Accepted Manuscript Direct pharmacological Akt activation rescues Alzheimer's disease like memory impairments and aberrant synaptic plasticity Jee Hyun Yi, Soo Ji Baek, Sunghoo Heo, Hye Jin Park, Huiyoung Kwon, Seungheon Lee, Jiwook Jung, Se Jin Jeon, Byung C. Kim, Young Choon Lee, Jong Hoon Ryu, Dong Hyun Kim PII:
S0028-3908(17)30494-X
DOI:
10.1016/j.neuropharm.2017.10.028
Reference:
NP 6913
To appear in:
Neuropharmacology
Received Date: 22 March 2017 Revised Date:
23 September 2017
Accepted Date: 21 October 2017
Please cite this article as: Yi, J.H., Baek, S.J., Heo, S., Park, H.J., Kwon, H., Lee, S., Jung, J., Jeon, S.J., Kim, B.C., Lee, Y.C., Ryu, J.H., Kim, D.H., Direct pharmacological Akt activation rescues Alzheimer's disease like memory impairments and aberrant synaptic plasticity, Neuropharmacology (2017), doi: 10.1016/j.neuropharm.2017.10.028. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT Direct pharmacological Akt activation rescues Alzheimer’s disease like memory impairments and aberrant synaptic plasticity
RI PT
Jee Hyun Yi1,#, Soo Ji Baek2,#, Sunghoo Heo2, Hye Jin Park3, Huiyoung Kwon3, Seungheon Lee4, Jiwook Jung5, Se Jin Jeon6, Byung C. Kim2, Young Choon Lee3,9,
1
SC
Jong Hoon Ryu7,8,*, Dong Hyun Kim3,9,*
School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol,
2
M AN U
Bristol, UK.
Department of Neurology, Chonnam National University Medical School, Gwangju,
Republic of Korea 3
Department of Medicinal Biotechnology, College of Health Sciences, Dong-A
4
TE D
University, Busan, Republic of Korea
Department of Aquatic Biomedical Sciences, School of Marine Biomedical Science,
College of Ocean Science, Jeju National University, Jeju, Republic of Korea Department of Herbal Medicinal Pharmacology, College of Herbal Bio-industry,
EP
5
Daegu Haany University, Kyungsan, Republic of Korea School of Natural Resources and Environmental Science, Kangwon National
AC C
6
University, ChoonCheon, Republic of Korea 7
Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee
University, 1 Hoeki-dong, Dongdaemoon-Gu, Seoul, Republic of Korea 8
Department of Life and Nanopharmaceutical Sciences, Kyung Hee University, 1
Hoeki-dong, Dongdaemoon-Gu, Seoul, Korea. 9
Institute of Convergence Bio-Health, Dong-A University, Busan, Republic of Korea
1
ACCEPTED MANUSCRIPT #
These authors are contributed equally in this study.
*Corresponding authors: Dong Hyun Kim
RI PT
Email:
[email protected] Tel: +82-51-200-7583 Fax: +82-51-200-7588
SC
Jong Hoon Ryu Email:
[email protected]
M AN U
Tel: +82-2-961-9230
AC C
EP
TE D
Fax: +82-2-961-0369
2
ACCEPTED MANUSCRIPT Abstract Amyloid β (Aβ) is a key mediator for synaptic dysfunction and cognitive impairment implicated in Alzheimer’s disease (AD). However, the precise mechanism of the toxic
RI PT
effect of Aβ is still not completely understood. Moreover, there is currently no treatment for AD. Protein kinase B (PKB, also termed Akt) is known to be aberrantly regulated in the AD brain. However, its potential function as a therapeutic target for
SC
AD-associated memory impairment has not been studied. Here, we examined the role of a direct Akt activator, SC79, in hippocampus-dependent memory impairments
M AN U
using Aβ-injected as well as 5XFAD AD model mice. Oligomeric Aβ injections into the 3rd ventricle caused concentration-dependent and time-dependent impairments in learning/memory and synaptic plasticity. Moreover, Aβ aberrantly regulated caspase-3, GSK-3β, and Akt signaling, which interact with each other in the
TE D
hippocampus. Caspase-3 and GSK-3β inhibitor ameliorated memory impairments and synaptic deficits in Aβ-injected AD model mice. We also found that pharmacological activation of Akt rescued memory impairments and aberrant
EP
synaptic plasticity in both Aβ-treated and 5XFAD mice. These results suggest that
AC C
Akt could be a therapeutic target for memory impairment observed in AD.
Key words: Amyloid β; Alzheimer’s disease; Akt; Long-term potentiation; Memory
1
ACCEPTED MANUSCRIPT 1
Introduction Alzheimer’s disease (AD) is a neurodegenerative disease, whose main symptom
3
is memory impairment. Amyloid β (Aβ) and Tau pathologies are known as main
4
processes in the progression of AD and blocking of these pathologies has long been
5
believed to be the therapeutic goal for AD. However, there is still no medical
6
approach approved to target these hallmarks of the disease.
RI PT
2
Aβ is aggregated and deposited in the AD brain and directly suppresses functions
8
of neurons (Palop and Mucke, 2010) and causes inflammation (Tuppo and Arias,
9
2005) and oxidative stress (Butterfield, 2002) as a secondary effect. Because
10
previous studies revealed that Aβ induces synaptic dysfunction and memory
11
impairment (Haass and Selkoe, 2007; Selkoe, 2008; Shankar et al., 2008) at an
12
early stage of AD, studies investigating the precise mechanisms and ways of
13
blocking the effects of Aβ have long been believed to help curing AD symptoms
14
(Klein, 2002; Klein et al., 2001; Kotilinek et al., 2008; Wang et al., 2016b). Therefore,
15
an excessive number of studies have been conducted to uncover the toxic
16
mechanisms of Aβ.
EP
TE D
M AN U
SC
7
Akt, protein kinase B, is an enzyme that plays important roles in cellular processes,
18
including glucose metabolism (Kohn et al., 1996; Sakoda et al., 2003), apoptosis
19
(Song et al., 2005), and cell proliferation (Vivanco and Sawyers, 2002). Activation of
20
Akt is induced via phosphorylation by mTORC2 (Sarbassov et al., 2005) and PDK1
21
(Franke, 2008) and this activation has been reported to play an important role in
22
synaptic plasticity and memory formation (Horwood et al., 2006). Aberrant regulation
23
of Akt has reported in AD brain. Several studies revealed increased activation of Akt
24
in AD (Griffin et al., 2005; Rickle et al., 2004; Talbot et al., 2012) but, opposite result
25
is also exist (Lee et al., 2009). However, Aβ, itself, may suppress the activity of Akt
AC C
17
1
ACCEPTED MANUSCRIPT (Abbott et al., 2008; Chen et al., 2009; Magrane et al., 2005) and several effective
2
drugs, which are reported as candidates for the treatment of AD, activate Akt
3
indirectly (Ali and Kim, 2015; Jin et al., 2005; Yi et al., 2016). Aβ induces Akt
4
cleavage through caspase-3 activation, resulting in activation of GSK-3β (Jo et al.,
5
2011). Moreover, because activation of Akt-related molecules with drugs ameliorates
6
Aβ toxicity (Ahmad et al., 2016; Baki et al., 2004; Ma et al., 2009; Xing et al., 2011),
7
the regulation of Akt might be a good target for investigation. However, the effect of
8
the direct regulation of Akt on AD pathology has not been studied.
SC
RI PT
1
In the present study, we examined the effect of AC79, a direct activator of Akt, on
10
Aβ-induced synaptic plasticity and memory impairments using an Aβ injection model
11
as well as a 5XFAD transgenic AD mouse model.
12 Methods and Materials
14
1. Animals
TE D
13
M AN U
9
Male C57BL6 mice (30 - 34 g, 10 weeks) were purchased from the SAMTAKO
16
biokorea (Osansi, Korea), and kept in the University Animal Care Unit for 1 week
17
prior to the experiments. The animals were housed 4 per cage, allowed access to
18
water and food ad libitum; the environment was maintained at a constant
19
temperature (23 ± 1 °C) and humidity (60 ± 10%) under a 12-h light/dark cycle (the
20
lights were on from 07:30 to 19:30). 5XFAD male mice were received from Jackson
21
Laboratory (Jackson Laboratory, USA) and crossbred with hybrid B6SJLF1 (from
22
Taconic) female mice. The offspring, heterozygous transgenic and littermate wild
23
type (WT) male mice, were used. The mice were housed in individual ventilated
24
cages (IVC) with access to water and food ad libitum, under a 12-h light/dark cycle
25
(the lights were on from 07:30 to 19:30). The treatment and maintenance of the
AC C
EP
15
2
ACCEPTED MANUSCRIPT animals were carried out in accordance with the Animal Care and Use Guidelines
2
Dong-A University, Korea. All of the experimental protocols using animals were
3
approved by the Institutional Animal Care and Use Committee of Dong-A University,
4
Korea.
RI PT
1
5 6
2. Cannulae implantation
Cannulae were implanted into the 3rd ventricle. Mice were anesthetized with Zoletil
8
50 (10 mg/kg, i.m.) and placed in a stereotaxic instrument (David Kopf Instruments,
9
Tujunga, CA). Guide cannulae (26 G) were aimed at the 3rd ventricle (stereotaxic
10
coordinates: AP, -2.00 mm; ML, 0 mm; DV, -2.00 mm) using an atlas of the mouse
11
brain (Paxinos and Franklin, 2004). The guide cannulae were fixed to the skul with
12
dental cement and covered with dummy cannulae. To inject drugs, mice were
13
carefully restrained by hand and infused with drugs through an injector cannula (30
14
G) at 0.25 µl/min). After 2 min of infusion, the infusion needle remained in the guide
15
cannula for 1 min more to ensure proper delivery of the drugs.
TE D
M AN U
SC
7
EP
16 3. Aβ or drugs injection
18
Add 1.0% NH4OH directly to the Aβ1-42 (35 - 40 µl to 0.5 mg peptide or 70-80 µl to 1
19
mg peptide). Then this solution was immediately diluted with 1 X PBS to a
20
concentration of 1 mg/ml. The solution was gently vortexed and sonicated at room
21
temperature until fully imscible. Aβ1-42 (10 µM) was incubated at 37 °C for 1 week,
22
and then 3 µl of Aβ or vehicle were acutely injected into the intracerebroventricle. Z-
23
DEVD-fmk, SC79, and TCS2002 were purchased from Tocris Bioscience (Ellisville,
24
MO). Z-DEVD-fmk (cell-permaeble, irreversible inhibitor of caspase-3/CPP32, Tocris
25
Bioscience, 30 µM/3 µl) (Fan et al., 2014), TCS2002 (Potent GSK-3β inhibitor, Tocris
AC C
17
3
ACCEPTED MANUSCRIPT Bioscience, 10 µM/3 µl) (Saitoh et al., 2009) and SC79 (Akt activator, Tocris
2
Bioscience, 100 µg/3 µl) (Wang et al., 2016a) was dissolved in 0.4%
3
dimethylsulphoxide (DMSO, MC/B, Norwood, OH, U.S.A.) in 0.1 M phosphate-
4
buffered saline (PBS; pH 7.4). Drugs were injected into 3rd ventricle.
RI PT
1
5 6
4. Inhibitory avoidance test
We assessed training and test trials for 2 days using the inhibitory avoidance task.
8
Testing was performed in a box consisting of two identical chambers (20 × 20 × 20
9
cm) with one chamber illuminated with a 50-W bulb and another non-illuminated; the
10
two chambers were separated by a guillotine door (5 × 5 cm). The floor of the non-
11
illuminated compartment was comprised of 2-mm stainless steel rods spaced 1 cm
12
apart as described previously (Kim et al., 2006). In the training trial, the mice were
13
initially placed in the illuminated compartment. The door between the two
14
compartments was opened 10 s later and was automatically closed when the mice
15
entered the non-illuminated compartment. A 3 s electrical foot shock (0.5 mA) was
16
then delivered through the stainless steel rods. The test trial was conducted 24 h
17
after the acquisition trial by returning the individual mice to the illuminated
18
compartment. In both trials, latency to enter was defined as the time it took for the
19
mouse to enter the dark compartment after the door was opened. During the
20
acquisition trial, the mice that did not enter the non-illuminated compartment within
21
60 s of the door opening the door were gently introduced into the dark chamber, and
22
the latency to enter was recorded as 60 s. Latencies were recorded for up to 300 s
23
during the retention trial.
AC C
EP
TE D
M AN U
SC
7
24 25
5. Y-maze test
4
ACCEPTED MANUSCRIPT The Y-maze is a three-arm horizontal maze (40 cm long and 3 cm wide with 12 cm
2
high walls) in which the arms are symmetrically disposed at 120° angles to each
3
other. The maze floor and walls were constructed from dark opaque polyvinyl plastic
4
as previously described (Kim et al., 2006). Mice were initially placed in one arm, and
5
the sequence and numbers of arm entries were manually recorded for each mouse
6
over an 8-min period. An alternation was defined as the consecutive entry into all
7
three arms (i.e., ABC, CAB, or BCA, but not BAB). The maze arms were thoroughly
8
cleaned with water between each test to remove residual odors and residues. The
9
alternation score (%) for each mouse was defined as the ratio of the actual number
10
of alternations to the total number possible (defined as the total number of arm
11
entries minus two) multiplied by 100 as shown by the following equation: %
12
Alternation = [(Number of alternations) / (Total arm entries – 2)] × 100. The number
13
of arm entries was used as an indicator of locomotor activity.
15
SC
M AN U
TE D
14
RI PT
1
6. Object recognition test
Mice were habituated to the open field (25 cm x 25 cm x 25 cm) with an internal
17
cue on one of the four walls for 10 min. Thirty minutes after the habituation; mice
18
were re-placed in the same box with two distinct objects. The objects consisted of a
19
glass box and plastic cylinder. Mice were allowed to freely explore the objects for 10
20
min. After 2 h, mice were placed back in the same box for the test phase. The two
21
objects were again present, but one object was now displaced to a novel spatial
22
location. Mice were allowed to freely explore the environment and the objects for 5
23
min. Time spent exploring the displaced and non-displaced objects were measured.
24
Preference ratio was calculated by following formula: Tdisplaced or Tnon-displaced/(Tdisplaced
AC C
EP
16
5
ACCEPTED MANUSCRIPT 1
+ Tnon-displaced) x 100. Tdisplaced, time spent exploring displaced object. Tnon-displaced, time
2
spent exploring non-displaced object.
3 7. Electrophysiology
RI PT
4
Mouse hippocampal slices were prepared using micro-vibratome (Lafayette-
6
campden neuroscience TM). The brain was rapidly removed and placed in ice-cold
7
artificial cerebrospinal fluid (ACSF; bubbled with 95% O2/5% CO2), which comprised:
8
(mM) NaCl, 124; KCl, 3; NaHCO3, 26; NaH2PO4, 1.25; CaCl2, 2; MgSO4, 1; D-
9
glucose, 10. Transverse hippocampal slices (400 µm thick) were prepared.
10
Hippocampal slices were submerged in ACSF (20–25 °C) for 2h before transfer to
11
the recording chamber (28–30 °C, flow rate ∼3 ml/min) as required. Field recordings
12
were made from stratum pyramidale in area CA1. Stimulating electrode was placed
13
in the Schaffer collateral-commissural pathway. Stimuli (constant voltage) were
14
delivered at 30 s intervals. To induce LTP, theta burst stimulation (5 trains of 4
15
pulses at 100 Hz) was delivered. The slope of the evoked field potential responses
16
were averaged from four consecutive recordings (fEPSPs) evoked at 30 s intervals.
M AN U
TE D
EP
18
8. Western blot analysis
AC C
17
SC
5
19
For the preparation of Western blot samples, coronal-sliced hippocampal tissues
20
were made using micro-vibratome (Lafayette-campden neuroscience TM). Then the
21
stratum radiatum, enriched in CA1 dendrites, were microdissected and homogenized
22
in an ice-cold Tris–HCl buffer (20 mM, pH 7.4) containing 0.32 M sucrose, 1 mM
23
EDTA, 1 mM EGTA, 1 mM PMSF, a Complete Protease Inhibitor Cocktail (1
24
tablet/50 ml) and a PhosSTOP Phosphatase Inhibitor Cocktail (1 tablet/10 ml).
25
Samples of the homogenates (30 µg of protein) were then subjected to SDS-PAGE
6
ACCEPTED MANUSCRIPT (8%) under reducing conditions. The proteins were transferred to PVDF membranes
2
in a transfer buffer [25 mM Tris–HCl buffer (pH 7.4) containing 192 mM glycine and
3
20% v/v methanol] and further separated at 400 mA for 2 h at 4 °C. The Western
4
blots were then incubated for 1 h with a blocking solution (2% BSA or 5% skim milk),
5
then with rabbit anti-cleaved caspase-3 (Cell Signaling Technology, Beverly, MA,
6
1:1000), rabbit anti-caspase-3 (Cell Signaling Technology, Beverly, MA, 1:1000),
7
rabbit anti-actin (Santa Cruz Biotechnology, Santa Cruz, CA, 1:1000), rabbit anti-
8
pAkt1 (Ser473) (Cell Signaling Technology, Beverly, MA, 1:1000), rabbit anti-Akt1
9
(Santa Cruz Biotechnology, Santa Cruz, CA, 1:1000), rabbit anti-pGSK3β (Ser9)
10
(Cell Signaling Technology, Beverly, MA, 1:1000), rabbit anti-GSK3β (Santa Cruz
11
Biotechnology, Santa Cruz, CA, 1:1000) antibody overnight at 4 °C, washed ten
12
times with Tween20/Tris-buffered saline (TTBS), incubated with a 1:2000 dilution of
13
horseradish peroxidase-conjugated secondary antibodies for 2 h at room
14
temperature, washed ten times with TTBS, and finally developed by enhanced
15
chemiluminescence (Amersham LifeScience, Arlington Heights, IL).
16 17
9. Statistics
EP
TE D
M AN U
SC
RI PT
1
The values are expressed as the means ± S.E.M. The data were analyzed using
19
one-way analysis of variance (ANOVA) followed by Student Newman–Keuls test for
20
multiple comparisons. The data from drug experiments were analyzed using two-way
21
ANOVA followed by Bonferroni test for multiple comparisons. The statistical
22
significance was set at P < 0.05.
AC C
18
23 24
Results
7
ACCEPTED MANUSCRIPT 1
1. Intracerebroventricular (i.c.v.) injection of Aβ causes hippocampal LTP and
2
memory impairment Since synthetic Aβ impair synaptic plasticity in the hippocampus and long-term
4
memory (Balducci et al., 2010), we examined Aβ under different experimental
5
conditions. To evaluate the precise concentration of Aβ that leads to memory
6
impairment, we first conducted inhibitory avoidance and object location recognition
7
tests after i.c.v. injection of various concentrations of Aβ. We found that 10 µM of Aβ
8
significantly impaired inhibitory avoidance memory (F3,36 = 3.020, P < 0.05, n =
9
10/group, supplementary Fig. S1A) and object location recognition memory (F3,36 =
10
2.961, P < 0.05, n = 10/group, supplementary Fig. S1B). Moreover, hippocampal
11
slices from Aβ (1 or 10 µM)-injected groups showed significantly lower LTP levels
12
compared to those of a vehicle-injected control group [Aβ (0 µM), 193 ± 8; Aβ (0.5
13
µM), 177 ± 12; Aβ (1 µM), 147 ± 10; Aβ (10 µM), 115 ± 12; F3,24 = 10.52, P < 0.05, n
14
= 7/group, supplementary Fig. S1C].
TE D
M AN U
SC
RI PT
3
Synthetic Aβ transiently impaired hippocampus-dependent memory (Balducci et
16
al., 2010). Therefore, we next examined the time course of the effect of Aβ on
17
memory and synaptic plasticity. Behavioral experiments were started at 1, 3, or 7
18
days after Aβ injection. The memory-impairing effect of Aβ persisted until 3 days
19
after injection in the inhibitory avoidance test (day 1, t18 = 2.922, P < 0.05, n =
20
10/group; day 3, t18 = 1.829, P < 0.05; day 7, t18 = 0.5026, P > 0.05, n = 10/group,
21
supplementary Fig. S1D) and in the object location recognition test (day 1, t18 =
22
2.100, P < 0.05; day 3, t18 = 2.618, P < 0.05; day 7, t18 = 0.6681, P > 0.05, n =
23
10/group, supplementary Fig. S1E). Moreover, hippocampal LTP levels were also
24
found to be impaired in the slices taken at 1, 3, or 7 days after Aβ injection (Day 1:
25
control = 166 ± 12, Aβ = 114 ± 10, t12 = 3.159, P < 0.05, n = 7/group, supplementary
AC C
EP
15
8
ACCEPTED MANUSCRIPT Fig. S1F; Day 3: control = 187 ± 11, Aβ = 135 ± 16, t12 = 2.557, P < 0.05, n = 7/group,
2
supplementary Fig. S1G; Day 7: control = 180 ± 17, Aβ = 146 ± 5, t12 = 1.889, P <
3
0.05, n = 7/group, supplementary Fig. S1H). Based on these results, we conducted
4
all other experiments at 3 days after Aβ (10 µM) injection.
RI PT
1
5 6
2. Aβ activates the CAG pathway
A previous report suggested that the caspase-3/Akt/GSK-3β cascade is important
8
in Aβ-impairing synaptic plasticity (Jo et al., 2011). However, these authors
9
conducted experiments with perfusion of Aβ into the recording chamber using
10
hippocampal slices. Here, we examined whether this system is active in vivo as well
11
as in vitro. To test this, we injected Aβ into the 3rd ventricle and first measured the
12
changes in the caspase-3/Akt/GSK-3β cascade in the radiatum of the hippocampal
13
CA1 region, which is mainly consists of dendrites of pyramidal neurons. Aβ injection
14
lead to significantly higher cleaved caspase-3 levels (t6 = 2.143, P < 0.05, n =
15
5/group, Fig. 1B and 1C). However, levels of the phosphorylated form of Akt1 and
16
GSK-3β were significantly lower in Aβ-injected groups (pAkt1: t6 = 1.999, P < 0.05, n
17
= 5/group; pGSK-3β: t6 = 1.994, P < 0.05, n = 5/group, Fig. 1B and 1C), while levels
18
of the Akt1 and GSK-3β did not change significantly. These results suggest that Aβ
19
inhibits Akt1 and activates caspase-3 and GSK-3β.
21
M AN U
TE D
EP
AC C
20
SC
7
3. Caspase-3 inhibition rescues Aβ-induced memory impairment
22
A previous report indicated that caspase-3 knockout mice showed normal LTP in
23
the presence of Aβ in an in vitro system (Jo et al., 2011). Moreover, pharmacological
24
inhibition of caspase-3 rescued contextual fear memory deficits and synaptic
25
dysfunctions in the Tg2576-APPswe mouse model of Alzheimer's disease (D'Amelio
9
ACCEPTED MANUSCRIPT et al., 2011). We, therefore, tested the effect of inhibition of caspase-3 in avoidance
2
memory, working memory, recognition memory, and long-term synaptic plasticity in
3
the hippocampus. To inhibit caspase-3, we used z-DEVD-fmk, a cell-permeable and
4
irreversible caspase-3/CPP32 inhibitor (IC50 = 18 µM). All experiments were
5
conducted 3 days after Aβ and/or z-DEVD-fmk (30 µM/3 µl) injection into the 3rd
6
ventricle. In the inhibitory avoidance test, all groups showed similar latency to enter
7
in the training session (two way ANOVA, z-DEVD-fmk, F1, 35 = 0.638, P = 0.430; Aβ,
8
F1, 35 = 0.035, P = 0.853; interaction, F1, 35 = 0.299, P = 0.587, n = 9 – 10/group, Fig.
9
2B). In the test session, Aβ-treated mice showed significantly shorter latency to enter
10
than vehicle-treated control mice. However, Aβ- and z-DEVD-fmk-co-treated mice
11
showed latency to enter similar to those found for z-DEVD-fmk-treated mice (two
12
way ANOVA, z-DEVD-fmk, F1, 35 = 6.209, P = 0.018; Aβ, F1, 35 = 4.778, P = 0.036;
13
interaction, F1, 35 = 4.808, P = 0.035, n = 9 – 10/group, Fig. 2A). In the object location
14
recognition test, total exploration time was not significantly different between groups
15
(two way ANOVA, z-DEVD-fmk, F1, 35 = 0.012, P = 0.913; Aβ, F1, 35 = 0.072, P =
16
0.789; interaction, F1, 35 = 0.412, P = 0.525, n = 9 – 10/group, Fig. 2D). However, Aβ-
17
treated mice could not distinguish moved object and fixed object (t18 = 1.167, P >
18
0.05, n = 10, Fig. 2C), while Aβ and z-DEVD-fmk-cotreated mice could distinguish
19
the objects (t18 = 1.846, P < 0.05, n = 10, Fig. 2C). Moreover, Aβ-treated mice
20
showed a significantly lower number of spontaneous alternations, while Aβ and z-
21
DEVD-fmk-cotreated mice showed a significantly higher spontaneous alternations
22
(two way ANOVA, z-DEVD-fmk, F1, 35 = 5.499, P = 0.025; Aβ, F1, 35 = 4.409, P =
23
0.043; interaction, F1, 35 = 4.589, P = 0.039, n = 9-10, Fig. 2E) in the Y-maze test
24
without total arm entries being affected (two way ANOVA, z-DEVD-fmk, F1, 35 = 0.118,
25
P = 0.734; Aβ, F1, 35 = 0.498, P = 0.485; interaction, F1, 35 = 0.003, P = 0.959, n = 9 –
AC C
EP
TE D
M AN U
SC
RI PT
1
10
ACCEPTED MANUSCRIPT 1
10/group, Fig. 2F). These results suggest that caspase-3 inhibition rescued spatial
2
fear memory and object location recognition memory deficits induced by Aβ. As reported above Aβ impaired LTP and facilitated LTD in the hippocampal
4
Schaffer collateral pathway. However, treatment of z-DEVD-fmk with Aβ attenuated
5
this effect of Aβ on LTP and LTD (LTP: two way ANOVA, z-DEVD-fmk, F1, 24 = 3.520,
6
P = 0.073; Aβ, F1, 24 = 4.856, P = 0.037; interaction, F1, 24 = 4.500, P = 0.044, n =
7
7/group, Fig. 2G; LTD: two way ANOVA, z-DEVD-fmk, F1, 24 = 2.529, P = 0.125; Aβ,
8
F1, 24 = 21.58, P = 0.001; interaction, F1, 24 = 15.68, P = 0.006, n = 7/group, Fig. 2H).
SC
RI PT
3
10
M AN U
9
4. GSK-3β inhibition rescues Aβ-induced synaptic and memory dysfunction A previous report indicated that GSK-3β inhibitor rescues Aβ-induced LTP in an in
12
vitro system (Jo et al., 2011). Therefore, we tested the effect of pharmacological
13
inhibition of GSK-3β on avoidance memory, working memory, recognition memory
14
and long-term synaptic plasticity in the hippocampus. To inhibit GSK-3β activity, we
15
used a potent inhibitor of GSK-3β (IC50 = 35 nM). In the inhibitory avoidance test, all
16
groups showed similar latency to enter in the training session (two way ANOVA,
17
TSC2002, F1, 36 = 1.087, P = 0.3.4; Aβ, F1, 36 = 0.716, P = 0.403; interaction, F1, 36 =
18
0.422, P = 0.520, n = 10/group, Fig. 3B). In the test session, Aβ-treated mice
19
showed significantly shorter latency to enter than vehicle-treated control mice.
20
However, Aβ and TCS2002 (10 µM/3 µl)-cotreated mice showed latency to enter
21
similar to those found for TCS2002-treated mice (two way ANOVA, TSC2002, F1, 36 =
22
2.840, P = 0.101; Aβ, F1, 36 = 6.055, P = 0.019; interaction, F1, 36 = 11.07, P = 0.002,
23
n = 10/group, Fig. 3A). In the object location recognition test, total exploration time
24
was not significantly different between groups (two way ANOVA, TSC2002, F1, 36 =
25
0.389, P = 0.537; Aβ, F1, 36 = 0.373, P = 0.545; interaction, F1, 36 = 0.012, P = 0.913,
AC C
EP
TE D
11
11
ACCEPTED MANUSCRIPT n = 10/group, Fig. 3D). However, Aβ-treated mice could not distinguish moved object
2
and fixed object (t18 = 0.4830, P > 0.05, n = 10, Fig. 3C), in the test session. Aβ and
3
TCS2002-cotreated mice could distinguish those objects (t18 = 1.947, P < 0.05, n =
4
10, Fig. 3C). Moreover, Aβ-treated mice showed a significantly lower number of
5
spontaneous alternations, while Aβ and z-DEVD-fmk-cotreated mice shower
6
significantly higher spontaneous alternations (two way ANOVA, TSC2002, F1,
7
3.733, P = 0.061; Aβ, F1, 36 = 3.877, P = 0.056; interaction, F1, 36 = 4.246, P = 0.047,
8
n = 10, Fig. 3E) in the Y-maze test, without total arm entries being affected (two way
9
ANOVA, TSC2002, F1, 36 = 0.582, P = 0.451; Aβ, F1, 36 = 0.009, P = 0.921; interaction,
36
=
M AN U
SC
RI PT
1
F1,
11
pharmacological inhibition of GSK-3β rescued spatial fear memory and object
12
location recognition memory deficits induced by Aβ. Treatment of TCS2002 with Aβ
13
also attenuated the effect of Aβ on LTP and LTD (LTP: two way ANOVA, TSC2002,
14
F1, 24 = 2.234, P = 0.148; Aβ, F1, 24 = 2.006, P = 0.169; interaction, F1, 24 = 5.032, P =
15
0.034, n = 7/group, Fig. 3G; LTD: two way ANOVA, TSC2002, F1, 324 = 5.642, P =
16
0.026; Aβ, F1,
17
10/group, Fig. 3H).
19
= 0.034, P = 0.854, n = 10/group, Fig. 3F). These results suggest that
= 1.520, P = 0.229; interaction, F1,
24
= 4.271, P = 0.049, n =
EP
24
AC C
18
36
TE D
10
5. Akt activation restores Aβ-induced synaptic and memory dysfunction
20
A previous report indicated that neurons injected with an Akt1 mutant mutated at
21
the caspase-3-cleavaging site show normal LTP in the presence of Aβ in an in vitro
22
system (Jo et al., 2011). Therefore, we tested the effect of pharmacological
23
activation of Akt in avoidance memory, working memory, recognition memory, and
24
long-term synaptic plasticity in the hippocampus. To activate Akt, we used SC79, an
25
activator of Akt; binds to the pleckstrin homology domain of Akt, which enhances Akt
12
ACCEPTED MANUSCRIPT phosphorylation by upstream protein kinases; also enables cytosolic activation of Akt.
2
In the inhibitory avoidance test, all groups showed similar latency to enter in the
3
training session (two way ANOVA, SC79, F1, 35 = 0.425, P = 0.519; Aβ, F1, 35 = 0.095,
4
P = 0.760; interaction, F1, 35 = 0.569, P = 0.456, n = 9 - 10/group, Fig. 4B). In the test
5
session, Aβ-treated mice showed significantly shorter latency to enter than vehicle-
6
treated control mice. However, Aβ and SC79 (100 µg/ 3 µl)-cotreated mice showed
7
latency to enter similar to those found for SC79-treated mice (two way ANOVA,
8
SC79, F1, 35 = 10.52, P = 0.003; Aβ, F1, 35 = 4.293, P = 0.046; interaction, F1, 35 =
9
4.153, P = 0.049, n = 9 - 10/group, Fig. 4A). In the object location recognition test,
10
total exploration time was not significantly different between groups (two way
11
ANOVA, SC79, F1, 35 = 0.104, P = 0.749; Aβ, F1, 35 = 0.479, P = 0.494; interaction, F1,
12
35
13
distinguish moved object and fix objects in the test session (t18 = 0.5181, P > 0.05, n
14
= 10, Fig. 4C), while Aβ and SC79-cotreated mice could distinguish those objects (t18
15
= 1.903, P < 0.05, n = 10, Fig. 4C). Moreover, Aβ-treated mice showed a significantly
16
lower number of spontaneous alternations and SC79 improved this (two way ANOVA,
17
SC79, F1, 35 = 3.263, P = 0.067; Aβ, F1, 35 = 4.133, P = 0.049; interaction, F1, 35 =
18
4.722, P = 0.037, n = 9 - 10, Fig. 4E) in the Y-maze test, without total arm entries
19
being affected (two way ANOVA, SC79, F1, 35 = 0.105, P = 0.748; Aβ, F1, 35 = 0.001,
20
P = 0.979; interaction, F1, 35 = 0.180, P = 0.674, n = 9 - 10/group, Fig. 4F). These
21
results suggest that direct activation of Akt rescued spatial fear memory and object
22
location recognition memory deficits induced by Aβ. Treatment of SC79 with Aβ also
23
attenuated the effect of Aβ on LTP (two way ANOVA, SC79, F1, 24 = 2.191, P = 0.152;
24
Aβ, F1, 24 = 1.984, P = 0.172; interaction, F1, 24 = 5.252, P = 0.031, n = 7/group, Fig.
M AN U
SC
RI PT
1
AC C
EP
TE D
= 0.014, P = 0.906, n = 9 - 10/group, Fig. 4D). However, Aβ-treated mice could not
13
ACCEPTED MANUSCRIPT 1
4G) and LTD (two way ANOVA, SC79, F1, 24 = 3.844, P = 0.062; Aβ, F1, 24 = 4.170, P
2
= 0.052; interaction, F1, 24 = 4.659, P = 0.041, n = 7/group, Fig. 4H).
3 6. Akt activation ameliorates synaptic and memory dysfunction in 5XFAD mice
RI PT
4
Since Injection of synthetic Aβ into the ventricle causes reversible deficits in
6
synaptic plasticity, learning, and memory, we next tested if direct Akt activation could
7
rescue memory and synaptic plasticity deficits in 6-month-old 5XFAD AD model mice.
8
SC79 was infused into the 3rd ventricle through cannulae once a day for 2 weeks
9
(Fig. 5A). Then, we conducted inhibitory avoidance and object location recognition
10
tests, and an LTP experiment. In the inhibitory avoidance test, all groups showed
11
similar latency to enter in the training trial (two way ANOVA, SC79, F1, 36 = 0.742, P
12
= 0.395; Tg, F1, 36 = 0.150, P = 0.701; interaction, F1,
13
10/group, Fig. 5B). However, vehicle-treated 5XFAD group showed significantly
14
shorter latency to enter than wild type group and this effect was ameliorated by
15
SC79 infusion (two way ANOVA, SC79, F1, 36 = 5.633, P = 0.023; Tg, F1, 36 = 4.587,
16
P = 0.039; interaction, F1, 36 = 4.138, P = 0.049, n = 10/group, Fig. 5C). Results from
17
the object location recognition test were similar to the data from the inhibitory
18
avoidance test. Vehicle-treated 5XFAD group showed a significantly lower
19
discrimination index than wild type group and this effect was ameliorated by SC79
20
infusion (two way ANOVA, SC79, F1, 36 = 1.832, P = 0.188; Tg, F1, 36 = 5.089, P =
21
0.033; interaction, F1,
22
exploration time being affected (two way ANOVA, SC79, F1, 36 = 0.988, P = 0.327; Tg,
23
F1, 36 = 0.469, P = 0.498; interaction, F1, 36 = 0.008, P = 0.927,n = 10/group, Fig. 5D).
24
Treatment with SC79 also improved the lower LTP level, which was shown in the
25
vehicle-treated 5XFAD group (two way ANOVA, SC79, F1, 28 = 2.921, P = 0.099; Tg,
M AN U
SC
5
= 0.007, P = 0.934, n =
AC C
EP
TE D
36
36
= 4.434, P = 0.046, n = 10/group, Fig. 5E), without total
14
ACCEPTED MANUSCRIPT 1
F1, 28 = 14.29, P = 0.001; interaction, F1, 28 = 6.681, P = 0.015, n = 7 - 9/group, Fig.
2
5F and 5G). Activity of GSK-3β, a direct target of Akt, was also increased in both wild type and
4
5XFAD groups after SC79 administration (two way ANOVA, SC79, F1, 16 = 11.93, P =
5
0.003; Tg, F1, 16 = 0.602, P = 0.449; interaction, F1, 16 = 6.007, P = 0.026, n = 5/group,
6
Fig. 5H and 5I). Akt1 activity was increased in 5XFAD group after SC79
7
administration (two way ANOVA, SC79, SC79, F1, 16 = 13.38, P = 0.002; Tg, F1, 16 =
8
3.201, P = 0.092; interaction, F1, 16 = 21.60, P = 0.003, n = 5/group, Fig. 5H and 5J).
SC
RI PT
3
10
M AN U
9 Discussion
Aberrant regulation of Akt is one of the key events in the AD brain. Since Akt is
12
involved in various up-stream signaling pathways including mTORC2 and PDK1, and
13
in down-stream pathways including GSK-3β, CREB, and Tau, all known to be
14
aberrantly regulated in AD pathology, the regulation of Akt has been identified as a
15
candidate for AD treatment. However, the effect of direct regulation of Akt has not
16
been investigated before. In the present study, we found reduced Akt activity in Aβ-
17
injected or 5XFAD mouse AD models. Therefore, we tested the effect of direct
18
activation of Akt with SC79, an Akt activator, on AD-like memory impairments and
19
aberrant synaptic plasticity. We found that short-term treatment with SC79
20
ameliorated memory and LTP impairments and attenuated facilitated LTD in Aβ-
21
injected as well as 5XFAD AD model mice.
AC C
EP
TE D
11
22
The Inhibitory avoidance task is notorious for its sensitivity to confounding factors.
23
At least hyperactivity, reduced anxiety and increased pain threshold can yield a false
24
impression of impaired memory. Total arm visits in the spontaneous alternation in Y-
25
maze can be used as a control for activity. In the present study, because total arm
15
ACCEPTED MANUSCRIPT visits were not significantly different, the effect of Aβ infusion and the applied drugs
2
on animal activities could be rule out. We also could infer anxiety level from time
3
spent in exploring the object on the first day of object location recognition task.
4
Because this was not significantly different among groups, the effect of Aβ infusion
5
and the applied drugs on anxiety level could be rule out. To test the effect of Aβ
6
infusion and the applied drugs on sensitivity to foot shock, we measured responses
7
to foot shock in training trial (no response, 0; immobilization, 1; vocalization, 2;
8
jumping 3) (supplemental Fig. 4). In this test, all groups showed similar scores in
9
responses to food shock. Therefore, we demonstrate that the data from inhibitory
SC
M AN U
10
RI PT
1
avoidance task used in this study could be acceptable.
Caspase-3 is a molecule which is linked to Akt (Chu et al., 2017; Jahani-Asl et al.,
12
2007; Li et al., 2010). Caspase-3 activation is a cause of neuronal death in AD
13
pathology (Bredesen, 2009; D'Amelio et al., 2011). Growth factors and neurotropic
14
factors protect neurons from Aβ toxicities via inhibition of caspase-3 through Akt
15
activation (Schindowski et al., 2008; Zheng et al., 2000). However, since neuronal
16
loss is a late event in AD patients, its significance has been under-estimated. Recent
17
reports show that caspase-3 involved in early synaptic dysfunction induced by Aβ
18
and blocking caspase-3 is effective in preventing AD-like symptoms (D'Amelio et al.,
19
2011; Jo et al., 2011). Moreover, caspase-3 is involved in physiological synaptic
20
depression, which is related to brain development and memory flexibility (Li et al.,
21
2010). Recent studies also found that caspase-3 exists in the synapse as well as the
22
cell body, suggesting that local caspase-3 activation can regulate local physiological
23
and pathological events without affecting cell fate. The Aβ-injected AD model mice
24
that we used showed transient memory impairments and aberrant synaptic plasticity
25
in the hippocampus without evident neuronal loss suggesting that this model can be
AC C
EP
TE D
11
16
ACCEPTED MANUSCRIPT used to mimic early AD patients. In the present study, inhibition of caspase-3
2
blocked memory impairments and aberrant synaptic plasticity. These findings
3
suggest that if good pharmacological inhibitors are good drug candidates for early
4
phase of AD patients. However, since caspase-3 also has various physiological
5
roles, further research is required.
RI PT
1
GSK-3β is a well-studied target in AD pathology and a down-stream molecule of
7
Akt (Hooper et al., 2008; Reddy, 2013). GSK-3β is involved in Aβ-induced synaptic
8
pathology and memory impairments (Bradley et al., 2012; Peineau et al., 2008).
9
Moreover, GSK-3β inhibitors show neuroprotection and regulation of symptoms in
10
various AD models (Dominguez et al., 2012; Martinez et al., 2011). Lithium is a
11
representative GSK-3β inhibitor. Unfortunately, a clinical study failed to show the
12
effectiveness of lithium on moderate AD pathology (Hampel et al., 2009). However,
13
Nunes et al. (2013) showed a delay of progression of mild cognitive impairments to
14
AD suggesting that GSK-3β inhibition may be effective before onset or in the very
15
early phase of AD (Nunes et al., 2013). This may due to the fact that the time course
16
of AD onset is similar Tau hyperphosphorylation, rather than to Aβ deposition
17
(Selkoe and Hardy, 2016). Since GSK-3β is a Tau kinase, once Tau is
18
phosphorylated, GSK-3β inhibition cannot restore this event. In the present study, we
19
first examined the effect of TCS2002, a cell permeable GSK-3β inhibitor, in an Aβ-
20
injected AD mouse model. TCS2002 ameliorated memory impairments and aberrant
21
synaptic plasticity. These results suggest that TCS2002 may be a good candidate for
22
preventing the progression of MCI to AD.
AC C
EP
TE D
M AN U
SC
6
23
Akt is both a regulator and a substrate of caspase-3. Akt not only suppresses
24
caspase-3 activation but is also cleaved by activated caspase-3 (Chu et al., 2017).
17
ACCEPTED MANUSCRIPT This suggests that Aβ can suppress activity and reduce the protein level of Akt. In
2
the present study, we found that Akt phosphorylation was significantly lower in Aβ-
3
injected AD mouse model compared to a vehicle-injected control in the dendrite
4
area. However, levels of the Akt were slightly lower in AD group; the effect was
5
however not significant in the dendrite area. This suggests that changes of activity
6
rather than protein levels of Akt are evident in the early phase of AD. This was
7
confirmed in the 5XFAD model (supplemental Fig. S2A). Early-phase (6-month-old)
8
AD brains showed a reduction of pAkt but normal Akt levels (supplemental Fig. S2A).
9
However, late-phase (1-year-old) AD brains showed a reduction of Akt levels
10
(supplemental Fig. S2B). These results suggest that the Akt activator could be more
11
useful in the early rather than the late phase of AD. In the present study, we
12
confirmed that SC79 ameliorates memory impairments and aberrant synaptic deficits
13
in 3-month-old 5XFAD AD model mice but not in 1-year-old 5XFAD AD model mice
14
(supplemental Fig S3). Although we cannot rule out that prolonged administration or
15
an increase of the dose of SC79 may be effective in late-phase AD as well, it is
16
obvious that treatment protocols and agents should be different for early- and late-
17
phase AD.
EP
TE D
M AN U
SC
RI PT
1
Akt activity is aberrantly regulated in AD brain (Lee et al., 2009; Sajan et al., 2016;
19
Talbot et al., 2012). Many researches showed controversial results in Akt activity in
20
brain of AD patients. Several findings showed increase of Akt activity in AD patients,
21
but others and we found opposite results in Aβ-treated neurons or mouse AD
22
models. In AD brain, there might be various pathological initiators beside Aβ. But, in
23
the Aβ-treated AD model system, Aβ is only involved in AD-like pathology. This might
24
be the reason of the controversy. Although Aβ-treated AD model system has long
25
been used to develop AD treatment, promising medicine for AD is still not developed.
AC C
18
18
ACCEPTED MANUSCRIPT 1
The controversial results may reflect the difficulty of developing AD treatment with
2
this system.
3 Funding and Disclosure
RI PT
4
This study was supported by the National Research Foundation of Korea (NRF)
6
grand funded by the Ministry of Education, Science and Technology (MEST)
7
(2016R1A5A2007009 and 2015R1A2A2A01007838).
8
SC
5
The authors declare no competing financial interests.
AC C
EP
TE D
M AN U
9
19
ACCEPTED MANUSCRIPT References
2
Abbott, J. J., Howlett, D. R., Francis, P. T., Williams, R. J., 2008. Abeta(1-42)
3
modulation of Akt phosphorylation via alpha7 nAChR and NMDA receptors.
4
Neurobiol Aging 29, 992-1001.
5
Ahmad, A., Ali, T., Park, H. Y., Badshah, H., Rehman, S. U., Kim, M. O., 2016.
6
Neuroprotective Effect of Fisetin Against Amyloid-Beta-Induced Cognitive/Synaptic
7
Dysfunction, Neuroinflammation, and Neurodegeneration in Adult Mice. Mol
8
Neurobiol.
9
Ali, T., Kim, M. O., 2015. Melatonin ameliorates amyloid beta-induced memory
10
deficits, tau hyperphosphorylation and neurodegeneration via PI3/Akt/GSk3beta
11
pathway in the mouse hippocampus. J Pineal Res 59, 47-59.
12
Baki, L., Shioi, J., Wen, P., Shao, Z., Schwarzman, A., Gama-Sosa, M., Neve, R.,
13
Robakis, N. K., 2004. PS1 activates PI3K thus inhibiting GSK-3 activity and tau
14
overphosphorylation: effects of FAD mutations. EMBO J 23, 2586-2596.
15
Balducci, C., Beeg, M., Stravalaci, M., Bastone, A., Sclip, A., Biasini, E., Tapella, L.,
16
Colombo, L., Manzoni, C., Borsello, T., Chiesa, R., Gobbi, M., Salmona, M., Forloni,
17
G., 2010. Synthetic amyloid-beta oligomers impair long-term memory independently
18
of cellular prion protein. Proc Natl Acad Sci U S A 107, 2295-2300.
19
Bradley, C. A., Peineau, S., Taghibiglou, C., Nicolas, C. S., Whitcomb, D. J.,
20
Bortolotto, Z. A., Kaang, B. K., Cho, K., Wang, Y. T., Collingridge, G. L., 2012. A
21
pivotal role of GSK-3 in synaptic plasticity. Front Mol Neurosci 5, 13.
22
Bredesen, D. E., 2009. Neurodegeneration in Alzheimer's disease: caspases and
23
synaptic element interdependence. Mol Neurodegener 4, 27.
AC C
EP
TE D
M AN U
SC
RI PT
1
20
ACCEPTED MANUSCRIPT Butterfield, D. A., 2002. Amyloid beta-peptide (1-42)-induced oxidative stress and
2
neurotoxicity: implications for neurodegeneration in Alzheimer's disease brain. A
3
review. Free Radic Res 36, 1307-1313.
4
Chen, T. J., Wang, D. C., Chen, S. S., 2009. Amyloid-beta interrupts the PI3K-Akt-
5
mTOR signaling pathway that could be involved in brain-derived neurotrophic factor-
6
induced Arc expression in rat cortical neurons. J Neurosci Res 87, 2297-2307.
7
Chu, J., Lauretti, E., Pratico, D., 2017. Caspase-3-dependent cleavage of Akt
8
modulates tau phosphorylation via GSK3beta kinase: implications for Alzheimer's
9
disease. Mol Psychiatry.
M AN U
SC
RI PT
1
D'Amelio, M., Cavallucci, V., Middei, S., Marchetti, C., Pacioni, S., Ferri, A.,
11
Diamantini, A., De Zio, D., Carrara, P., Battistini, L., Moreno, S., Bacci, A.,
12
Ammassari-Teule, M., Marie, H., Cecconi, F., 2011. Caspase-3 triggers early
13
synaptic dysfunction in a mouse model of Alzheimer's disease. Nat Neurosci 14, 69-
14
76.
15
Dominguez, J. M., Fuertes, A., Orozco, L., del Monte-Millan, M., Delgado, E.,
16
Medina, M., 2012. Evidence for irreversible inhibition of glycogen synthase kinase-
17
3beta by tideglusib. J Biol Chem 287, 893-904.
18
Fan, W., Dai, Y., Xu, H., Zhu, X., Cai, P., Wang, L., Sun, C., Hu, C., Zheng, P., Zhao,
19
B. Q., 2014. Caspase-3 modulates regenerative response after stroke. Stem Cells
20
32, 473-486.
21
Franke, T. F., 2008. PI3K/Akt: getting it right matters. Oncogene 27, 6473-6488.
22
Griffin, R. J., Moloney, A., Kelliher, M., Johnston, J. A., Ravid, R., Dockery, P.,
23
O'Connor, R., O'Neill, C., 2005. Activation of Akt/PKB, increased phosphorylation of
24
Akt substrates and loss and altered distribution of Akt and PTEN are features of
25
Alzheimer's disease pathology. J Neurochem 93, 105-117.
AC C
EP
TE D
10
21
ACCEPTED MANUSCRIPT Haass, C., Selkoe, D. J., 2007. Soluble protein oligomers in neurodegeneration:
2
lessons from the Alzheimer's amyloid beta-peptide. Nat Rev Mol Cell Biol 8, 101-
3
112.
4
Hampel, H., Ewers, M., Burger, K., Annas, P., Mortberg, A., Bogstedt, A., Frolich, L.,
5
Schroder, J., Schonknecht, P., Riepe, M. W., Kraft, I., Gasser, T., Leyhe, T., Moller,
6
H. J., Kurz, A., Basun, H., 2009. Lithium trial in Alzheimer's disease: a randomized,
7
single-blind, placebo-controlled, multicenter 10-week study. J Clin Psychiatry 70,
8
922-931.
9
Hooper, C., Killick, R., Lovestone, S., 2008. The GSK3 hypothesis of Alzheimer's
M AN U
SC
RI PT
1
disease. J Neurochem 104, 1433-1439.
11
Horwood, J. M., Dufour, F., Laroche, S., Davis, S., 2006. Signalling mechanisms
12
mediated by the phosphoinositide 3-kinase/Akt cascade in synaptic plasticity and
13
memory in the rat. Eur J Neurosci 23, 3375-3384.
14
Jahani-Asl, A., Basak, A., Tsang, B. K., 2007. Caspase-3-mediated cleavage of Akt:
15
involvement of non-consensus sites and influence of phosphorylation. FEBS Lett
16
581, 2883-2888.
17
Jin, Y., Yan, E. Z., Fan, Y., Zong, Z. H., Qi, Z. M., Li, Z., 2005. Sodium ferulate
18
prevents amyloid-beta-induced neurotoxicity through suppression of p38 MAPK and
19
upregulation of ERK-1/2 and Akt/protein kinase B in rat hippocampus. Acta
20
Pharmacol Sin 26, 943-951.
21
Jo, J., Whitcomb, D. J., Olsen, K. M., Kerrigan, T. L., Lo, S. C., Bru-Mercier, G.,
22
Dickinson, B., Scullion, S., Sheng, M., Collingridge, G., Cho, K., 2011. Abeta(1-42)
23
inhibition of LTP is mediated by a signaling pathway involving caspase-3, Akt1 and
24
GSK-3beta. Nat Neurosci 14, 545-547.
AC C
EP
TE D
10
22
ACCEPTED MANUSCRIPT Kim, D. H., Hung, T. M., Bae, K. H., Jung, J. W., Lee, S., Yoon, B. H., Cheong, J. H.,
2
Ko, K. H., Ryu, J. H., 2006. Gomisin A improves scopolamine-induced memory
3
impairment in mice. Eur J Pharmacol 542, 129-135.
4
Klein, W. L., 2002. Abeta toxicity in Alzheimer's disease: globular oligomers (ADDLs)
5
as new vaccine and drug targets. Neurochem Int 41, 345-352.
6
Klein, W. L., Krafft, G. A., Finch, C. E., 2001. Targeting small Abeta oligomers: the
7
solution to an Alzheimer's disease conundrum? Trends Neurosci 24, 219-224.
8
Kohn, A. D., Summers, S. A., Birnbaum, M. J., Roth, R. A., 1996. Expression of a
9
constitutively active Akt Ser/Thr kinase in 3T3-L1 adipocytes stimulates glucose
M AN U
SC
RI PT
1
uptake and glucose transporter 4 translocation. J Biol Chem 271, 31372-31378.
11
Kotilinek, L. A., Westerman, M. A., Wang, Q., Panizzon, K., Lim, G. P., Simonyi, A.,
12
Lesne, S., Falinska, A., Younkin, L. H., Younkin, S. G., Rowan, M., Cleary, J., Wallis,
13
R. A., Sun, G. Y., Cole, G., Frautschy, S., Anwyl, R., Ashe, K. H., 2008.
14
Cyclooxygenase-2 inhibition improves amyloid-beta-mediated suppression of
15
memory and synaptic plasticity. Brain 131, 651-664.
16
Lee, H. K., Kumar, P., Fu, Q., Rosen, K. M., Querfurth, H. W., 2009. The insulin/Akt
17
signaling pathway is targeted by intracellular beta-amyloid. Mol Biol Cell 20, 1533-
18
1544.
19
Li, Z., Jo, J., Jia, J. M., Lo, S. C., Whitcomb, D. J., Jiao, S., Cho, K., Sheng, M.,
20
2010. Caspase-3 activation via mitochondria is required for long-term depression
21
and AMPA receptor internalization. Cell 141, 859-871.
22
Ma, R., Xiong, N., Huang, C., Tang, Q., Hu, B., Xiang, J., Li, G., 2009. Erythropoietin
23
protects PC12 cells from beta-amyloid(25-35)-induced apoptosis via PI3K/Akt
24
signaling pathway. Neuropharmacology 56, 1027-1034.
AC C
EP
TE D
10
23
ACCEPTED MANUSCRIPT Magrane, J., Rosen, K. M., Smith, R. C., Walsh, K., Gouras, G. K., Querfurth, H. W.,
2
2005. Intraneuronal beta-amyloid expression downregulates the Akt survival
3
pathway and blunts the stress response. J Neurosci 25, 10960-10969.
4
Martinez, A., Gil, C., Perez, D. I., 2011. Glycogen synthase kinase 3 inhibitors in the
5
next horizon for Alzheimer's disease treatment. Int J Alzheimers Dis 2011, 280502.
6
Nunes, M. A., Viel, T. A., Buck, H. S., 2013. Microdose lithium treatment stabilized
7
cognitive impairment in patients with Alzheimer's disease. Curr Alzheimer Res 10,
8
104-107.
9
Palop, J. J., Mucke, L., 2010. Amyloid-beta-induced neuronal dysfunction in
10
Alzheimer's disease: from synapses toward neural networks. Nat Neurosci 13, 812-
11
818.
12
Paxinos, G., Franklin, K. B. J., 2004. The Mouse Brain in Stereotaxic Coordinates.
13
Gulf Professional Publishing.
14
Peineau, S., Bradley, C., Taghibiglou, C., Doherty, A., Bortolotto, Z. A., Wang, Y. T.,
15
Collingridge, G. L., 2008. The role of GSK-3 in synaptic plasticity. Br J Pharmacol
16
153 Suppl 1, S428-437.
17
Reddy, P. H., 2013. Amyloid beta-induced glycogen synthase kinase 3beta
18
phosphorylated VDAC1 in Alzheimer's disease: implications for synaptic dysfunction
19
and neuronal damage. Biochim Biophys Acta 1832, 1913-1921.
20
Rickle, A., Bogdanovic, N., Volkman, I., Winblad, B., Ravid, R., Cowburn, R. F.,
21
2004. Akt activity in Alzheimer's disease and other neurodegenerative disorders.
22
Neuroreport 15, 955-959.
23
Saitoh, M., Kunitomo, J., Kimura, E., Iwashita, H., Uno, Y., Onishi, T., Uchiyama, N.,
24
Kawamoto, T., Tanaka, T., Mol, C. D., Dougan, D. R., Textor, G. P., Snell, G. P.,
25
Takizawa, M., Itoh, F., Kori, M., 2009. 2-{3-[4-(Alkylsulfinyl)phenyl]-1-benzofuran-5-
AC C
EP
TE D
M AN U
SC
RI PT
1
24
ACCEPTED MANUSCRIPT yl}-5-methyl-1,3,4-oxadiazole derivatives as novel inhibitors of glycogen synthase
2
kinase-3beta with good brain permeability. J Med Chem 52, 6270-6286.
3
Sajan, M., Hansen, B., Ivey, R., 3rd, Sajan, J., Ari, C., Song, S., Braun, U., Leitges,
4
M., Farese-Higgs, M., Farese, R. V., 2016. Brain Insulin Signaling Is Increased in
5
Insulin-Resistant States and Decreases in FOXOs and PGC-1alpha and Increases in
6
Abeta1-40/42 and Phospho-Tau May Abet Alzheimer Development. Diabetes 65,
7
1892-1903.
8
Sakoda, H., Gotoh, Y., Katagiri, H., Kurokawa, M., Ono, H., Onishi, Y., Anai, M.,
9
Ogihara, T., Fujishiro, M., Fukushima, Y., Abe, M., Shojima, N., Kikuchi, M., Oka, Y.,
10
Hirai, H., Asano, T., 2003. Differing roles of Akt and serum- and glucocorticoid-
11
regulated kinase in glucose metabolism, DNA synthesis, and oncogenic activity. J
12
Biol Chem 278, 25802-25807.
13
Sarbassov, D. D., Guertin, D. A., Ali, S. M., Sabatini, D. M., 2005. Phosphorylation
14
and regulation of Akt/PKB by the rictor-mTOR complex. Science 307, 1098-1101.
15
Schindowski, K., Belarbi, K., Buee, L., 2008. Neurotrophic factors in Alzheimer's
16
disease: role of axonal transport. Genes Brain Behav 7 Suppl 1, 43-56.
17
Selkoe, D. J., 2008. Soluble oligomers of the amyloid beta-protein impair synaptic
18
plasticity and behavior. Behav Brain Res 192, 106-113.
19
Selkoe, D. J., Hardy, J., 2016. The amyloid hypothesis of Alzheimer's disease at 25
20
years. EMBO Mol Med 8, 595-608.
21
Shankar, G. M., Li, S., Mehta, T. H., Garcia-Munoz, A., Shepardson, N. E., Smith, I.,
22
Brett, F. M., Farrell, M. A., Rowan, M. J., Lemere, C. A., Regan, C. M., Walsh, D. M.,
23
Sabatini, B. L., Selkoe, D. J., 2008. Amyloid-beta protein dimers isolated directly
24
from Alzheimer's brains impair synaptic plasticity and memory. Nat Med 14, 837-842.
AC C
EP
TE D
M AN U
SC
RI PT
1
25
ACCEPTED MANUSCRIPT Song, G., Ouyang, G., Bao, S., 2005. The activation of Akt/PKB signaling pathway
2
and cell survival. J Cell Mol Med 9, 59-71.
3
Talbot, K., Wang, H. Y., Kazi, H., Han, L. Y., Bakshi, K. P., Stucky, A., Fuino, R. L.,
4
Kawaguchi, K. R., Samoyedny, A. J., Wilson, R. S., Arvanitakis, Z., Schneider, J. A.,
5
Wolf, B. A., Bennett, D. A., Trojanowski, J. Q., Arnold, S. E., 2012. Demonstrated
6
brain insulin resistance in Alzheimer's disease patients is associated with IGF-1
7
resistance, IRS-1 dysregulation, and cognitive decline. J Clin Invest 122, 1316-1338.
8
Tuppo, E. E., Arias, H. R., 2005. The role of inflammation in Alzheimer's disease. Int
9
J Biochem Cell Biol 37, 289-305.
M AN U
SC
RI PT
1
Vivanco, I., Sawyers, C. L., 2002. The phosphatidylinositol 3-Kinase AKT pathway in
11
human cancer. Nat Rev Cancer 2, 489-501.
12
Wang, Y. J., Ren, Q. G., Gong, W. G., Wu, D., Tang, X., Li, X. L., Wu, F. F., Bai, F.,
13
Xu, L., Zhang, Z. J., 2016a. Escitalopram attenuates beta-amyloid-induced tau
14
hyperphosphorylation in primary hippocampal neurons through the 5-HT1A receptor
15
mediated Akt/GSK-3beta pathway. Oncotarget 7, 13328-13339.
16
Wang, Z. X., Tan, L., Liu, J., Yu, J. T., 2016b. The Essential Role of Soluble Abeta
17
Oligomers in Alzheimer's Disease. Mol Neurobiol 53, 1905-1924.
18
Xing, G., Dong, M., Li, X., Zou, Y., Fan, L., Wang, X., Cai, D., Li, C., Zhou, L., Liu, J.,
19
Niu, Y., 2011. Neuroprotective effects of puerarin against beta-amyloid-induced
20
neurotoxicity in PC12 cells via a PI3K-dependent signaling pathway. Brain Res Bull
21
85, 212-218.
22
Yi, J. H., Park, H. J., Lee, S., Jung, J. W., Kim, B. C., Lee, Y. C., Ryu, J. H., Kim, D.
23
H., 2016. Cassia obtusifolia seed ameliorates amyloid beta-induced synaptic
24
dysfunction
25
Ethnopharmacol 178, 50-57.
AC C
EP
TE D
10
through
anti-inflammatory
26
and
Akt/GSK-3beta
pathways.
J
ACCEPTED MANUSCRIPT 1
Zheng, W. H., Kar, S., Dore, S., Quirion, R., 2000. Insulin-like growth factor-1 (IGF-
2
1): a neuroprotective trophic factor acting via the Akt kinase pathway. J Neural
3
Transm Suppl, 261-272.
AC C
EP
TE D
M AN U
SC
RI PT
4
27
ACCEPTED MANUSCRIPT 1
Figure legends
2 Figure 1. Effect of Aβ on signaling molecules in the hippocampus. Aβ (10 µM/3 µl)
4
was injected into third ventricle and hippocampal tissues were collected 3 days after
5
the injection. a. Photograph of blots. b. Quantitative analysis of the expression level
6
of cleaved caspase-3, pAkt and pGSK-3β, which were normalized to actin. c.
7
Quantitative analysis of the expression level of caspase-3, Akt and GSK-3β, which
8
were normalized to actin. Data are expressed as mean ± SEM. *P < 0.05 vs vehicle
9
group.
M AN U
SC
RI PT
3
10
Figure 2. Effect of Z-DEVD-fmk on Aβ-induced memory impairments and aberrant
12
synaptic plasticity. Aβ (10 µM/3 µl) and/or Z-DEVD-fmk (30 µM/3 µl) was injected
13
into third ventricle and hippocampal tissues were collected 3 days after the injection.
14
a. Latency to enter in retention trial of inhibitory avoidance test. b. Latency to enter in
15
acquisition trial of inhibitory avoidance test. c. Preference ratio in test session of
16
object location recognition test. d. Total arm entry in test session of object location
17
recognition test. e. Spontaneous alternations in Y-maze test. f. Total arm entry in Y-
18
maze test. g. Long-term potentiation (LTP) induced by theta burst stimulation (TBS).
19
h. Long-term depression (LTD) induced by low frequency stimulation (1 Hz, 900
20
pulses). Data are expressed as mean ± SEM. *P < 0.05 vs vehicle group.
EP
AC C
21
TE D
11
22
Figure 3. Effect of TCS2002 on Aβ-induced memory impairments and aberrant
23
synaptic plasticity. Aβ (10 µM/3 µl) and/or TCS2002 (10 µM/3 µl) was injected into
24
third ventricle and hippocampal tissues were collected 3 days after the injection. a.
25
Latency to enter in retention trial of inhibitory avoidance test. b. Latency to enter in
28
ACCEPTED MANUSCRIPT acquisition trial of inhibitory avoidance test. c. Preference ratio in test session of
2
object location recognition test. d. Total arm entry in test session of object location
3
recognition test. e. Spontaneous alternations in Y-maze test. f. Total arm entry in Y-
4
maze test. g. Long-term potentiation (LTP) induced by theta burst stimulation (TBS).
5
h. Long-term depression (LTD) induced by low frequency stimulation (1 Hz, 900
6
pulses). Data are expressed as mean ± SEM. *P < 0.05 vs vehicle group.
RI PT
1
SC
7
Figure 4. Effect of SC79 on Aβ-induced memory impairments and aberrant synaptic
9
plasticity. Aβ (10 µM/3 µl) and/or SC79 (100 µg/3 µl) was injected into third ventricle
10
and hippocampal tissues were collected 3 days after the injection. a. Latency to
11
enter in retention trial of inhibitory avoidance test. b. Latency to enter in acquisition
12
trial of inhibitory avoidance test. c. Preference ratio in test session of object
13
recognition test. d. Total arm entry in test session of object recognition test. e.
14
Spontaneous alternations in Y-maze test. f. Total arm entry in Y-maze test. g. Long-
15
term potentiation (LTP) induced by theta burst stimulation (TBS). h. Long-term
16
depression (LTD) induced by low frequency stimulation (1 Hz, 900 pulses). Data are
17
expressed as mean ± SEM. *P < 0.05 vs vehicle group.
TE D
EP
AC C
18
M AN U
8
19
Figure 5. Effect of SC79 on memory impairments and aberrant synaptic plasticity of
20
5XFAD mice. a. Experimental schedule. SC79 (10 µg/3 µl) was once a day for 11
21
days. Inhibitory avoidance test was conducted 8 and 9 d after the start of SC79
22
injection. Object recognition test (ORM) was conducted 10 and 11 d after the start of
23
SC79 injection. b. Latency to enter in acquisition test of inhibitory avoidance test. c.
24
Latency to enter in retention trial of inhibitory avoidance test. d. Total exploration
25
time in test session of object recognition test. e. Discrimination index in test session
29
ACCEPTED MANUSCRIPT of object recognition test. f. Long-term potentiation (LTP) induced by theta burst
2
stimulation (TBS). g. Level of LTP at last 5 min. h. Western blot analysis of Akt1 and
3
GSK-3β activity. I and J. Quantitative analysis of immunoreactivity of western blot of
4
GSK-3β (I) and Akt1 (J). Data are expressed as mean ± SEM. *P < 0.05.
RI PT
1
AC C
EP
TE D
M AN U
SC
5
30
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT Highlight
Oligomeric Aβ impaired learning and memory and synaptic plasticity
Akt activation ameliorated Aβ-induced deficits
RI PT
Caspase inhibition ameliorated Aβ-induced deficits
GSK-3β inhibition ameliorated Aβ-induced deficits
SC
Akt activation ameliorated learning and memory and synaptic deficits in
AC C
EP
TE D
M AN U
5XFAD mice
1