You are on page 1of 11

J. Pineal Res. 2014; 56:213–223 © 2013 John Wiley & Sons A/S.

Published by John Wiley & Sons Ltd


Doi:10.1111/jpi.12114 Journal of Pineal Research

Melatonin improves neuroplasticity by upregulating the growth-


Molecular, Biological, Physiological and Clinical Aspects of Melatonin

associated protein-43 (GAP-43) and NMDAR postsynaptic density-


95 (PSD-95) proteins in cultured neurons exposed to glutamate
excitotoxicity and in rats subjected to transient focal cerebral
ischemia even during a long-term recovery period
Abstract: Recent evidence shows that the NMDAR postsynaptic density-95 Wei-Sheng Juan1,*, Sheng-Yang
(PSD-95), growth-associated protein-43 (GAP-43), and matrix Huang1,*, Che-Chao Chang1,
metalloproteinase-9 (MMP-9) protein enhance neuroplasticity at the subacute Yu-Chang Hung1, Yu-Wen Lin1,
stage of stroke. Here, we evaluated whether melatonin would modulate the Tsung-Ying Chen1,2, Ai-Hua Lee1,
Ai-Chiang Lee1, Tian-Shung Wu1,3
PSD-95, GAP-43, and MMP-9 proteins in cultured neurons exposed to
and E-Jian Lee1
glutamate excitotoxicity and in rats subjected to experimental stroke. Adult
1
male Sprague–Dawley rats were treated with melatonin (5 mg/kg) or vehicle at Neurophysiology Laboratory, Institute of
Biomedical Engineering & Department of
reperfusion onset after transient occlusion of the right middle cerebral artery Surgery, National Cheng Kung University
(tMCAO) for 90 min. Animals were euthanized for Western immunoblot Hospital, College of Medicine, National Cheng
analyses for the PSD-95 and GAP-43 proteins and gelatin zymography for the Kung University, Tainan, Taiwan; 2Department
of Anesthesiology, Buddhist Tzu-Chi University
MMP-9 activity at 7 days postinsult. Another set of animals was sacrificed for and Buddhist Tzu Chi General Hospital,
histologic and Golgi–Cox-impregnated sections at 28 days postinsult. In Hualien, Taiwan; 3Department of Chemistry,
cultured neurons exposed to glutamate excitotoxicity, melatonin significantly National Cheng Kung University, Tainan,
Taiwan
upregulated the GAP-43 and PSD-95 expressions and improved dendritic
aborizations (P < 0.05, respectively). Relative to controls, melatonin-treated
stroke animals caused a significant improvement in GAP-43 and PSD-95 Key words: glutamate excitotoxicity, melatonin,
neuroplasticity, neuroprotection, stroke
expressions as well as the MMP-9 activity in the ischemic brain (P < 0.05).
Address reprint requests to E-Jian Lee,
Consequently, melatonin also significantly promoted the dendritic spine
Department of Surgery, National Cheng Kung
density and reduced infarction in the ischemic brain, and improved University Medical center and Medical School,
neurobehaviors as well at 28 days postinsult (P < 0.05, respectively). Together, 138 Sheng-Li Road, Tainan 70428, Taiwan.
E-mail: ejian@mail.ncku.edu.tw
melatonin upregulates GAP-43, PSD-95, and MMP-9 proteins, which likely
*These two authors have made equal
accounts for its actions to improve neuroplasticity in cultured neurons exposed contributions to the study.
to glutamate excitotoxicity and to enhance long-term neuroprotection, Received November 4, 2013;
neuroplasticity, and brain remodeling in stroke rats. Accepted December 13, 2013.

Recent studies have highlighted the involvement of


Introduction matrix metalloproteinase-9 (MMP-9) protein and NMDA-
Current treatment modality for ischemic stroke is still con- type glutamate receptors (NMDAR) in synaptic plasticity
fined to thrombolysis administration that benefits only a and brain remodeling following stroke [9–13]. At an early
small proportion of stroke patients at the acute stage [1]. stage of stroke, activated and overexpressed matrix metal-
There is an urgent need to rectify the current thrombolytic loproteinases (MMPs) induce endothelial and extracellular
strategy so as to improve clinical outcome of stroke matrix (ECM) damage, leading to transmigration of
patients at the subacute and chronic stages of recovery inflammatory cells and large, toxic molecules into the
[2–8]. Thus, functional deficits caused by ischemic stroke brain parenchyma [14–16]. Conversely, MMPs enzymes
may be restored either by enhancing the rewiring process also played essential roles in the neuroplasticity and brain
of the damaged brain tissues or by decreasing the remote remodeling at the subacute stage of stroke. In particular,
injury that occurs distal to the ischemic territory. More- the MMP-mediated proteolysis is essential for regulating
over, functional deficits may be replaced or compensated synaptic plasticity and neurogenesis at the subacute and
for by de novo neurogenesis or using other intact brain late healing stages following stroke [11, 17–19].
tissues. Strategies that have the potential to modulate neu- The NMDA-type glutamate receptors (NMDAR) medi-
roplasticity and/or neurogenesis, in addition to the neuro- ate both trophic and excitotoxic signaling in CNS neurons
protective ability to reduce acute brain damage, have [12, 13, 20–22]. Inhibitors of the NMDAR postsynaptic
therefore been suggested by us and others to improve density-95 (PSD-95) within 3 hr postinsult reduced brain
current stroke treatment [4–6, 8]. damage following permanent or transient focal cerebral

213
Juan et al.

ischemia [22]. A major pathway of NMDAR signaling to


Neuronal cultures, glutamate-induced excitotoxicity,
cell death in cortical and hippocampal neurons requires
and assay for dendritic aborizations
the scaffolding protein PSD-95 and activation of neuronal
nitric oxide synthase (nNOS) [23]. Neuronal protection Based on a method described previously [36, 42], cultured
mediated by PSD-95, thus, correlates well with reduced neurons were obtained from cerebral cortices of 1-day-old
activation of nNOS. Accordingly, MMPs and PSD-95 are Sprague–Dawley rats. Experiments were undertaken on
thought to be essential proteins for the ECM remodeling cultured neurons between 7 and 10 days in vitro (DIV).
and synaptic plasticity cascades, although each protein vir- Neurons were incubated melatonin (20–100 lM) or vehicle
tually has shown a Janus face of either toxic or neuronal (0.1% DMSO). Cultured neurons (n = 10–14) were pre-
plasticity/trophic effects depending on different time treated with melatonin (20–100 lM) or vehicle (0. 1%
course and/or molecular basis of injury. DMSO) for 30 min, and then, they were exposed to gluta-
The 43-kDa growth-associated protein (GAP-43) is a mate (300 lM) for 48 hr. Thereafter, neurons were
nervous tissue-specific protein that is synthesized at high co-incubated with mouse primary monoclonal antibodies
levels during axonal growth in neuronal development and antimicrotubule-associated protein-2 (MAP-2; dilution
axonal regrowth in regeneration in the peripheral and 1:1000, Santa Cruz Biotechnology, Santa Cruz, CA, USA)
central nervous systems [14–26]. GAP-43, therefore, repre- antibody. Anti-rabbit IgG secondary antibody conjugated
sents an important marker for axonal regeneration and with biotin (1:100, Jackson ImmunoResearch, Inc., West
sprouting. Grove, PA, USA) was then added, followed by fluorescein
We and several other investigators have previously dem- (DTAF)-conjugated streptavidin (1:150, Jackson Immuno-
onstrated that melatonin is highly effective in reducing Research). Neurons were co-incubated with 4’,-6-
acute ischemic brain damage [8, 14, 15, 27–29]. We used diamidino-2-phenylindole (DAPI; 2 lM, Sigma-Aldrich).
melatonin, primarily because the agent and its metabolites Fluorescence was assessed at excitation 365 nm and emis-
are potent, blood-brain barrier-permeable antioxidants, sion >420 nm for DAPI and was at excitation 450–490 nm
and radical scavengers [30–34]. Thus, melatonin protects and emission >515 nm for DTAF detection. Dendritic
the brain against acute ischemic damage to both gray and aborizations (length and branching) of the immunostained
white matter, ECM, blood-brain barrier permeability, and neurons were determined using a microscope (Olympus
functional neurovascular unit [14, 15, 27, 35–38]. More IX71, Olympus Optical Co. Ltd, Tokyo, Japan) equipped
recently, we have shown that melatonin improved the den- with epifluorescence and a computerized image analyzer
dritic spine density and the somatosensory field potentials (Image Pro plus 5.1, Media Cybernetics, Silver Spring,
of the ischemic brain at the subacute stage of insult, and MD, USA).
this was associated with an elevated level of SNAP-25, but
not synaptophysin (marker of synaptogenesis), protein
Animal preparation, anesthesia, and monitoring
expression [8]. Whereas melatonin has been shown to con-
fer some presynaptic and dendritic neuroplasticity at a Male Sprague–Dawley rats, weighing 240–290 g, were sup-
subacute stage of an ischemic insult, there is still a need to plied by the University Laboratory Animal Center and
further clarify whether melatonin offers neuroplasticity at were allowed free access to food and water. Animals were
the postsynaptic, axonal sprouting and ECM signaling anesthetized with 1–2% halothane in 70% N2O/30% O2.
cascades. In particular, we presumed that melatonin and During surgery, body temperature was maintained at
its active metabolites function to inhibit nNOS activity 37.0  0.5°C, which corresponded to brain temperature of
[38–41] and therefore might be suited to enhance the PSD- 37.2  0.5°C [43], using a thermostatically controlled
95-mediated synaptic plasticity. It was also important to heating blanket and rectal probe (Harvard Apparatus,
determine whether melatonin-induced neuroplasticity South Natick, MA, USA). The right femoral artery was
remains effectively during a long-lasting recovery period. cannulated for measuring arterial blood gases, glucose,
In the current study, we therefore examined whether hematocrit, and blood pressure.
melatonin reduces brain infarct volumes and dendritic
spine damage as well as improves the postsynaptic plastic-
Experimental model and grouping
ity, axonal sprouting, and ECM remodeling during a long-
term recovery after transient focal cerebral ischemia in Focal cerebral ischemia was employed by intra-arterial
rats. In addition, we explored possible contributions of the suture occlusion of the proximal right middle cerebral
melatonin-mediated neuroplasticity in cultured neurons artery (MCA) for 90 min, as described previously [8, 14,
exposed to glutamate excitotoxicity. 27, 36, 43–45]. Laser Doppler flowmetry (LDF, Laserflo
BMP2, Vasamedics, St. Paul, MN, USA) was used for
local cortical blood perfusion (LCBF) measurements, and
Materials and methods
the data of LCBF were expressed as a percentage of the
All procedures performed were approved by the Subcom- baseline values.
mittee on Research Animal Care of the University Medical Melatonin (Sigma-Aldrich) was dissolved in a mixture
Center. All chemicals were purchased from Sigma-Aldrich of PEG 400 (Sigma-Aldrich) and 0.9% normal saline (3:7,
Co. (St Louis, MO, USA) unless otherwise indicated. Mel- vol./vol.). A dose of melatonin of 5 mg/kg was chosen
atonin was dissolved in polyethylene glycol 400 (PEG 400, based on the pharmacokinetic study of exogenous melato-
Sigma-Aldrich) or dimethylsulfoxide (DMSO, Sigma- nin in animal models [46] and the neuroprotective
Aldrich). dose–response studies of melatonin in experimental stoke

214
Melatonin enhanced postinsult neuroplasticity

[36–39]. In a randomized fashion, animals were given were sacrificed. Based on the changes measured in LCBF,
intravenously either melatonin at 5 mg/kg (n = 39) or we defined an imaginary line, which was parallel to the
vehicle (PEG-saline, n = 40) at the initiation time of reper- superior sagittal sinus (SSS) and 3.0 mm lateral to the
fusion (i.e., 90 min after the onset of ischemia). In the first bregma, to divide the superior limb of the penumbral (left
series of experiments, animals treated either with melato- side to the line) and the ischemic core (right side to the
nin (5 mg/kg, n = 10) or vehicle (PEG-saline, n = 10) were line) cortices in the ischemic brain [8]. Cortical dissection
assigned for evaluating brain infarction and neuronal was continued between the rhinal fissure and the lateral
damage and were euthanized after 28 days following the olfactory tract. The inferior margin of ischemic core was
ischemic insult (Day 28). In the second set of experiments, defined as the cortical brain tissues located superiorly to
animals treated either with melatonin (5 mg/kg, n = 17) or the level of the rhinal fissure.
vehicle (n = 18) were assigned to assess the dendritic spine Samples were homogenized in lysis buffer, containing
density by the Golgi–Cox staining at Day 1 and Day 28 1% Triton X-100, 20 mM Tris-HCl (pH 7.5), 150 mM
(n = 8 for both groups, respectively). The third series of NaCl, 0.5% deoxycholate, 1 mM EDTA, 0.1% SDS, and
animals, received melatonin (5 mg/kg, n = 12) or vehicle were centrifuged at 18,000 g for 60 min at 4°C. Protein
(n = 12) upon reperfusion, were assigned to evaluate the concentrations of each sample were determined with a
synaptic integrity by Western blotting and the ECM BCA protein assay kit (Pierce, Rockford, IL, USA).
remodeling by zymography at Day 7. Additional 16 Cell lysates of 25 lg protein were separated by 10%
animals which received a sham-operated procedure served sodium dodecyl sulfate–polyacrylamide gels (SDS-PAGE)
as the nonischemic controls. and were transferred onto polyvinylidene difluoride
(PVDF) microporous membranes (IPVH00010, Millipore,
Billerica, MA, USA) [8, 45]. Membranes were blocked
Animal sacrifice and histologic outcome measure
with 5% milk, then probed with primary antibodies
Sacrifice was performed under anesthesia by decapitation against GAP-43 and PSD-95 (1:10,000 and 1:1000; Chem-
after transcardial perfusion with normal saline at Day 28. icon International, Temecula, CA, USA), and finally
By the method described previously [8, 14, 36, 44, 45], the incubated with horseradish peroxidase–conjugated immu-
brains were quickly removed and sectioned coronally, noglobulin (1:5000; Chemicon International) [8, 45].
followed by the staining of 0.5% cresyl violet. Brain Bound antibody was visualized with the Amersham ECL
infarction volumes were measured using a computerized system (GE Healthcare Bio-Sciences Corp., Piscataway,
image analyzer (MCID Elite, Imaging Research Inc., NJ, USA). Membranes were then probed for actin
Ontario, Canada) and were expressed as a percentage of (1:10,000; Chemicon International). Optical densities were
the contralateral hemisphere volume [8, 14, 36]. In addi- measured using a Luminescent Image Analyzer (Fujifilm
tion, individual cortical and subcortical (caudoputaminal LAS-3000; Fuji Photo Film Co., Tokyo, Japan).
and hippocampal) infarct sizes and surviving neurons were
separately calculated.
Gelatin zymography
Brain tissues were homogenized and centrifuged by the
Golgi–Cox analysis
method described previously [14–16]. Gelatinase activity in
At Day 1 or Day 28, animals were sacrificed, and the the supernatants was extracted and purified with gelatin-
brains were rapidly removed and immersed in a modified Sepharose 4B (GE Healthcare Bio-Sciences Corp., Piscata-
Golgi–Cox solution for 14 days and, then, in 30% way, NJ, USA) [14, 15]. Samples normalized for protein
sucrose solution for another 5 days [8]. Coronal brain concentration were mixed with sample buffer and loaded
sections of 200 lm were harvested using a vibratome onto 7.5% sodium dodecyl sulfate (SDS)–polyacrylamide
(VSLM1 motorized Vibroslice, Campden Instrument, gel containing 1.25 mg/mL gelatin. After electrophoresis,
Silbey, UK), mounted, and stained. The spine density of the gel was washed with 2.5% Triton X-100 buffer for
the second- and third-order basilar dendrites of the pyra- three times. The gel was incubated in 100 mL of 50 mM
midal neurons at regions corresponding to the ischemic Tris-HCl (pH 7.4) containing 5 mM CaCl2, 200 mM NaCl,
core (layer II–III), the inner (layer III–IV), and the outer and 0.2% Brij 35 at 37°C overnight. Gel was stained with
(layer V–VI) boundary zones of the forepaw and the 0.25% Coomassie blue R-250 (Sigma-Aldrich) and
hindpaw areas of the ischemic hemisphere, as well as at destained appropriately.
the layer V–VI region of the contralateral, intact brain,
was quantified and traced at 1000 magnification by a
Neurobehavioral testing and body weight
Zeiss Axioskop 2 Mot fluorescent microscope equipped
measurements
with an Axiocam high-resolution camera and AxioVision
image analysis system (Carl Zeiss, Oberkochen, Body weight measurement was taken daily. A battery of
Germany). sensory-motor tests was conducted prior to and on a daily
basis after reperfusion by two observers unaware of treat-
ment protocol. Briefly, two neurologic grading systems
Western blot analysis
were used: (i) a sensorimotor grading scale modified from
Samples were obtained from cultured neurons and from previously published methods [8, 14, 29, 36, 44, 45, 47], (ii)
brain tissues of the ischemic core and penumbral regions, a grading scale of 0–28 developed by Clark et al. [44, 45,
which were quickly dissected on dry ice after the animal 48].

215
Juan et al.

45.0%, compared with controls (n = 6; P < 0.05, respec-


Statistical analysis
tively; Fig. 2A,B).
All data were expressed as the mean standard error of Seven animals (10.1%) died prior to completing the
the mean (S.E.M.). Neurobehavioral scores and the den- recovery protocol and were excluded: Five were in the
dritic spine densities were analyzed by Mann–Whitney vehicle-injected group, and two was in the melatonin-trea-
U-test. Paired Student’s t-test was used to evaluate the ted group. Relative to controls, melatonin did not signifi-
response to a change in conditions, and unpaired Student’s cantly affect the LCBF either at the ischemic core and the
t-test/one-way ANOVA with Fisher’s protected least signifi- penumbral regions, or at the contralateral cortical brain,
cant difference (LSD) post hoc comparison was used to as assessed before and within 40 min after treatment
evaluate differences between groups. P < 0.05 was selected (P > 0.05; data not shown). The other physiologic param-
for statistical significance. eters of the animals were kept within normal physiologic
limits during the course of experiments and did not differ
significantly between melatonin-treated animals and vehi-
Results
cle-injected controls (P > 0.05; data not shown).
At 10-DIV cultured neurons, the addition of glutamate Brain infarction was reduced by 35.3% (P < 0.05) in
(300 lM) induced significant reductions in the dendritic the melatonin-treated animals (n = 9; Fig. 3), when com-
lengths and branches by 43.5% and 43.6%, respectively pared with controls (n = 7), following a recovery period of
(n = 10–14; P < 0.05; Fig. 1), relative to sham treatment, 28 days. This reflected the melatonin-mediated reductions
cultured neurons. Melatonin (20–100 lM), however, exhib- in cortical and subcortical infarct sizes by 36.9 and 30.0%
ited a neuroplasticity response by improving the dendritic (P < 0.05, respectively; Fig. 3) and increases in the num-
lengths and branches by 56.7–84.0 and 53.9–78.2%, bers of the surviving neurons in the penumbral cortex and
respectively, compared with the control-treated neurons caudoputamen by 246.7 and 289.2% (P < 0.001, respec-
exposed to glutamate excitotoxicity (P < 0.05; Fig. 1). tively; Fig. 3). Additionally, melatonin significantly
Additionally, melatonin (n = 6) improved the expressions improved sensory, motor, and the 28-point neurologic
of the GAP-43 and PSD-95 proteins by 116.4% and scores, but not the loss of body weight, taken at 1–28 days

Fig. 1. Melatonin improves dendritic


aborizations in cultured neurons expose to
glutamate excitotoxicity. In the microtu-
bule-associated protein-2 (MAP-2)-
immunostained neurons, treatment with
melatonin (20–100 lM) significantly
enhanced the numbers of total dendritic
lengths and branches, compared with
controls. *P < 0.05 versus vehicle data;
n = 10–14.

216
Melatonin enhanced postinsult neuroplasticity

(A) (B)

Fig. 2. Western immunoblot analysis for


GAP-43 and PSD-95 in cultured neurons
expose to glutamate excitotoxicity.
Melatonin significantly improved the
expressions of GAP-43 (A) and PSD-95
(B), compared with vehicle-treated
controls. *P < 0.05 versus vehicle data;
n = 6 for each groups.

Fig. 3. Treatment with melatonin


reduced ischemic brain damage in rats
subjected to middle cerebral artery
(MCA) occlusion following a recovery
period of 28 days. The cresyl violet-
stained coronal sections were from
representative animals, which received an
intravenous injection of vehicle (PEG-
saline) or melatonin (5 mg/kg) at 90 min
after the ischemic onset. Six random and
nonoverlapping (500 9 400 lm²) regions
in the borders of the ischemic parietal
cortex and caudoputamen were selected
for counting the surviving neurons. Scale
bar=5 mm. melatonin-treated animals
(n = 9) had significantly reduced the
infarction volume and individual cortical
and striatal lesion sizes, and significantly
increased numbers of surviving neurons,
compared with controls (n = 7).
*P < 0.05 versus vehicle data.

after the ischemic onset (P < 0.05; Table 1), compared B), compared with vehicle-treated controls (n = 6). Mela-
with controls. tonin also significantly improved the expressions of PSD-
Intravenous injection of melatonin (n = 6) at 5 mg/kg 95 at the penumbral cortical zones (P < 0.05; Fig. 5D),
effectively improved the activity of the pro- and MMP-9 but did not affected the PSD-95 levels at the ischemic core
at penumbral cortical zones (Fig. 4A–C) by 24.0 and regions (P > 0.05; Fig. 5C).
15.0% (P < 0.05, respectively), compared with controls At Day 1 and Day 28, vehicle-treated controls had a sig-
(n = 6). Melatonin, however, did not affect the levels of nificantly lower branches and density of the second- and
the pro- and MMP-9 activity at the ischemic core third-order basilar dendrites of the pyramidal neurons at
(P > 0.05). Melatonin also did not affect the activity of the ischemic core (layer II–III), the inner (layer III–IV)
MMP-2 either at the ischemic core and the penumbral cor- and the outer (layer V–VI) boundary zones of the ischemic
tical zones, but enhanced the MMP-2 activity by 22.6% at cortex, compared to sham-operated, intact controls
the contralateral intact brain (Fig. 4A,D; P < 0.05). (P < 0.01, respectively; Fig. 6A). Melatonin-treated
Cerebral ischemia–reperfusion insult induced prolonged animals (n = 8 for both groups), however, had a signifi-
reductions in the expressions of the PSD-95 and GAP-43 cantly improved numbers of mushroom spine density of
at the ischemic core and the penumbral cortical zones the second- and third-order basilar dendrites of the pyra-
(P < 0.05, respectively; Fig. 5A–D). Melatonin-treated midal neurons at the layers II–III, III–IV, and V–VI of
animals (n = 6), however, had a significantly improved the ischemic hemisphere at Day 1 and 28 (P < 0.05,
protein levels of the GAP-43 at the ischemic core and the respectively; Fig. 6A–C), relative to vehicle-injected con-
penumbral cortical zones (P < 0.05, respectively; Fig. 5A, trols (n = 8 for both groups). Melatonin-treated animals

217
Juan et al.
Table 1. Melatonin improves sensorimotor behavioral scores and weight loss after cerebral ischemia–reperfusion

Neurologic behavioral score

Time Weight loss (g) Motor Sensory 28-point clinical scale

Day 1 Vehicle (n = 7) 29  3.1 2 (1.6–2.4) 4 (3.7–4.3) 17 (15.3–18.7)


Melatonin (n = 9) 25  16.3 1 (0.6–1.4)* 2 (1.5–2.5)* 9 (6.6–11.4)*
Day 7 Vehicle (n = 7) 20  17.4 2 (1.5–2.4) 3 (2.7–3.3) 15.5 (14.6–16.5)
Melatonin (n = 9) 15  50.0 1 (0.6–1.4)* 2 (1.7–2.3)* 8 (6.7–9.3)*
Day 14 Vehicle (n = 7) 28  19.0 2.5 (2.1–2.9) 3.5 (2.9–4.1) 16 (14.4–17.6)
Melatonin (n = 9) 20  56.4 1 (0.7–1.3)* 2 (1.6–2.4)* 8 (6.6–9.4)*
Day 21 Vehicle (n = 7) 72  5.8 2 (1.6–2.4) 3 (2.5–3.5) 16 (14.3–17.7)
Melatonin(n = 9) 51  56.0 1 (0.6–1.4)* 2 (1.6–2.4)* 8 (7.2–8.8)*
Day 28 Vehicle (n = 7) 82  31 2 (15–2.5) 3 (2.4–3.6) 14 (12.9–15.1)
Melatonin (n = 9) 87  24.8 1 (0.8–1.2)* 2 (1.7–2.3)* 8 (7.0–9.0)*

Weight data and neurologic behavioral scores are expressed by mean  S.E.M. and by median (95% CI), respectively. Intravenous injec-
tion of melatonin (5 mg/kg) 90 min after the onset of MCA occlusion significantly improved sensorimotor neurologic scores, but not
weight loss, compared with vehicle-injected control values.
*P < 0.05 versus vehicle data.

(B)
(A)

(C) (D)

Fig. 4. Zymography of MMP-2 and


MMP-9 in the ischemic brain. (A) The
photographs show typical pattern of
changes in MMP-2 and MMP-9 at the
ischemic core (IC), penumbra (P), and the
contralateral intact, homotopic (L) areas.
Melatonin not only significantly improved
the pro- and MMP-9 activity (B and C,
respectively) at the ischemic penumbral,
but not ischemic core, areas, but also
enhanced the MMP-2 activity (D) at the
contralateral intact brain, compared with
controls. *P < 0.05 versus vehicle data;
n = 6, respectively.

also had a significant improvement in the density of den- (aborizations) in cultured neurons exposed to glutamate
dritic spines of pyramidal neurons at the layer V–VI excitotoxicity. We also demonstrated that melatonin
regions of the contralateral, intact brain at Day 1 not only improved neuronal dendritic spine density,
(P < 0.05, respectively; Fig. 6D,E), but did not affect the but also enhanced the PSD-95 and GAP-43 protein
contralateral dendritic spine density at Day 28 (P > 0.05; expressions and the MMP-9 activity at a subacute
Fig. 6D,E), compared with vehicle-treated controls. stage of recovery following cerebral ischemia–reperfu-
sion [9, 20, 24]. Another novel finding was that treat-
ment with melatonin reduced neuronal damage,
Discussion
improved neuronal survival and plasticity [8, 35], and,
This study showed that melatonin increased the thus, enhanced neurobehavioral outcomes following a
expressions of the PSD-95 and GAP-43 proteins, and long-term recovery period after transient focal cerebral
thus, improved dendritic spine lengths and branches ischemia in rats.

218
Melatonin enhanced postinsult neuroplasticity

(A) (B)

(C) (D)

Fig. 5. Western immunoblot analysis for


GAP-43 and PSD-95 in the ischemic
brain. The photographs showed typical
changes in the GAP-43 and PSD-95
protein expressions in the ischemic core
(A, C) and the penumbral (B, D) cortices
at Day 7 after ischemia. Densitometric
analyses showed that melatonin
significantly improved the expression in
the GAP-43 protein in the ischemic core
(A) and penumbral (B) cortices.
Melatonin also improved the PSD-95
protein at the penumbral (D), but not the
ischemic core (C), brain tissues.
*P < 0.05 versus control; n = 6 (each
column).

The neuroprotection and neuroplasticity observed here contralateral diaschisis in dendritic spine density returned
at the levels of the neuronal stoma, dendritic spine density, to normal levels after a recovery period of 28 days in con-
postsynaptic spines, axonal sprouting, and ECM remodel- trols and melatonin-treated animals. Interestingly, melato-
ing cannot be accounted for by changes in hemodilution (as nin not only improved the MMP-9 activity and the
measured by blood hematocrit), arterial blood pressure, expressions of PSD-95 and GAP-43 proteins in the pen-
heart rate, glucose, core temperature, or differences in umbral brain at Day 7, but also enhanced the MMP-2
LCBF, because these values were not significantly different levels at the contralateral homotopic intact brain [9, 20,
when compared between vehicle-injected and melatonin- 25]. This melatonin-mediated increase in the MMP-2 levels
treated animals [44, 48]. The finding that improvements for at Day 7 was correlated with the improved contralateral
the dendritic aborizations, postdendritic spine, and axonal neuroplasticity observed in melatonin-treated animals dur-
sprouting were also be observed in the cultured neurons ing the early recovery period (Day 1 and Day 7), as
exposed to glutamate further justified the melatonin-medi- observed here and reported previously [8]. Accordingly,
ated long-lasting neuroplasticity, as shown here. the data supported that melatonin improved functional
We have observed two specific patterns of reductions of deficits after stroke by providing direct neuroplasticity to
the dendritic spine density in macroscopically intact brain damaged brain tissues, and also by reducing remote brain
tissues at Day 1 after transient MCA occlusion: (i) changes injury, or, using other intact brain to compensate for the
in the peri-infarct penumbral regions and (ii) remote lost function during a course of healing.
changes contralateral to the side of the infarct [8, 27, 48, Optimal sensorimotor function requires both anatomical
49]. Consistent with the Day-7 data reported previously and functional linkages among the neuronal soma, axons,
[8], we found that at Day 1, both the two patterns of dendritic spines, synapses as well as the ECM integrity [8,
reduced dendritic spine density could be improved with 26]. These structures and their axonal synaptodendritic
melatonin either in the peri-infarct penumbral or the connections as well as the ECM integrity are involved in
remote contralateral, intact brain regions. At Day 28, mel- the processing and integration of sensorimotor informa-
atonin, however, only significantly enhanced the reduced tion [8, 9, 26]. The marked and prolonged reductions of
dendritic spine density in the penumbral brain, because the dendritic spine density in the peri-infarct penumbral

219
Juan et al.

(A)

Fig. 6. Photomicrographs of representative


dendritic segments from pyramidal neurons
sampled at regions corresponding to the
inner (layer III–IV) boundary zones of the
ischemic damage in sham-operated controls
(A, right upper panel), vehicle-injected
ischemic controls (A, left lower panel),
and melatonin-treated animals (A, right
(B) (C) lower panel) at Day 28 following stroke.
Scale bar = 40 lm; = 15 lm in the
magnification inset. The left upper panel
in A indicates the 4 nonoverlapping
sampling sites selected for dendritic spine
counting in the study. Vehicle-injected
ischemic controls had a decreased
dendritic spine density in the ischemic
core, inner and outer boundary zones of
ischemia (Fig. A), compared with sham-
operated controls. At Day 1 and Day 28,
melatonin-treated animals, however, had
a significantly improved density of
mushroom spines of the second- (A, B)
and third-order (A, C) dendrites at the
layers II–III, III–IV, and V–VI of the
(D) (E) ischemic hemisphere, relative to vehicle-
injected controls. In addition, we have
observed that, at Day 1 but not Day 28,
vehicle-injected ischemic controls had a
significantly lower density of the second-
and third-order basilar dendritic spines of
pyramidal neuron at the layer V–VI
region of the contralateral, intact brain
(D, E), compared with sham-operated
controls. Melatonin treatment could
effectively reverse the reduction in the
dendritic spines at Day 1 in the
contralateral, intact brain (D, E). n = 8
for each group; *P < 0.05 versus vehicle
data.

region indicate that there may be relatively widespread and ipsilateral and remote excitability changes observed after
persisting functional disturbances in this region [8, 48, 49]. stroke; this needs further evaluation [8, 50]. One of the
The findings with improved neurobehavioral outcomes most obvious explanations for the diaschisis changes in
observed with melatonin following a long-term recovery dendritic spine density observed at Day 1 and Day 7 at
further justify its ability to improve neuroplasticity at the the contralateral homotopic brain is transhemispheric/
postsynaptic, axonal sprouting, dendritic spine density, and transcallosal deafferentation [51]. Whether the changes in
the ECM levels following ischemic stroke, as observed here. the MMP-2 level of the contralateral brain observed here
Previous findings that melatonin and its main brain metab- account for the severity of the contralateral diaschisis
olites worked well to inhibit nNOS activity in the ischemic needs further evaluation [9, 10]. Both the peri-infarct and
brain also validated its ability to enhance the PSD-95-medi- the remote changes in the dendritic spine density and other
ated synaptic plasticity observed here [36–39]. molecular signaling cascades may turn out to be valuable
A variety of mechanisms including free radical forma- indicators of functional disturbances, which could become
tion, disordered neurotransmitter storage, uptake and targets of therapeutic interventions at the early and
release, or a general stress response, may underlie the subacute stage after stroke [8].

220
Melatonin enhanced postinsult neuroplasticity

We have observed that, perhaps via different mecha- 2. ROMERO JR, BABIKIAN VL, KATZ DI et al. Neuroprotection
nisms of action (e.g., MMP-9 and MMP-2, respectively), and stroke rehabilitation: modulation and enhancement of
melatonin overcame poststroke functional deficits by recovery. Behav Neurol 2006; 17:17–24.
improving in situ and remote neuroplasticity at both the 3. RAMIC M, EMERICK AJ, BOLLNOW MR et al. Axonal plasticity
penumbral brain and those intact brain tissues distal from is associated with motor recovery following amphetamine
the area of injury [8, 49, 51]. The present study and our treatment combined with rehabilitation after brain injury in
previous work also indicate that melatonin can modulate the adult rat. Brain Res 2006; 1111:176–186.
4. SIZONENKO SV, BEDNAREK N, GRESSENS P. Growth factors
MMPs and therefore exhibit neuroprotective or plasticity/
and plasticity. Semin Fetal Neonatal Med 2007; 12:241–249.
trophic actions depending on different time course and/or
5. LEGOS JJ, BARONE FC. Update on pharmacological strategies
molecular basis of ischemic stroke [14–16]. Thus, melato-
for stroke: prevention, acute intervention and regeneration.
nin is a potent regulator of MMPs at various stages of
Curr Opin Investig Drugs 2003; 4:847–858.
injury [14–16, 52]. Recent evidence also strongly support 6. HAAS S, WEIDNER N, WINKLER J. Adult stem cell therapy in
that tight binding of melatonin in the active site is stroke. Curr Opin Neurol 2005; 18:59–64.
involved in modulating the catalytic activity of MMP-9 7. HAGGT T. From neurotransmitters to neurotrophic factors to
[52]. Additional studies are, however, required to decipher neurogenesis. Neuroscientist 2009; 15:20–27.
more molecular events through which melatonin regulates 8. CHEN HY, HUNG YC, CHEN TY et al. Melatonin improves
the activity of the MMPs and leads to the improved long- presynaptic protein, SNAP-25, expression and dendritic spine
term neuroplasticity observed here. density and enhances functional and electrophysiological
It has been shown that neuroprotective effects of mela- recovery following transient focal cerebral ischemia in rats.
tonin against acute brain damage do not depend on mel- J Pineal Res 2009; 47:260–270.
atonin receptors in a mouse model of ischemic stroke 9. MICHALUK P, WAWRZYNIAK M, ALOT P et al. Influence of
[53]. Further studies are needed to clarify whether mela- matrix metalloproteinase MMP-9 on dendritic spine morphol-
tonin receptors are involved in the melatonin-mediated ogy. J Cell Sci 2011; 124:3369–3380.
neuronal growth/maturation, neuroplasticity, and neuro- 10. FRAGKOULI A, PAPATHEODOROPOULOS C, GEORGOPOULOS S
genesis during the subacute and long-term stages of et al. Enhanced neuronal plasticity and elevated endogenous
ischemic stroke as observed here and reported previously sAPPa levels in mice over-expressing MMP9. J Neurochem
[8, 53–55]. Additional studies are also needed to clarify 2012; 121:239–251.
the melatonin’s beneficial role in the field of human 11. OLIVEIRA-SILVA P, JURGILAS PB, TRINDADE P et al. Matrix me-
talloproteinase-9 is involved in the development and plasticity
stroke, although a lack of economic benefits for pharma-
of retinotectal projections in rats. NeuroImmunoModulation
cological companies may actually hinder its further
2007; 14:144–149.
clinical trials.
12. HAYAKAWA K, NAKANO T, IRIE K et al. Inhibition of reactive
In summary, we demonstrate that administration with
astrocytes with fluorocitrate retards neurovascular remodeling
melatonin induces in vitro and in vivo neuroprotection and and recovery after focal cerebral ischemia in mice. J Cereb
neuroplasticity through improving the PSD-95 and GAP- Blood Flow Metab 2010; 30:871–882.
43 protein expressions. In addition, we show that melato- 13. BELL KF, BENT RJ, MEESE-TAMURI S et al. Calmodulin
nin decreases poststroke diaschisis and enhances long-term kinase IV-dependent CREB activation is required for neuro-
neuroplasticity probably via upregulating the MMP-2 and protection via NMDA receptor-PSD95 disruption. J Neuro-
MMP-9 in the nonischemic and ischemic brain, respec- chem 2013; 126:274–287.
tively. As melatonin offers the advantage for improving 14. HUNG YC, CHEN TY, LEE EJ et al. Melatonin decreases
postischemic neuroplasticity for both the penumbral and matrix metalloproteinase-9 activation and expression and
the contralateral, intact brain, it may be worthwhile fur- attenuates reperfusion-induced hemorrhage following tran-
ther investigating in the field of ischemic stroke in human sient focal cerebral ischemia in rats. J Pineal Res 2008;
beings. 45:459–461.
15. TAI SH, CHEN HY, LEE EJ et al. Melatonin inhibits postis-
chemic matrix metalloproteinase-9 (MMP-9) activation via
Acknowledgements dual modulation of plasminogen/plasmin system and endoge-
This research was supported by a grant from the National nous MMP inhibitor in mice subjected to transient focal cere-
Science Council of Taiwan (NSC No. 99-2314-B-006-022- bral ischemia. J Pineal Res 2010; 49:332–341.
MY3). A.H.L. was the recipient of the summer student- 16. CHANG CC, TIEN CH, LEE EJ et al. Melatonin inhibits matrix
metalloproteinase-9 (MMP-9) activation in the lipopolysac-
ship of Department of Occupational Safety and Heath,
charide (LPS)-stimulated RAW 264.7 and BV2 cells and a
Chang Jung University, Tainan, Taiwan. A.C.L. was the
mouse model of meningitis. J Pineal Res 2012; 53:188–197.
recipient of the summer studentship of Chemistry and
17. BOZDAGI O, NAGY V, KWEI KT et al. In vivo roles for matrix
Biochemistry Department, University of California Los
metalloproteinase-9 in mature hippocampal synaptic physiol-
Angeles, CA, USA. ogy and plasticity. J Neurophysiol 2007; 98:334–344.
18. BARKHO BZ, MUNOZ AE, LI X et al. Endogenous matrix
References metalloproteinase (MMP)-3 and MMP-9 promote the differ-
entiation and migration of adult neural progenitor cells in
1. DEMAERSCHALK BM, YIP TR. Economic benefit of increasing response to chemokines. Stem Cells 2008; 26:3139–3149.
utilization of intravenous tissue plasminogen activator for 19. WOJCIK L, SAWICKA A, RIVERA S et al. Neurogenesis in
acute ischemic stroke in the United States. Stroke 2005; gerbil hippocampus following brain ischemia: focus on the
36:2500–2503.

221
Juan et al.
involvement of metalloproteinases. Acta Neurobiol Exp estrogen on its hormetic dose-response. J Pineal Res 2011;
(Wars) 2009; 69:52–61. 50:292–303.
20. FAN J, VASUTA OC, ZHANG LY et al. N-methyl-D-aspartate 37. CHEN HY, CHEN TY, LEE MY et al. Melatonin decreases
receptor subunit- and neuronal-type dependence of excitotox- neurovascular oxidative/nitrosative damage and protects
ic signaling through post-synaptic density 95. J Neurochem against early increases in the blood-brain barrier permeability
2010; 115:1045–1056. after transient focal cerebral ischemia in mice. J Pineal Res
21. YAN BC, PARK JH, AHN JH et al. Postsynaptic density pro- 2006; 41:175–182.
tein (PSD)-95 expression is markedly decreased in the hippo- 38. PEI Z, CHEUNG RT. Pretreatment with melatonin exerts anti-
campal CA1 region after experimental ischemia-reperfusion inflammatory effects against ischemia/reperfusion injury in a
injury. J Neurol Sci 2013; 330:111–116. rat middle cerebral artery occlusion stroke model. J Pineal
22. SUN HS, DOUCETTE TA, LIU Y et al. Effectiveness of PSD95 Res 2004; 37:85–91.
inhibitors in permanent and transient focal ischemia in the 39. TJONG YW, LI MF, HUNG MW et al. Melatonin ameliorates
rat. Stroke 2008; 39:2544–2553. hippocampal nitric oxide production and large conductance
23. ZHOU L, LI F, XU HB et al. Treatment of cerebral ischemia calcium-activated potassium channel activity in chronic inter-
by disrupting ischemia-induced interaction of nNOS with mittent hypoxia. J Pineal Res 2008; 44:234–243.
PSD-95. Nat Med 2010; 16:1439–1443. 40. BETTAHI I, POZO D, OSUNA C et al. Melatonin reduces nitric
24. STROEMER RP, KENT TA, HULSEBOSCH CE. Neocortical oxide synthase activity in rat hypothalamus. J Pineal Res
neural sprouting, synaptogenesis, and behavioral recovery 1996; 20:205–210.
after neocortical infarction in rats. Stroke 1995; 26:2135– 41.  J, ESCAMES G, RODRIGUEZ MI et al. Inhibition of neuro-
LEON
2144. nal nitric oxide synthase activity by N1-acetyl-5-methoxyky-
25. BUFFO A, HOLTMAAT AJ, SAVIO T et al. Targeted overexpres- nuramine, a brain metabolite of melatonin. J Neurochem
sion of the neurite growth-associated protein B-50/GAP-43 in 2006; 98:2023–2033.
cerebellar Purkinje cells induces sprouting after axotomy but 42. LEE WT, LIN MH, LEE EJ et al. Magnolol reduces gluta-
not axon regeneration into growth-permissive transplants. mate-induced neuronal excitotoxicity and protects against
J Neurosci 1997; 17:8778–8791. permanent focal cerebral ischemia up to 4 hr. PLoS ONE
26. STROEMER RP, KENT TA, HULSEBOSCH CE. Enhanced neocor- 2012; 7:e39952.
tical neural sprouting, synaptogenesis, and behavioral recov- 43. LEE EJ, LEE MY, CHANG GL et al. Delayed treatment with
ery with D-amphetamine therapy after neocortical infarction magnesium: reduction of brain infarction and improvement
in rats. Stroke 1998; 29:2381–2395. of electrophysiological recovery following transient focal cere-
27. LEE EJ, WU TS, LEE MY et al. Delayed treatment with bral ischemia in rats. J Neurosurg 2005; 102:1085–1093.
melatonin enhances electrophysiological recovery following 44. CHEN TY, TAI SH, LEE EJ et al. Cinnamophilin offers pro-
transient focal cerebral ischemia in rats. J Pineal Res 2004; longed neuroprotection against gray and white matter dam-
36:33–42. age and improves functional and electrophysiological
28. REITER RJ, TAN DX, LEON J et al. When melatonin gets on outcomes after transient focal cerebral ischemia. Crit Care
your nerves: its beneficial actions in experimental models of Med 2011; 39:1130–1137.
stroke. Exp Biol Med 2005; 230:104–117. 45. CHEN TY, LIN MH, LEE WT et al. Nicotinamide inhibits
29. KOH PO. Melatonin attenuates decrease of protein phospha- nuclear factor-kappa B translocation after transient focal
tase 2A subunit B in ischemic brain injury. J Pineal Res 2012; cerebral ischemia. Crit Care Med 2012; 40:532–537.
52:57–61. 46. YELESWARAM K, MCLAUGHLIN LG, KNIPE JO et al. Pharma-
30. TAN DX, CHEN LD, POEGGELER B et al. Melatonin: a potent, cokinetics and oral bioavailability of exogenous melatonin in
endogenous hydroxyl radical scavenger. Endocr J 1993; 1:57– preclinical animal models and clinical implications. J Pineal
60. Res 1997; 22:45–51.
31. TAN DX, MANCHESTER LC, TERRON MP et al. One molecule, 47. LEE EJ, WU TS, CHANG GL et al. Delayed treatment with
many derivatives: a never-ending interaction of melatonin nicotinamide inhibits brain energy depletion, improves cere-
with reactive oxygen and nitrogen species? J Pineal Res 2007; bral microperfusion, reduces brain infarct volume, but does
42:28–42. not alter neurobehavioral outcome following permanent focal
32. PEYROT F, DUCROCQ C. Potential role of tryptophan deriva- cerebral ischemia in Sprague Dawley rats. Curr Neurovasc
tives in stress responses characterized by the generation of Res 2006; 3:203–213.
reactive oxygen and nitrogen species. J Pineal Res 2008; 48. CLARK WM, RINKER LG, LESSOV NS et al. Lack of interleu-
45:235–246. kin-6 expression is not protective against focal central
33. GALANO A, TAN DX, REITER RJ. On the free radical scaveng- nervous system ischemia. Stroke 2000; 31:1715–1720.
ing activities of melatonin’s metabolites, AFMK and AMK. 49. HUNG YC, CHOU YS, CHANG CH et al. Early reperfusion
J Pineal Res 2013; 54:245–257. improves the recovery of contralateral electrophysiological di-
34. TAN DX, REITER RJ, MANCHESTER LC et al. Chemical and aschisis following focal cerebral ischemia in rats. Neurol Res
physical properties and potential mechanisms: melatonin as a 2010; 32:828–834.
broad spectrum antioxidant and free radical scavenger. Curr 50. NEUMANN-HAEFELIN T, WITTE OW. Periinfarct and remote
Top Med Chem 2002; 2:181–197. excitability changes after transient middle cerebral artery
35. LEE EJ, LEE MY, CHEN HY et al. Melatonin attenuates gray occlusion. J Cereb Blood Flow Metab 2000; 20:45–52.
and white matter damage in a mouse model of transient focal 51. REDECKER C, WANG W, FRITSCHY JM et al. Widespread and
cerebral ischemia. J Pineal Res 2005; 38:42–52. long-lasting alterations in GABA(A)-receptor subtypes after
36. TAI SH, HUNG YC, LEE EJ et al. Melatonin protects against focal cortical infarcts in rats: mediation by NMDA-depen-
transient focal cerebral ischemia in both reproductively active dent processes. J Cereb Blood Flow Metab 2002; 22:1463–
and estrogen-deficient female rats: the impact of circulating 1475.

222
Melatonin enhanced postinsult neuroplasticity
52. RUDRA DS, PAL U, MAITI NC et al. Melatonin inhibits 54. YIP HK, CHANG YC, WALLACE CG et al. Melatonin treat-
matrix metalloproteinase-9 activity by binding to its active ment improves adipose-derived mesenchymal stem cell ther-
site. J Pineal Res 2013; 54:398–405. apy for acute lung ischemia-reperfusion injury. J Pineal Res
53. KILIC U, YILMAZ B, UGUR M et al. Evidence that 2013; 54:207–221.
membrane-bound G protein-coupled melatonin receptors 55. LIU J, SOMERA-MOLINA KC, HUDSON RL et al. Melatonin
MT1 and MT2 are not involved in the neuroprotective effects potentiates running wheel-induced neurogenesis in the dentate
of melatonin in focal cerebral ischemia. J Pineal Res 2012; gyrus of adult C3H/HeN mice hippocampus. J Pineal Res
52:228–235. 2013; 54:222–231.

223

You might also like