical inhibition and paired-
there are few reports that rTMS modulates other neuro- association stimulation effects (Cantello et al., 2002), this
trophic factors except BDNF. study focused on the alterations of neurotrophic/growth
GDNF has been shown to promote the survival of DA factors after rTMS in a PD animal model. Because this study
neurons and has powerful protective effects for neurotoxin- did not evaluate neurophysiologic function, such as motor
induced degenerated DA neurons (Lin et al., 1993; Hoffer et al., evoked potentials, we could not interpret behavioral out-
1994; Gill et al., 2003; Slevin et al., 2005; Lang et al., 2006). In the comes with respect to modulation of the cortical network or
DA-depleted rat model, GDNF protects 6-OHDA, 1-methyl-4- activation of the nigrostriatal dopaminergic pathway.
phenyl-1,2,3,6-tetrahydropyridine and medial forebrain bundle The other limitation of this study is that repeated amphe-
axotomy-induced degenerated midbrain DA neurons (Kearns tamine administration has been reported to induce sensitiza-
and Gash, 1995; Tomac et al., 1995; Cohen et al., 2003; Bradley tion and priming effects in hemiparkinson rats (Erhardt et al.,
et al., 2010). In addition, NGF supports sympathetic and 2004). Therefore, it is possible that rTMS can affect the
sensory neurons, and functions in the development and priming process instead of behavioral improvement from
maintenance of cholinergic neurons in the basal forebrain preserved DA neuronal damage in the SNc and upregulation
(Sofroniew et al., 2001). NGF level decreased in experimental of neurotrophic/growth factors. However, amphetamine-
models and patients with PD. DA neurons and NGF are induced rotation test is a method that has been widely used
strongly believed to be functionally related. When NGF was to evaluate motor function related to endogenous DA depletion
administrated in a 6-OHDA-lesioned rat model of PD, a in hemiparkinson rat models. In this study, amphetamine-
significant increase of THþ neurons was also observed in the induced rotational test revealed that both groups continuously
SN, showing potential neuroprotective effects of NGF on increased in the number of rotations until 6 weeks after the 6-
nigrostriatal DA neurons (Lorigados Pedre et al., 2002; OHDA injection. A possible explanation for the increased
Chaturvedi et al., 2006). PDGF is a major mitogen for fibro- rotations is that this experimental model was characterized
blasts, smooth muscle cells and other cells (Heldin and by evolving progressive nigrostriatal degeneration, resembling
Westermark, 1999). PDGF also has the neuroprotective and the progressive nature of the neurodegenerative process of
neurotrophic functions such as neuronal survival and neurite human PD (Sauer and Oertel, 1994; Blandini et al., 2007).
formation not only in cultures of rat DA neurons (Nikkhah Another limitation of this study is that we did not provide
et al., 1993; Othberg et al., 1995; Pietz et al., 1996), but also in evidence for neurorestorative effects, including the repopula-
DA neurons in the SN of 6-OHDA-lesioned rats (Funa et al., tion of neurons, increased endogenous neural stem/progenitor
1996). It has been suggested that PDGF agonists may be cells, synaptogenesis and angiogenesis. On the other hand, this
potential therapeutic agents for patients with PD (Othberg study discerned the upregulation of neurotrophic/growth fac-
et al., 1995). VEGF is an established angiogenic factor with tors in rTMS-treated hemispheres and SNc. Recent literature
specificity for endothelial cells (Jin et al., 2000; Rosenstein and demonstrated that GDNF involved neurorestorative processes
Krum, 2004). Recently, VEGF has been reported to have (Airavaara et al., 2012). VEGF and PDGF were also shown to be
neuroprotective effects in cerebral ischemia and ischemic related with neurorestoration by angiogenesis (Zhang and
peripheral neuropathy, and neurogenesis effects in neurologi- Chopp, 2009). Therefore, we need to validate the neurorestora-
cal disorders (Schratzberger et al., 2000; Yasuhara et al., 2004a). tive processes described above in future studies.
The increased survival of the DA neurons by VEGF might be In conclusion, rTMS treatment improved motor functions
mediated by neuroprotective mechanisms. Appropriate neo- and preserved DA neuronal damage by 6-OHDA administra-
vascularization could improve microcirculation around DA tion in a rat model of PD. Additionally, rTMS promoted
fibers in the striatum, and protect DA neurons (Jin et al., expression of various growth factors such as BDNF, GDNF,
2000; Yasuhara et al., 2004a, 2004b, 2005). Taken together, NGF, PDGF-AA, and VEGF. The neuroprotective effects might
enhanced various neurotrophic/growth factors might protect be induced by upregulation of neurotrophic/growth factors.
neuronal populations against excitotoxic, oxidative, and meta- Collectively, our results could support a therapeutic potential
bolic insults as described in the previous reports (Mattson role of rTMS in the treatment of PD, extending its application
et al., 2002; Mattson and Lindvall, 1997). to other neurological diseases.

Please cite this article as: Lee, J.Y., et al., Therapeutic effects of repetitive transcranial magnetic stimulation in an animal
model of Parkinson's disease. Brain Research (2013), http://dx.doi.org/10.1016/j.brainres.2013.08.051
brain research ] (]]]]) ]]]–]]] 9

4.4. Amphetamine-induced rotation test


4. Experimental procedure
Rotational behaviors were evaluated in response to intraper-
4.1. Animals itoneal injection of D-amphetamine (5 mg/kg; Sigma-Aldrich,
St. Louis, MO, USA) on day 10 after the 6-OHDA injection.
Animal use and care protocols were approved by the Institu- Animals were injected with the amphetamine and placed into
tional Review Board, and the Institutional Animal Care and an automated multichannel rotometer system (ROTORAT,
Use Committee. Thirty adult male Sprague-Dawley rats (body MED Associates, Inc., St Albans, USA). Each animal was placed
weight, 250–300 g; Charles River Lab., Wilmington, MA, USA) into a cylindrical test chamber for 50 min. Animals with more
were used. All efforts were made to minimize the number of than 100 asymmetric turns per 50 min were used for experi-
animals required and to ensure minimal suffering. Rats were ments. Amphetamine-induced rotation test was performed at
housed with free access to food and water in a room 2 and 4 weeks after treatment.
maintained at constant room temperature (20–22 1C) with
alternate 12-h light/dark cycles according to animal protec- 4.5. Treadmill locomotion test
tion regulations.
The rats were placed on a treadmill which was covered with a
see-through box with lines drawn from 0 to 40 cm. The
treadmill belt moved at 72 rpm during the on cycles. After
4.2. 6-hydroxydopamine (6-OHDA) lesion the rats adapted to running for 1 min, the lengths from the
beginning of the box to the end of the rats' noses were
Animals were pretreated with norepinephrine transporter measured. One cycle was defined as 20 s of running and 20 s
blocker desipramine hydrochloride (12.5 mg/kg; Sigma- of rest. The treadmill locomotion test measurement was
Aldrich, St. Louis, MO, USA) 30 min before surgery. For the repeated five times, and the average result was used.
surgery, rats were deeply anesthetized with an intraperito-
neal injection of ketamine (70 mg/kg body weight) and xyla- 4.6. Immunohistochemistry
zine (8 mg/kg body weight). Each animal was positioned
within a stereotaxic apparatus (Stoelting Co., USA). The Six rats of each group were deeply anesthetized as described
20 μg/4 μl/site of 6-OHDA (Sigma-Aldrich, St. Louis, MO, USA) above, and the thorax was opened. The rats were perfused
was injected into two sites (total of 40 μg) in the right striatum transcardially with 100 ml of 0.9% sodium chloride, then with
according to the brain atlas (Paxinos and Watson, 2009). The 200 ml of 4% paraformaldehyde in 0.1 M phosphate buffer (PB,
6-OHDA was injected at the following coordinates relative to pH 7.4). The brain was carefully removed from the skull, fixed
bregma and dura: anterior–posterior (AP) þ0.5 mm, medial– for 12 h in the same fixative, and infiltrated with 30% sucrose
lateral (ML) 2.5 mm, dorsal–ventral (DV) 5.0 mm and AP solution at 4 1C until they sank. The brains were then placed
0.5 mm, ML 4.2 mm, and DV 5.0 mm at a rate of 1 μl/min on a brain blocker (David Kopf Instruments, St. Tujunga, USA)
using a 26 G Hamilton syringe (Fig. 1B). The inserted needle and sliced into a block containing the striatum or SNc, based
was withdrawn from each location after 5 min, and the skin on the atlas of Paxinos and Watson (2009). The block was
was sutured. Afterward, animals were kept on a heating pad then rapidly frozen in 2-methylbutane, chilled on dry ice, and
at 37 1C until they completely recovered. Ten days after the mounted in Tissue-Tek OCT compound (Sakura Finetechnical
injections, animals showing more than 100 rotations per Co., Tokyo, Japan). Serial coronal sections 40 μm in thickness
50 min were considered to be successfully altered and were were obtained using a cryostat microtome (Leica Microsys-
used for further experiments (n ¼24). Animals were sacrificed tems Inc., Wetzlar, Germany) and were distributed sequen-
at 6-week time intervals post-lesion. The contralateral side of tially into a set of 12 tissue wells containing 0.1 M PB. After a
the brain was used as the internal control. brief wash with 0.1 M PB, the brain sections were transferred
to another set of 12 tissue wells containing cryoprotection
solution and then stored in a freezer at a temperature below
20 1C until use. The sections were washed three times with
4.3. Repetitive transcranial magnetic stimulation 0.1 M PBS and treated with 3% hydrogen peroxide for 15 min
to suppress endogenous peroxidase activity. After washing
The rats with PD were randomly allocated into the following with 0.1 M PBS, the sections were treated for 1 h with 5%
two groups: rTMS group and untreated controls (n¼ 12 each). normal serum obtained from the same host species to reduce
The rats were immobilized with an acrylic holder, and the non-specific binding and were then incubated with rabbit
distance between the rat's head and the coil was maintained anti-TH monoclonal antibody (1:1000; Chemicon, Temecula,
at 1 cm. The coil was placed parallel to the skull of the rat. CA, USA), rabbit anti-BDNF (1:200; Abcam, Cambridge, UK),
The rTMS was administrated to awaken animals by using a rabbit anti-GDNF (1:200; Abcam, Cambridge, UK), rabbit anti-
round coil (5-cm diameter) and the Bicon-100 (M-cube Tech, NGF (1:200; Abcam, Cambridge, UK), rabbit anti-PDGF (1:200;
Korea) at a rate of 10 Hz for 1 s with 100% power that Abcam, Cambridge, UK) and rabbit anti-VEGF(1:200; Abcam,
generates a field of approximately 1 T and the duration of Cambridge, UK) diluted in 0.1 M PBST for 12 h. Sections were
stimulation was 20 min per day. The stimulation intensity washed in PBS and incubated for 1 h with biotinylated goat
was set to 100% machine output. The treatments were anti-rabbit IgG (1:200; Vector, Burlingame, CA, USA) for TH
performed daily for 4 weeks (Fig. 1A). and goat-anti rabbit Alexa-488 for TH and growth factors.

Please cite this article as: Lee, J.Y., et al., Therapeutic effects of repetitive transcranial magnetic stimulation in an animal
model of Parkinson's disease. Brain Research (2013), http://dx.doi.org/10.1016/j.brainres.2013.08.051
10 brain research ] (]]]]) ]]]–]]]

After rinsing, sections were exposed to avidin–biotin perox- Boston, MA, USA) antibody in TBST (10 mM Tris, pH 7.5, 150 mM
idase complex (Vector) for 1 h prior to incubation. For perox- NaCl, and 0.02% Tween 20) containing 5% nonfat dry milk. On
idase detection, chromogens including 3,3′-diaminobenzidine the next day, blots were washed three times with TBST and
tetrahydrochloride (DAB; Sigma-Aldrich, St. Louis, MO, USA) incubated for 1 h with horseradish peroxidase-conjugated sec-
and DAB-nickel (Vector) chromogen were used for 3–5 min. ondary antibodies (1:3000, Santa Cruz Biotech, Santa Cruz, CA,
USA) in TBST containing 3% nonfat dry milk at room tempera-
4.7. Image analysis ture. After washing three times with TBST, proteins were
visualized with an ECL detection system (Amersham Pharmacia
TH positive neurons on both sides of the SNc were counted Biotech, Piscataway, NJ, USA).
with the analySIS image analyzer system (Olympus Soft
Imaging System, Münster, Germany), and the percentage of 4.9. Multiplex ELISA
THþ neurons was calculated by dividing the average number
of THþ neurons in the ipsilateral SNc by the number of THþ To identify growth factors regulated by rTMS, a multiplex
neurons in the contralateral SNc. Light photomicrographic ELISA assay (Quantibodys array, RayBio-tech, Norcross, GA)
images were acquired on a Nikon Optiphot microscope was used to determine which of the following growth factors
(Nikon Inc., Tokyo, Japan) fitted with a Nikon digital camera were detectable in the ipsilateral hemispheres (n¼ 3 per
(DXM1200), using Nikon ACT-1 image capture software group): NGF, CNTF, PDGF-AA and VEGF. Expression of growth
(ver. 2.2). TH positive neurons on both sides of the striatum factors was detected using an array scanner (Gene PIX™
were measured using the MetaMorph Imaging System (Mole- 4000B, Axon instruments, USA).
cular Device, Sunnyvale, CA), and the percentage of THþ fibers
was calculated by dividing the average volume of THþ fibers in 4.10. Data analysis
the ipsilateral striatum by the volume of fibers in the contral-
ateral striatum. The area of the SNc was obtained using the Statistical analysis was performed using the t-test (Prism
MetaMorph Imaging System (Molecular Device, Sunnyvale, CA) Graph Pad Software, San Diego, CA, USA) and two-way
and converted to volume by multiplying the area by the section repeated ANOVA, followed by a Bonferroni post-hoc compar-
thickness (40 μm). BDNFþ, GDNFþ, NGFþ, PDGFþ and VEGFþ ison (SAS version 9.2, SAS Institute Inc., Cary, NC, USA). The
cells (%) in the SNc (/mm2) were quantified using the Meta- data were expressed as a mean7standard error of the mean
Morph Imaging System (Molecular Device, Sunnyvale, CA). The (SEM). The significance level was assumed at po0.05, unless
images were imported into Adobe Photoshop (ver. 7.0, Adobe otherwise indicated.
Systems Inc., San Jose, CA, USA) and were adjusted for bright-
ness and contrast to optimize photographic representation of
the images obtained by the microscope. Author contributions

4.8. Western blotting Lee JY: Conception and design, collection and/or assembly of
data, manuscript writing; Kim SH: Conception and design,
To compare the expression levels of neurotrophic factors, data analysis and interpretation, manuscript writing; Ko AR,
ipsilateral hemispheres (n¼ 5 per group) were lysed in 500 μl Lee JS,Yu JH, Seo JH: Collection and/or assembly of data; Cho
of cold RIPA buffer (50 mM Tris–HCl, pH 7.5, 1% Triton X-100, BP: Administrative support, conception and design, data
150 mM NaCl, 0.1% sodium dodecyl sulfate (SDS), and 1% analysis and interpretation; Cho SR: Data analysis and inter-
sodium deoxycholate) with a protease inhibitor cocktail pretation, manuscript writing, final approval of manuscript.
(Sigma-Aldrich, St. Louis, MO, USA). Tissue lysate was centri-
fuged at 13,000  g for 15 min at 4 1C. The supernatant was
harvested, and protein concentration was analyzed using a Acknowledgments
Qunt-iT protein assay kit (Molecular Probes, Eugene, Oregon,
USA). For electrophoresis, 50 μg of protein was dissolved in This study was supported by grants from the National
sample buffer (60 mM Tris–HCl, pH 6.8, 14.4 mM β-mercap- Research Foundation Research 2009-0077194; 2010-0020408;
toethanol, 25% glycerol, 2% SDS, and 0.1% bromophenol blue), 2010-0024334) funded by the Ministry of Education, Science
boiled for 10 min and separated on a 10% SDS reducing gel. and Technology, Republic of Korea, and the Korea Health
Separated proteins were transferred onto polyvinylidene Technology R&D project, Ministry of Health & Welfare
difluoride (PVDF) membranes (Invitrogen, Carlsbad, CA) using (A100054).
a trans-blot system. Blots were blocked for 1 h in Tris-buffered
saline (TBS) (10 mM Tris–HCl, pH 7.5, 150 mM NaCl) containing r e f e r e n c e s
5% nonfat dry milk at room temperature, washed three times
with TBS and incubated at 4 1C overnight with an anti-rabbit
polyclonal BDNF (1:1000, Abcam, Cambridge, MA, USA), anti- Airavaara, M., Voutilainen, M.H., Wang, Y., Hoffer, B., 2012.
Neurorestoration. Parkinsonism Relat. Disord. 18, S143–S146.
rabbit glial cell-derived neurotrophic factor (GDNF, 1:1000,
Altar, C.A., Boylan, C.B., Fritsche, M., Jones, B.E., Jackson, C.,
Abcam), anti-rabbit NGF (1:1000; Abcam, Cambridge, UK), anti-
Wiegand, S.J., Lindsay, R.M., Hyman, C., 1994. Efficacy of brain-
rabbit PDGF (1:1000; Abcam, Cambridge, UK), anti-rabbit VEGF derived neurotrophic factor and neurotrophin-3 on neuro-
(1:1000; Abcam, Cambridge, UK) and anti-rabbit TH (1:1000; chemical and behavioral deficits associated with partial
Abcam, Cambridge, UK) and anti-GAPDH (1:3000, Cell signaling, nigrostriatal dopamine lesions. J. Neurochem. 63, 1021–1032.

Please cite this article as: Lee, J.Y., et al., Therapeutic effects of repetitive transcranial magnetic stimulation in an animal
model of Parkinson's disease. Brain Research (2013), http://dx.doi.org/10.1016/j.brainres.2013.08.051
brain research ] (]]]]) ]]]–]]] 11

Angelucci, F., Oliviero, A., Pilato, F., Saturno, E., Dileone, M., Fitzgerald, P.B., Daskalakis, Z.J., 2011. The effects of repetitive
Versace, V., Musumeci, G., Batocchi, A.P., Tonali, P.A., Di transcranial magnetic stimulation in the treatment of
Lazzaro, V., 2004. Transcranial magnetic stimulation and depression. Expert Rev. Med. Devices 8, 85–95.
BDNF plasma levels in amyotrophic lateral sclerosis. Funa, K., Yamada, N., Brodin, G., Pietz, K., Ahgren, A., Wictorin, K.,
Neuroreport 15, 717–720. Lindvall, O., Odin, P., 1996. Enhanced synthesis of platelet-derived
Arias-Carrión, O., 2008. Basic mechanisms of rTMS: implications growth factor following injury induced by 6-hydroxydopamine in
in Parkinson's disease. Int. Arch. Med. 1, 2. rat brain. Neuroscience 74, 825–833.
Arias-Carrion, O., Machado, S., Paes, F., Velasques, B., Teixeira, S., Gedge, L., Beaudoin, A., Lazowski, L., du Toit, R., Jokic, R., Milev, R.,
Cardenas-Morales, L., Piedade, R., Ribeiro, P., Nardi, A.E., 2011. 2012. Effects of electroconvulsive therapy and repetitive
Is rTMS an effective therapeutic strategy that can be used to transcranial magnetic stimulation on serum brain-derived
treat Parkinson's disease?. CNS Neurol. Disord. Drug Targets neurotrophic factor levels in patients with depression. Front.
10, 693–702. Psychiatry 3, 1–8.
Baquet, Z.C., Bickford, P.C., Jones, K.R., 2005. Brain-derived Grealish, S., Mattsson, B., Draxler, P., Bjorklund, A., 2010.
neurotrophic factor is required for the establishment of the Characterisation of behavioural and neurodegenerative
proper number of dopaminergic neurons in the substantia changes induced by intranigral 6- hydroxydopamine lesions
nigra pars compacta. J. Neurosci. 25, 6251–6259. in a mouse model of Parkinson's disease. Eur. J. Neurosci. 31,
Bentwich, J., Dobronevsky, E., Aichenbaum, S., Shorer, R., Peretz, 2266–2278.
R., Khaigrekht, M., Marton, R.G., Rabey, J.M., 2011. Beneficial Glick, S.D., Shapiro, R.M., Drew, K.L., Hinds, P.A., Carlson, J.N.,
effect of repetitive transcranial magnetic stimulation 1986. Differences in spontaneous and amphetamine-induced
combined with cognitive training for the treatment of rotational behavior and in sensitization to amphetamine,
Alzheimer's disease: a proof of concept study. J. Neural among Sprague-Dawley derived rats from different sources.
Transm. 118, 463–471. Physiol. Behav. 38, 67–70.
Blandini, F., Levandis, G., Bazzini, E., Nappi, G., Armentero, M.T., Gill, S.S., Patel, N.K., Hotton, G.R., O'Sullivan, K., McCarter, R.,
2007. Time-course of nigrostriatal damage, basal ganglia Bunnage, M., Brooks, D.J., Svendsen, C.N., Heywood, P., 2003.
metabolic changes and behavioural alterations following Direct brain infusion of glial cell line-derived neurotrophic
intrastriatal injection of 6-hydroxydopamine in the rat: new factor in Parkinson disease. Nat. Med. 9, 589–595.
clues from an old model. Eur. J. Neurosci. 25, 397–405. Hausmann, A., Weis, C., Marksteiner, J., Hinterhuber, H., Humpel,
Bradley, L.H., Fuqua, J., Richardson, A., Turchan-Cholewo, J., Ai, C., 2000. Chronic repetitive transcranial magnetic stimulation
Y., Kelps, K.A., Glass, J.D., He, X., Zhang, Z., Grondin, R., Littrell, enhances c-fos in the parietal cortex and hippocampus. Brain
O.M., Huettl, P., Pomerleau, F., Gash, D.M., Gerhardt, G.A., 2010. Res. Mol. Brain Res. 76, 355–362.
Dopamine neuron stimulating actions of a GDNF propeptide. Heldin, C.H., Westermark, B., 1999. Mechanism of action and
PLoS One 5, e9752. in vivo role of platelet-derived growth factor. Physiol. Rev. 79,
Cantello, R., Tarletti, R., Civardi, C., 2002. Transcranial magnetic 1283–1316.
stimulation and Parkinson's disease. Brain Res. Brain Res. Rev. Heuer, A., Smith, G.A., Lelos, M.J., Lane, E.L., Dunnett, S.B., 2012.
38, 309–327. Unilateral nigrostriatal 6-hydroxydopamine lesions in mice
Chang, J.Y., Shi, L.H., Luo, F., Woodward, D.J., 2003. High I: motor impairments identify extent of dopamine depletion at
frequency stimulation of the subthalamic nucleus improves three different lesion sites. Behav. Brain Res. 228, 30–43.
treadmill locomotion in unilateral 6-hydroxydopamine Hoffer, B.J., Hoffman, A., Bowenkamp, K., Huettl, P., Hudson, J.,
lesioned rats. Brain Res. 983, 174–184. Martin, D., Lin, L.F., Gerhardt, G.A., 1994. Glial cell line-derived
Chaturvedi, R.K., Shukla, S., Seth, K., Agrawal, A.K., 2006. Nerve neurotrophic factor reverses toxin-induced injury to midbrain
growth factor increases survival of dopaminergic graft, dopaminergic neurons in vivo. Neurosci. Lett. 182, 107–111.
rescue nigral dopaminergic neurons and restores functional Howells, D.W., Porritt, M.J., Wong, J.Y., Batchelor, P.E., Kalnins, R.,
deficits in rat model of Parkinson's disease. Neurosci. Lett. 398, Hughes, A.J., Donnan, G.A., 2000. Reduced BDNF mRNA
44–49. expression in the Parkinson's disease substantia nigra. Exp.
Cho, S.S., Strafella, A.P., 2009. rTMS of the left dorsolateral Neurol. 166, 127–135.
prefrontal cortex modulates dopamine release in the Ji, R.R., Schlaepfer, T.E., Aizenman, C.D., Epstein, C.M., Qiu, D.,
ipsilateral anterior cingulate cortex and orbitofrontal cortex. Huang, J.C., Rupp, F., 1998. Repetitive transcranial magnetic
PLoS One 4, e6725. stimulation activates specific regions in rat brain. Proc. Natl.
Cohen, A.D., Tillerson, J.L., Smith, A.D., Schallert, T., Zigmond, Acad. Sci. USA 95, 15635–15640.
M.J., 2003. Neuroprotective effects of prior limb use in 6- Jin, K.L., Mao, X.O., Greenberg, D.A., 2000. Vascular endothelial
hydroxydopamine-treated rats: possible role of GDNF. J. growth factor: direct neuroprotective effect in in vitro
Neurochem. 85, 299–305. ischemia. Proc. Natl. Acad. Sci. USA 97, 10242–10247.
Corti, M., Patten, C., Triggs, W., 2012. Repetitive transcranial Johnson, R.E., Schallert, T., Becker, J.B., 1999. Akinesia and
magnetic stimulation of motor cortex after stroke: a focused postural abnormality after unilateral dopamine depletion.
review. Am. J. Phys. Med. Rehabil. 91, 254–270. Behav. Brain Res. 104, 189–196.
Doi, W., Sato, D., Fukuzako, H., Takigawa, M., 2001. c-Fos Kearns, C.M., Gash, D.M., 1995. Protects nigral dopamine neurons
expression in rat brain after repetitive transcranial magnetic against 6-hydroxydopamine in vivo. Brain Res. 672, 104–111.
stimulation. Neuroreport 12, 1307–1310. Keck, M.E., Sillaber, I., Ebner, K., Welt, T., Toschi, N., Kaehler, S.T.,
Erhardt, A., Sillaber, I., Welt, T., Müller, M.B., Singewald, N., Keck, Singewald, N., Philippu, A., Elbel, G.K., Wotjak, C.T., Holsboer,
M.E., 2004. Repetitive transcranial magnetic stimulation F., Landgraf, R., Engelmann, M., 2000. Acute transcranial
increases the release of dopamine in the nucleus accumbens magnetic stimulation of frontal brain regions selectively
shell of morphine-sensitized rats during abstinence. modulates the release of vasopressin, biogenic amines and
Neuropsychopharmacology 29, 2074–2080. amino acids in the rat brain. Eur. J. Neurosci. 12, 3713–3720.
Fisher, B.E., Wu, A.D., Salem, G.J., Song, J., Lin, C.H., Yip, J., Cen, S., Khedr, E.M., Rothwell, J.C., Shawky, O.A., Ahmed, M.A., Foly, N.,
Gordon, J., Jakowec, M., Petzinger, G., 2008. The effect of Hamdy, A., 2007. Dopamine levels after repetitive transcranial
exercise training in improving motor performance and magnetic stimulation of motor cortex in patients with
corticomotor excitability in people with early Parkinson's Parkinson's disease: preliminary results. Mov. Disord. 22,
disease. Arch. Phys. Med. Rehabil. 89, 1221–1229. 1046–1050.

Please cite this article as: Lee, J.Y., et al., Therapeutic effects of repetitive transcranial magnetic stimulation in an animal
model of Parkinson's disease. Brain Research (2013), http://dx.doi.org/10.1016/j.brainres.2013.08.051
12 brain research ] (]]]]) ]]]–]]]

Kole, M.H., Fuchs, E., Ziemann, U., Paulus, W., Ebert, U., 1999. Pellis, S.M., Pellis, V.C., Chesire, R.M., Rowland, N., Teitelbaum, P.,
Changes in 5-HT1A and NMDA binding sites by a single rapid 1987. Abnormal gait sequence in locomotion after atropine
transcranial magnetic stimulation procedure in rats. Brain treatment of catecholamine-deficient akinetic rats. Proc. Natl.
Res. 826, 309–312. Acad. Sci. USA 84, 8750–8753.
Lang, A.E., Gill, S., Patel, N.K., Lozano, A., Nutt, J.G., Penn, R., Peterson, A.L., Nutt, J.G., 2008. Treatment of Parkinson's disease
Brooks, D.J., Hotton, G., Moro, E., Heywood, P., Brodsky, M.A, with trophic factors. Neurotherapeutics 5, 270–280.
Burchiel, K., Kelly, P., Dalvi, A., Scott, B., Stacy, M., Turner, D., Pietz, K., Odin, P., Funa, K., Lindvall, O., 1996. Protective effect of
Wooten, V.G., Elias, W.J., Laws, E.R., Dhawan, V., Stoessl, A.J., platelet-derived growth factor against 6-hydroxydopamine-
Matcham, J., Coffey, R.J., Traub, M., 2006. Randomized induced lesion of rat dopaminergic neurons in culture.
controlled trial of intraputamenal glial cell line-derived Neurosci. Lett. 204, 101–104.
neurotrophic factor infusion in Parkinson disease. Ann. Rosenstein, J.M., Krum, J.M., 2004. New roles for VEGF in nervous
Neurol. 59, 459–466. tissue—beyond blood vessels. Exp. Neurol. 187, 246–253.
Lau, Y.S., Patki, G., Das-Panja, K., Le, W.D., Ahmad, S.O., 2011. Sauer, H., Oertel, W.H., 1994. Progressive degeneration of
Neuroprotective effects and mechanisms of exercise in a nigrostriatal dopamine neurons following intrastriatal
chronic mouse model of Parkinson's disease with moderate terminal lesions with 6-hydroxydopamine: a combined
neurodegeneration. Eur. J. Neurosci. 33, 1264–1274. retrograde tracing and immunocytochemical study in the rat.
Lee, H.Y., Hsieh, T.H., Liang, J.I., Yeh, M.L., Chen, J.J., 2012. Neuroscience 59, 401–415.
Quantitative video-based gait pattern analysis for Schratzberger, P., Schratzberger, G., Silver, M., Curry, C., Kearney,
hemiparkinsonian rats. Med. Biol. Eng. Comput. 50, 937–946. M., Magner, M., Alroy, J., Adelman, L.S., Weinberg, D.H.,
Lin, L.-F., Doherty, D.H., Lile, J.D., Bektesh, S., Collins, F., 1993. Ropper, A.H., Isner, J.M., 2000. Favorable effect of VEGF gene
GDNF: a glial cell line-derived neurotrophic factor for transfer on ischemic peripheral neuropathy. Nat. Med. 6,
midbrain dopaminergic neurons. Science 260, 1130–1132. 405–413.
Lorigados Pedre, L., Pavón Fuentes, N., Alvarez González, L., Shen, R.Y., Altar, C.A., Chiodo, L.A., 1994. Brain-derived
McRae, A., Serrano Sánchez, T., Blanco Lescano, L., Macías neurotrophic factor increases the electrical activity of pars
González, R., 2002. Nerve growth factor levels in Parkinson compacta dopamine neurons in vivo. Proc. Natl. Acad. Sci.
disease and experimental Parkinsonian rats. Brain Res. 952, USA 91, 8920–8924.
122–127. Shults, C.W., Kimber, T., Altar, C.A., 1995. BDNF attenuates the
Mattson, M.P., Lindvall, O., 1997. Neurotrophic factor and cytokine effects of intrastriatal injection of 6-hydroxydopamine.
signalingin the aging brain. In: Mattson, M.P., Geddes, J.W. Neuroreport 6, 1109–1112.
(Eds.), The Aging Brain, Advances in Cell Aging and Siebner, H.R., Mentschel, C., Auer, C., Conrad, B., 1999. Repetitive
Gerontology, vol. 2. JAI Press, Greenwich CT, pp. 299–345. transcranial magnetic stimulation has a beneficial effect
Mattson, M.P., Duan, W., Chan, S.L., Cheng, A., Haughey, N., Gary, on bradykinesia in Parkinson's disease. Neuroreport 10,
D.S., Guo, Z., Lee, J., Furukawa, K., 2002. Neuroprotective and 589–594.
neurorestorative signal transduction mechanisms in brain Slevin, J.T., Gerhardt, G.A., Smith, C.D., Gash, D.M., Kryscio, R.,
aging: modification by genes, diet and behavior. Neurobiol. Young, B., 2005. Improvement of bilateral motor functions in
Aging 23, 695–705. patients with Parkinson disease through the unilateral
Martin-Iverson, M.T., Todd, K.G., Altar, C.A., 1994. Brain-derived intraputaminal infusion of glial cell line-derived neurotrophic
neurotrophic factor and neurotrophin-3 activate striatal factor. J. Neurosurg. 102, 216–222.
dopamine and serotonin metabolism and related behaviors: Sofroniew, M.V., Howe, C.L., Mobley, W.C., 2001. Nerve growth
interactions with amphetamine. J. Neurosci. 14, 1262–1270. factor signaling, neuroprotection, and neural repair. Annu.
Mogi, M., Togari, A., Kondo, T., Mizuno, Y., Komure, O., Kuno, S., Rev. Neurosci. 24, 1217–1281.
Ichinose, H., Nagatsu, T., 1999. Brain-derived growth factor Spina, M.B., Hyman, C., Squinto, S., Lindsay, R.M., 1992. Brain-
and nerve growth factor concentrations are decreased in the derived neurotrophic factor protects dopaminergic cells from
substantianigra in Parkinson's disease. Neurosci. Lett. 270, 6-hydroxydopamine toxicity. Ann. N. Y. Acad. Sci. 648, 348–350.
45–48. Strafella, A.P., Paus, T., Barrett, J., Dagher, A., 2001. Repetitive
Müller, M.B., Toschi, N., Kresse, A.E., Post, A., Keck, M.E., 2000. transcranial magnetic stimulation of the human prefrontal
Long-term repetitive transcranial magnetic stimulation cortex induces dopamine release in the caudate nucleus.
increases the expression of brain-derived neurotrophic factor J. Neurosci. 21, 1–4.
and cholecystokinin mRNA, but not neuropeptide tyrosine Strafella, A.P., Paus, T., Fraraccio, M., Dagher, A., 2003. Striatal
mRNA in specific areas of rat brain. dopamine release induced by repetitive transcranial magnetic
Neuropsychopharmacology 23, 205–215. stimulation of the human motor cortex. Brain 126, 2609–2615.
Nikkhah, G., Odin, P., Smits, A., Tingström, A., Othberg, A., Theilig, S., Podubecka, J., Bösl, K., Wiederer, R., Nowak, D.A., 2011.
Brundin, P., Funa, K., Lindvall, O., 1993. Platelet-derived Functional neuromuscular stimulation to improve severe
growth factor promotes survival of rat and human hand dysfunction after stroke: does inhibitory rTMS enhance
mesencephalic dopaminergic neurons in culture. Exp. Brain therapeutic efficiency?. Exp. Neurol. 230, 149–155.
Res. 92, 516–523. Tomac, A., Widenfalk, J., Lin, L.F., Kohno, T., Ebendal, T., Hoffer,
Nozaki, T., Sugiyama, K., Yagi, S., Yoshikawa, E., Kanno, T., B.J., Olson, L., 1995. Retrograde axonal transport of glial cell
Asakawa, T., Ito, T., Terada, T., Namba, H., Ouchi, Y., 2013. line-derived neurotrophic factor in the adult nigrostriatal
Effect of subthalamic nucleus stimulation during exercise on system suggests a trophic role in the adult. Proc. Natl. Acad.
the mesolimbocortical dopaminergic region in Parkinson's Sci. USA 92, 8274–8278.
disease: a positron emission tomography study. J. Cereb. Blood Tuon, T., Valvassori, S.S., Lopes-Borges, J., Luciano, T., Trom, C.B.,
Flow Metab. 33, 415–421. Silva, L.A., Quevedo, J., Souza, C.T., Lira, F.S., Pinho, R.A., 2012.
Othberg, A., Odin, P., Ballagi, A., Ahgren, A., Funa, K., Lindvall, O., Physical training exerts neuroprotective effects in the
1995. Specific effects of platelet derived growth factor (PDGF) regulation of neurochemical factors in an animal model of
on fetal rat and human dopaminergic neurons in vitro. Exp. Parkinson's disease. Neuroscience 227, 305–312.
Brain Res. 105, 111–122. Wang, H.Y., Crupi, D., Liu, J., Stucky, A., Cruciata, G., Di Rocco, A.,
Paxinos, G., Watson, C., 2009. The Rat Brain in Stereotaxic Friedman, E., Quartarone, A., Ghilardi, M.F., 2011. Repetitive
Coordinates. Academic Press, London. transcranial magnetic stimulation enhances BDNF-TrkB

Please cite this article as: Lee, J.Y., et al., Therapeutic effects of repetitive transcranial magnetic stimulation in an animal
model of Parkinson's disease. Brain Research (2013), http://dx.doi.org/10.1016/j.brainres.2013.08.051
brain research ] (]]]]) ]]]–]]] 13

signaling in both brain and lymphocyte. J. Neurosci. 31, 2005. Neurorescue effects of VEGF on a rat model of
11044–11054. Parkinson's disease. Brain Res. 1053, 10–18.
Wu, A.D., Fregni, F., Simon, D.K., Deblieck, C., Pascual-Leone, A., Yue, L., Xiao-Lin, H., Tao, S., 2009. The effects of chronic repetitive
2008. Noninvasive brain stimulation for Parkinson's disease transcranial magnetic stimulation on glutamate and gamma-
and dystonia. Neurotherapeutics 5, 345–361. aminobutyricacid in rat brain. Brain Res. 1260, 94–96.
Yang, X., Song, L., Liu, Z., 2010. The effect of repetitive Yukimasa, T., Yoshimura, R., Tamagawa, A., Uozumi, T., Shinkai,
transcranial magnetic stimulation on a model rat of K., Ueda, N., Tsuji, S., Nakamura, J., 2006. High-frequency
Parkinson's disease. Neuroreport 21, 268–272. repetitive transcranial magnetic stimulation improves
Yang, Y.R., Tseng, C.Y., Chiou, S.Y., Liao, K.K., Cheng, S.J., Lai, K.L., refractory depression by influencing catecholamine and brain-
Wang, R.Y., 2013. Combination of rTMS and treadmill training derived neurotrophic factors. Pharmacopsychiatry 39, 52–59.
modulates corticomotor inhibition and improves walking in Zamir, O., Gunraj, C., Ni, Z., Mazzella, F., Chen, R., 2012. Effects of
Parkinson disease: a randomized trial. Neurorehabil. Neural theta burst stimulation on motor cortex excitability in
Repair 27, 79–86. Parkinson's disease. Clin. Neurophysiol. 123, 815–821.
Yasuhara, T., Shingo, T., Date, I., 2004a. The potential role of Zhang, Z.G., Chopp, M., 2009. Neurorestorative therapies for
vascular endothelial growth factor in the central nervous stroke: underlying mechanisms and translation to the clinic.
system. Rev. Neurosci. 15, 293–307. Lancet Neurol. 8, 491–500.
Yasuhara, T., Shingo, T., Kobayashi, K., Takeuchi, A., Yano, A., Zwanzger, P., Ella, R., Keck, M.E., Rupprecht, R., Padberg, F., 2002.
Muraoka, K., Matsui, T., Miyoshi, Y., Hamada, H., Date, I., Occurrence of delusions during repetitive transcranial
2004b. Neuroprotective effects of vascular endothelial growth magnetic stimulation (rTMS) in major depression. Biol.
factor (VEGF) upon dopaminergic neurons in a rat model of Psychiatry 51, 602–603.
Parkinson's disease. Eur. J. Neurosci. 19, 1494–1504.
Yasuhara, T., Shingo, T., Muraoka, K., Kameda, M., Agari, T.,
WenJi, Y., Hayase, H., Hamada, H., Borlongan, C.V., Date, I.,

Please cite this article as: Lee, J.Y., et al., Therapeutic effects of repetitive transcranial magnetic stimulation in an animal
model of Parkinson's disease. Brain Research (2013), http://dx.doi.org/10.1016/j.brainres.2013.08.051

You might also like