You are on page 1of 17

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/334617845

Avenanthramide-C Restores Impaired Plasticity and Cognition in Alzheimer’s


Disease Model Mice

Article  in  Molecular Neurobiology · January 2020


DOI: 10.1007/s12035-019-01707-5

CITATIONS READS

7 350

12 authors, including:

Vijay Sankar Samidurai Manikandan


Chonnam National University Iowa State University
8 PUBLICATIONS   83 CITATIONS    11 PUBLICATIONS   184 CITATIONS   

SEE PROFILE SEE PROFILE

Hyungseok Kim
Chonnam National University
125 PUBLICATIONS   2,061 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

bacterio-therapy View project

Nanoparticles for inflammatory diseases View project

All content following this page was uploaded by Hyungseok Kim on 17 December 2019.

The user has requested enhancement of the downloaded file.


Molecular Neurobiology
https://doi.org/10.1007/s12035-019-01707-5

Avenanthramide-C Restores Impaired Plasticity and Cognition


in Alzheimer’s Disease Model Mice
Vijay Sankar Ramasamy 1,2 & Manikandan Samidurai 1,2 & Hyung Joon Park 3 & Ming Wang 1,2 & Ra Young Park 4 &
Seon Young Yu 5 & Hee Kyung Kang 1 & Semi Hong 3 & Won-Seok Choi 3 & Yu Young Lee 6 & Hyung-Seok Kim 7,8 &
Jihoon Jo 1,2,9,10

Received: 23 April 2019 / Accepted: 10 July 2019


# Springer Science+Business Media, LLC, part of Springer Nature 2019

Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease characterized by cognitive decline and dementia with no
effective treatment. Here, we investigated a novel compound from oats named avenanthramide-C (Avn-C), on AD-related
memory impairment and behavioral deficits in transgenic mouse models. Acute hippocampal slices of wild-type or AD trans-
genic mice were treated with Avn-C in the presence or absence of oligomeric Aβ42. LTP analyses and immunoblotting were
performed to assess the effect of Avn-C on Aβ-induced memory impairment. To further investigate the effect of Avn-C on
impaired memory and Aβ pathology, two different AD transgenic mice (Tg2576 and 5XFAD) models were orally treated with
either Avn-C or vehicle for 2 weeks. They were then assessed for the effect of the treatment on neuropathologies and behavioral
impairments. Avn-C reversed impaired LTP in both ex vivo- and in vivo-treated AD mice hippocampus. Oral administration
(6 mg/kg per day) for 2 weeks in AD mice leads to improved recognition and spatial memory, reduced caspase-3 cleavage,
reversed neuroinflammation, and to accelerated glycogen synthase kinase-3β (pS9GSK-3β) and interleukin (IL-10) levels. Avn-
C exerts its beneficial effects by binding to α1A adrenergic receptors to stimulate adenosine monophosphate-activated kinase
(AMPK). All of the beneficial effects of Avn-C on LTP retrieval could be blocked by prazosin hydrochloride, a specific inhibitor
of α1A adrenergic receptors. Our findings provide evidence, for the first time, that oats’ Avn-C reverses the AD-related memory

Vijay Sankar Ramasamy, Manikandan Samidurai and Hyung Joon Park


contributed equally to this work.
Electronic supplementary material The online version of this article
(https://doi.org/10.1007/s12035-019-01707-5) contains supplementary
material, which is available to authorized users.

* Won-Seok Choi 4
Bio-Medical treatment technology development projects, Institute for
Biomedical Science, Chonnam National University Hwasun
Hospital, Gwangju, Republic of Korea
* Yu Young Lee
5
Forensic Medicine Division, National Forensic Service Daegu
* Hyung-Seok Kim Institute, Daegu 39872, Republic of Korea
6
Department of Central Area, National Institute of Crop Science,
* Jihoon Jo Rural Development Administration, Suwon 16429, Republic of
Jihoon.Jo@chonnam.ac.kr Korea
7
1
NeuroMedical Convergence Laboratory, Biomedical Research Department of Forensic Medicine, Chonnam National University
Institute, Chonnam National University Hospital, Jebong-ro, Medical School, Gwangju, South Korea
Gwangju 501-757, Republic of Korea 8
Center for Creative Biomedical Scientists, Chonnam National
2
Department of Biomedical Sciences, BK21 PLUS Center for University Medical School, Gwangju 501-757, Republic of Korea
Creative Biomedical Scientists at Chonnam National University, 9
Research Institute of Medical Sciences, Chonnam National Department of Neurology, Chonnam National University Medical
University Medical School, Gwangju 501-757, Republic of Korea School, Gwangju 501-757, Republic of Korea
3 10
School of Biological Sciences and Technology, College of Natural NeuroMedical Convergence Laboratory, Department of Biomedical
Sciences, Chonnam National University, Gwangju 500-757, Sciences and Neurology, Chonnam National University Medical
Republic of Korea School, Gwangju 501-746, Republic of Korea
Mol Neurobiol

and behavioral impairments, and establish it as a potential candidate for Alzheimer’s disease drug development.

Keywords Alzheimer’s disease . Avn-C . Oats . LTP . Amyloid-β . Memory . Mouse model
Introduction avenanthramide-C (Avn-C), a low molecular weight polyphe-
nolic compound exclusively found in oats (Avena sativa), on
Alzheimer’s disease (AD) is the most common form of de- synaptic dysfunction and memory impairment in Tg2576 and
mentia in the elderly population worldwide, characterized by 5XFAD mouse models of AD. Previous studies have shown
the memory and cognition impairment. Though there are sev- that Avn-C have the anti-inflammatory effects of decreasing
eral mechanisms and factors proposed for AD condition, β- pro-inflammatory markers (IL-6 and TNFα) through decreased
amyloid is thought to be the major risk factor, driving oxida- NF-κB activity [22–24]. Rats fed with Avn-C supplemented
tive stress, tau hyperphosphorylation, inflammation, apoptotic diet for 50 days display lower reactive oxygen species genera-
signaling, and neuronal loss [1, 2]. Acetylcholinesterase inhib- tion and lipid peroxidation in skeletal muscle [25]. Also,
itors and an N-methyl-d-aspartate receptor blocker represent 8 weeks of Avn-C supplementation in women abolishes down-
the few currently available drugs approved for AD treatment, hill running-induced inflammation [26]. Furthermore, orally
but these drugs are minimally efficacious and have adverse administered avenanthramides are shown to reduce titanium
effects, limiting their use [3, 4]. Many clinical trials targeting dioxide-induced neuronal degeneration and serum TNFα levels
Aβ with monoclonal antibodies or small peptide inhibitors in Sprague-Dawley rats [27]. Based on these observations, we
have failed because their effects were too limited [5]. hypothesized that the cytoprotective properties of Avn-C would
A critical early event in AD is the synaptic dysfunction and be beneficial for AD-related memory impairment.
impaired synaptic plasticity. Spatial learning and memory im-
pairment, an early clinical feature of AD, is caused by synaptic
dysfunction rather than neuronal loss in AD transgenic mice Materials and Methods
[6]. Soluble oligomeric form of Aβ activates a variety of mo-
lecular cascades that culminate in early synapse dysfunction. Mouse Models
Acute application of oligomeric Aβ42 to hippocampal slices
inhibits long-term potentiation (LTP)—a form of synaptic Wild-type (C57BL/6J, 7–8 months old), Tg2576 (APP
plasticity, by activating caspase-3 and GSK3β [7], reducing KM670/671NL, 7–8 months old, Taconic), and 5XFAD
NMDA receptor surface expression [8] and increasing AMPA (APP KM670/671NL [Swedish], APP I716V [Florida], APP
receptors internalization [9]. In addition, Aβ induce synaptic V717I [London], PSEN1 M146L, PSEN1 L286V, 5–6 months
dysfunction by promoting pro-inflammatory cytokine produc- old, Jackson Laboratory) mice were used for the experiments.
tion. Mouse hippocampal slices exposed to Aβ oligomers In all experiments, male mice were used. Animals were housed
shown to increase the production of tumor necrosis factor-α in individually ventilated cages with free access to food and
(TNFα) and reduction in LTP. In support of this, increased water. The breeding room was controlled with a 12-h light/dark
levels of pro-inflammatory cytokines such as interleukin-1β cycle (lights on at 8:00 p.m.) and the temperature maintained at
(IL1β) [10], interleukin-6 (IL-6) [11], and TNFα [12] have 22–30 °C. All experiments involving animals followed ap-
been observed in brain samples from both AD patients and proved protocols from the Institutional Animal Care and Use
AD mice models. Aberrant microglial activation and cytokine Committee of Chonnam National University Medical School,
production have been correlated with cognitive decline [13, Republic of Korea. In the study, n refers to the number of
14]. Furthermore, in an AD mouse model of tauopathy, animals used in any particular experiment or the number of
microglial activation coincided with hippocampal synapse individual replicates (in vitro).
loss and impaired synaptic function prior to the onset of fibril-
lary tau tangles [15, 16]. Such observations highlight targeting Hippocampal Slice Preparation and Avn-C Treatment
these pathways would be beneficial in AD treatment. In sup-
port of this, administration of nonsteroidal anti-inflammatory Animals were sacrificed between 9:00 and 10:00 a.m. by cer-
drugs ameliorates Aβ pathogenesis and memory impairment vical dislocation. After cervical dislocation, the brain was
[17], although efficacy in clinical trials remains to be shown. quickly removed and transferred to ice-cold artificial cerebro-
In searches for new anti-AD drugs, natural products have spinal fluid (aCSF; 124 mM NaCl, 3 mM KCl, 26 mM
become an important avenue because of their unique composi- NaHCO3, 1.25 mM NaH2PO4, 2 mM CaCl2, 1 mM MgSO4,
tion of therapeutic compounds, for example epigallocatechin and 10 mM glucose). A midsagittal cut was made in the brain
gallate (green tea component), gallic acid, resveratrol, and and one hemisphere was returned to ice-cold aCSF until re-
curcumin were identified as multifunctional compounds with quired. Hippocampal slices were cut transversely (400 μm
anti-AD properties, yet their poor bioavailability limits their thick) using a Mcilwain tissue chopper (Mickle Laboratory
therapeutic effects [18–21]. Here, we report the effects of Engineering Co. Ltd., UK) and stabilized for 1 h in aCSF
Mol Neurobiol

constantly perfused with a gas mixture of 95% O2 and 5% Avenanthramide-C Administration


CO2 at room temperature. For treatment, slices were trans-
ferred to a treatment chamber containing aCSF constantly Avn-C was purchased from Sigma-Aldrich. For treatment,
perfused with a gas mixture of 95% O2 and 5% CO2. To final concentration of 1 mg/mL Avn-C was prepared by dis-
examine the effect of Avn-C on Aβ42-induced LTP impair- solving in 10% Kolliphor (Sigma-Aldrich, USA). Mice
ment, the mouse hippocampal slices were pretreated with de- (5XFAD or Tg2576) were divided into three groups: Tg
sired concentrations of Avn-C for 0.5 h, followed by 0.5 μM Avn-C-treated (n = 10), Tg vehicle-treated (n = 7), and non-
Aβ42 for another 2 h in the same aCSF medium at room transgenic vehicle-treated (n = 9). At 6 months-of-age, Avn-
temperature. In case of transgenic mice experiments, slices C or vehicle was administered orally to each group of mice
were treated with 50 μM Avn-C in the aCSF medium for once a day (2, 4, and 6 mg/kg, at approximately 4:00 p.m.) for
2 h at room temperature. For inhibitor studies with transgenic desired days.
mice hippocampi, slices were pretreated with antagonists for
1 h followed by 50 μM Avn-C exposure in the same aCSF
medium. Morris Water Maze Test

Electrophysiology At 2 weeks after the first Avn-C administration, mice were


subjected to the Morris water maze (5XFAD: n = 8; Avn-C-
Slices were allowed approximately 60 min to recover from the treated 5XFAD: n = 10; WT: n = 8). Mice were placed in a
slice procedure and for stable responses to be obtained. circular pool (114 cm in diameter, 25 cm in height) filled with
Extracellular field potentials were recorded in the CA1 region water that was maintained at 25 °C and made opaque using
using microcapillary electrodes (3–5 MΩ) containing NaCl nontoxic paint. A fixed-position, clear escape platform (17 ×
(3 M). Stimulating electrodes were placed in the subiculum 10.5 cm) was submerged 1.5 cm below the surface of the
and CA3 (Schaffer collateral pathway). Stimuli were delivered water for all training sessions. In each trial, mice were ran-
alternatively to the two electrodes (each electrode 0.016 Hz). domly placed facing the wall in any of the three quadrants of
After establishing a stable baseline for 30 min, LTP was the pool that did not contain the platform. A total of 16 trials
evoked by two trains of tetanus stimuli (each 100 Hz, 1 s: (four trials per day for four consecutive days) were performed.
repeated after a 30-s interval), field excitatory postsynaptic Mice were allowed to swim for 40 s. Mice that did not find the
potentiation (fEPSP) was recorded for at least 60 min. The platform within 40 s were guided to it. The animals were
slope of the evoked field potential response was measured allowed to stay on the platform for 10 s. A probe test was
and expressed relative to the normalized preconditioning performed without the platform in place 24 h after the last
baseline. Data were collected by an NI USB-6251 data acqui- training trial. In the probe test, the mice were allowed to swim
sition module (National Instruments, Texas, USA), amplified for 60 s to search for the removed escape platform.
by an Axopatch 200B amplifier (Axon Instruments, CA,
USA), and captured and analyzed using WinLTP software
(www.winltp.com). CSF Analysis

Amyloid-β Preparation Cerebrospinal fluid (CSF) collection was always performed


between 10 a.m. and 2 p.m. Mice were injected with a mixture
The amyloid-β 1-42 peptide (Aβ42; Abcam, Cambridge, UK) of 10 IU ketamine and Rompun in 3:1 ratio and placed in a
was initially dissolved at a concentration of 1 mg/mL in 100% chamber until they were deeply anesthetized. CSF was then
HFIP (1,1,1,3,3,3-hexafluoro-propanol; Sigma-Aldrich). This immediately collected from cisterna magna with the help of
solution was then incubated at room temperature for 1 h, with magnification glasses. To this end, the overlying skin was
occasional mixing on a vortex at a moderate speed. Next, the incised to expose the skull and the posterior neck muscles.
solution was sonicated for 10 min in a water bath sonicator. The latter were cut off layer by layer until the cisterna magna
The HFIP/peptide solution was then dried under a gentle was visible through the translucent dura matter. After cleaning
stream of nitrogen gas. DMSO (100%) was used to resuspend any blood residue from the surface with a cotton swab, the
the peptide, which was then incubated at room temperature for dura was perforated with a capillary tube and CSF was col-
12 min with occasional mixing using a vortex. The final so- lected. Then the capillary tube was connected with a 3-mL
lution was aliquoted into smaller volumes and stored at − syringe through a polyethylene tubing that has a 1-mm inter-
80 °C. Oligomeric Aβ preparation was done as described nal diameter and the collected CSF was transferred in to a pre-
previously [28]; 500–1000 μL of D-PBS (Invitrogen, UK) marked polypropylene tubes and immediately placed on dry
was added to the peptide stock solution and incubated for ice before stored to − 80 °C freezer for further use. Typically, a
2 h at room temperature to allow for peptide aggregation. total of 15 to 20 μL of CSF was collected.
Mol Neurobiol

Open Field Test explored the objects for 10 min. The percentage of preference
index was calculated as the time exploring the novel object (n)
At 3 weeks after the first Avn-C administration, mice were divided by the time exploring both novel and familiar objects
subjected to behavior tests (5XFAD: n = 8; Avn-C-treated (n + f) [% preference index = n/(n + f) × 100].
5XFAD: n = 10; WT: n = 9). Locomotor activity was mea-
sured in open field test as described [29]. Mice were placed
Immunohistochemistry
in the center of the arena (40 × 40 × 40 cm). Mice were pre-
habituated in arena for 5 min and then freely explored arena
Wild-type (WT) and transgenic mice (5XFAD and Tg2576)
for 20 min. Horizontal movement of mice in arena was re-
treated with vehicle or Avn-C (n = 3) were euthanized after
corded and total distance that mice moved in the arena were
2 weeks of vehicle or drug treatment. The brain was isolated,
measured by ANY-maze (Stoelting, IL, USA).
fixed in 4% paraformaldehyde, and transferred to 30% sucrose
overnight to prevent tissue damage. The tissues were embed-
Rotarod
ded in OCT compound and sliced to a thickness of 20 μm
using a Leica CM1860 tissue processor (Leica Biosystems,
Mice were placed on the stationary cylinder of rotarod
Germany). Brain sections were mounted onto glass slides
(5XFAD: n = 8; Avn-C-treated 5XFAD: n = 10; WT: n = 8)
and permeabilized with 0.3% Triton X-100 for 1 h. After
and the test was performed as described [30]. During pre-
washing with Tris-buffered saline, the sections are incubated
training, mice were habituated on the cylinder (B.S.
in 5% BSA. Primary antibodies for anti IBA1 (1:1000, Wako)
Technolab INC., Seoul, Korea) rotating at a constant speed
and anti-β-amyloid (6E10, 1:500, BioLegend) were applied to
(4 rpm for 60 s). Once the mice were able to stay on the
the samples overnight at 4 °C. After being washed with PBS,
cylinder for 60 s, test was performed 3 h later. After placing
sections were probed with FITC- or TRITC- conjugated sec-
the animal on the cylinder, rotating speed was accelerated
ondary antibodies (1:500) at room temperature for 2 h, follow-
from 4 to 40 rpm over 5 min. Four trials were performed
ed by washing with PBS. Slides were mounted with anti-fade
and minimum 5 min breaks were given between trials. Time
mounting media. Confocal images were obtained using an
to fall from the cylinder was recorded with a cutoff time of
LSM 520 confocal microscope (Carl Zeiss).
300 s. Total eight trials were performed and the data from the
For analysis of the amyloid aggregates, the ImageJ soft-
last trials were averaged.
ware (https://rsbweb.nih.gov/ij/) was used. After adjusting
for threshold, ImageJ was used to measure total area of the
Pole Test
plaques and the percentage of total brain area occupied by
plaques. Data were pooled from 3~5 sections at ×200
Pole test was performed as described [31]. A vertical pole with
magnification of each mouse and three mice were used for
a rough surface (1.5 cm diameter, 43 cm height) was placed in
the statistical analysis. For quantification of Iba-1, 3 coronal
the home cage. The mice were placed facing upward on the
sections per mouse were selected. Optical density was
top of a pole (5XFAD: n = 8; Avn-C-treated 5XFAD: n = 10;
determined with ImageJ software. Interested cells have been
WT: n = 8). The mice were monitored for 60 s or until they
selected, and area integrated intensity and mean gray value
arrive at the bottom. The pole test score was calculated as
have been measured. Finally, the total cell fluorescence
follows: fell from the pole immediately: 1; fell from the pole
calculated. The number of animals in each group was
in 0–10 s: 2, 11–20 s: 3, 21–30 s: 4, 31–40 s: 5, 41–50 s: 6, 51–
indicated in the figure legends.
60 s: 7; stayed on the pole for 60 s and climbed halfway down:
8; climbed to lower half of pole: 9; and climbed down and off
in: 51–60 s: 10, 41–50 s: 11, 31–40 s: 12, 21–30 s: 13, 11– Sandwich Enzyme-Linked Immunosorbent Assay
20 s: 14, 1–10 s: 15.
Quantitative determination of pro-inflammatory cytokines
Novel Object Recognition Test TNFα, IL-6, and IL-10 was performed using the Mouse
(TNF alpha, IL-6, and IL-10) enzyme-linked immunosorbent
Mice were subjected to the novel object recognition test assay kit (eBioscience, San Diego, USA) according to manu-
(5XFAD: n = 8; Avn-C-treated 5XFAD: n = 10; WT: n = 9). facturer’s instructions. Tissue lysate was prepared from the hip-
The test consists of two exploration phase: training and test pocampus using tissue extracting reagent I (Thermo Scientific
phase. During the training phase, mice were placed in the FNN0071). Mouse TNF alpha ELISA Ready-SET-Go!® kit
open-top arena (40 × 40 × 40 cm) for 10 min in the presence (colorimetric; eBioscience; sensitivity 4 pg/mL; range 4–
of two identical objects. In the test phase, mice were returned 500 pg/mL), Mouse IL-6 alpha ELISA Ready-SET-Go!® kit
to the arena with one novel object and the other object which (colorimetric; sensitivity 8 pg/mL; range 8–1000 pg/mL;
is identical to the one used in the training phase and then eBioscience), and Mouse IL-10 alpha ELISA Ready-SET-
Mol Neurobiol

Go!® kit (colorimetric; sensitivity 8 pg/mL; range 8–1000 pg/ significance of the data was analyzed by Student’s t test. A P
mL; eBioscience) were used according to following protocol. value < 0.05 was considered statistically significant.
ELISA plate coated with 100 μL/well of capture antibody
in coating buffer and the sealed plate incubated overnight at Biotinylation and Streptavidin Pull-Down Assays
4 °C. The wells aspirated and washed three times with >
250 μL/well wash buffer. Allowing time for soaking (~ Biotinylation of Avn-C was performed according to the man-
1 min) during each wash step increases the effectiveness of ufacturer’s G-Biosciences (catalog number: BS-16, BS-17)
the washes. The plate was blotted on absorbent paper to re- instructions. For pull-down experiments, slices were treated
move any residual buffer. The wells blocked with 200 μL/well with biotinylated Avn-C (50 μM final concentration) in the
of 1× ELISA/ELISPOT diluent at room temperature for 1 h. presence or absence of prazosin hydrochloride. After treat-
One hundred microliters of top standard concentration was ment, tissue was homogenized in lysis buffer containing
added to the appropriate wells. Twofold serial dilutions of 25 mM Tris (pH 7.6), 150 mM NaCl, 1% NP-40, 1% sodium
the top standards performed to make the standard curve for a deoxycholate, 0.1% SDS, 1 mM NaF, and a cocktail of prote-
total of 8 points. Then 100 μL of samples added to the appro- ase inhibitors (Sigma). The lysate was centrifuged at 11,000×g
priate wells. The sealed plate incubates at room temperature for 15 min at 4 °C to remove nuclei and cellular debris. Total
for 2 h (or overnight at 4 °C for maximal sensitivity). The protein concentration was determined in a BCA assay
wells aspirated and washed three to five times. Detection an- (Pierce). A small amount of the lysate was removed for later
tibody diluted in 1× ELISA/ELISPOT diluent and 100 μL whole-cell analysis. Subsequently, 100 μL of streptavidin
added to each well and allowed to incubate at room tempera- beads (Thermo Scientific Inc.) was added to 600 μg of protein
ture for 1 h. Again, the wells aspirated and washed three to lysate and placed on a rotator at 4 °C for 2 h. Samples were
five times. Each well was added with 100 μL of Avidin-HRP then washed five times in wash buffer (25 mM Tris pH 7.6,
diluted in 1× ELISA/ELISPOT diluent and incubated at room 150 nM NaCl, 0.5% Triton X-100); beads were pulled down
temperature for 30 min. One hundred microliters of 1× TMB after each wash by gentle centrifugation. Bound proteins were
Solution was added to each well followed by five to seven eluted in 2× SDS reducing buffer and gently heated at 60 °C
washes. After the 15 min of incubation, 50 μL of Stop for 30 min. The resulting supernatant was transferred to new
Solution was added to each well. The plate was read at tubes and heated at 90 °C for 5 min before gel loading.
450 nm.
Statistical Analysis
Immunoblotting
All data are means ± s.e.m. of at least three independent ex-
Hippocampal tissue lysates were prepared using 1× RIPA periments. Statistical analyses were performed with two-
Buffer (Cell Biolabs, Inc., Catalog number: AKR 190) con- tailed, unpaired Student’s t test. Values of P > 0.05 were con-
taining protease inhibitor cocktail (immediately added before sidered not significant (n.s.); values of P < 0.05 and P < 0.01
use). Protein concentrations were determined in a BCA assay. were considered significant and highly significant,
Proteins were separated in 10–12% gels and transferred to respectively.
PVDF membranes (Millipore, Bedford, MA, USA), which
were incubated overnight with primary antibodies. The fol-
lowing primary antibodies were purchased from Cell Results
Signaling Technology (MA, USA): anti-caspase-3 (1:1000
dilution), anti-cleaved caspase-3 (1:500), anti-NF-κB-p65 Avenanthramide-C Restores Aβ-Mediated LTP
(1:1000), anti-phospho-NF-κB-p65 (1:1000), anti-phospho- Inhibition and Impaired LTP in AD Mouse Models
IKKα/β (1:1000), anti-GSK-3α/β (1:1000), anti-phospho-
GSK-3β(Ser9) (1:1000), and anti-β-actin (1:1000). The anti- To analyze the protective effects of Avn-C on Aβ-mediated
IKKα/β (1:1000) antibody was purchased from Santa Cruz impairment of synaptic function, we conducted LTP
Biotechnology (Santa Cruz, CA, USA). Membranes were analysis—a technique used to study synaptic events occurring
then incubated with rabbit IgG antibodies (1:5000, Cell during learning and memory. We treated slices of WT mouse
Signaling Technology) conjugated to horseradish peroxidase hippocampi (an acute preparation) with Aβ42 oligomers in the
and immunoblotted using an ECL detection system presence or absence of different concentrations of Avn-C (10,
(Millipore, Bedford, MA, USA). Densitometric analysis of 25, and 50 μM) and analyzed LTP. Slices treated with Aβ42
the immunoreactive bands was carried out using ImageJ soft- (0.5 μM, 2 h) alone showed impaired LTP, whereas transient
ware (National Institutes of Health, Bethesda, MD). Intensity pretreatment with Avn-C (50 μM, 30 min) followed by co-
of bands from the protein of interest were normalized to the treatment with Aβ42 restored LTP (Fig. 1a). Avn-C at 10 and
intensity of actin bands of the respective blots. The statistical 25 μM concentrations did not improve the LTP significantly
Mol Neurobiol

Fig. 1 Avn-C restores Aβ-mediated LTP inhibition and cognitive deficits. 4%, n = 5); P < 0.01. d Levels of active forms of GSK3β (Ser9) and
a High-frequency stimulation (2× tetanus) induces LTP in control slices caspase-3 in WT, 5XFAD control, and Avn-C-treated hippocampal slices
(closed circles, 158 ± 5%, n = 6), but not following Aβ treatment (open (n = 3). e LTP is induced in hippocampal slices from another
circles, 102 ± 7%, n = 6). However, Avn-C pretreatment blocks Aβ- amyloidogenic mouse model Tg2576 mice pretreated with Avn-C (trian-
mediated LTP inhibition (triangles, 152 ± 10%, n = 6); P < 0.05. b Aβ gles, 138 ± 3%, n = 5) and WT (open circles, 151.2 ± 4%, n = 5) but not in
treatment alters the levels of GSK3β (Ser9) and caspase-3 (C-3, cleaved Tg2576 control (closed circles, 105 ± 3%, n = 5); P < 0.05. f Levels of
C-3; Clv. C-3) active forms, whereas Avn-C pretreatment normalizes these GSK3β (Ser9) and caspase-3 active forms in WT, Tg2576 control, and
protein levels (n = 3). c LTP was induced in hippocampal slices from WT Avn-C-treated hippocampal slices (n = 3). In all panels, error bars indicate
(open circles, 150.8 ± 3%, n = 5) and Avn-C pretreated 5XFAD mice (tri- the SEM; *P < 0.05, **P < 0.01, ***P < 0.001. Black box in LTP data
angles, 135 ± 3%, n = 5) but not in 5XFAD controls (closed circles, 112 ± indicates tetanus stimulation
Mol Neurobiol

(Additional file 1: Fig. S1a, b). Involvement of caspase-3 and were observed in (6.0 mg/kg) Avn-C-treated 7–8 months aged
GSK3β in Aβ42-mediated LTP impairment has been reported Tg2576 mice (Fig. 2b, c).
[7]. To confirm whether Avn-C exerts its effects by inhibiting Based on these observations, we decided to check whether
these molecules, we performed immunoblot analyses to detect the oral treatment with Avn-C could improve cognition in AD
activity of GSK3β and caspase-3 in hippocampal slices; con- mice. To do that, we orally administered 5XFAD mice (5–
sistent with previous reports we found increased cleaved 6 months old) with Avn-C (6 mg/kg) or vehicle once daily
caspase-3 levels and decreased phospho-GSK3β (Ser9) for 2 weeks. At the end of the treatment, CSF was collected
levels, in Aβ42-treated samples (Fig. 1b) [7, 32]; however, from 5XFAD mice and analyzed for the presence of Avn-C.
50 μM Avn-C treatment restored these to basal levels. Also, CSF analysis confirmed that Avn-C can cross through the
we investigated 50 μM Avn-C’s effect on WT mouse hippo- blood-brain barrier (Additional file 3: Fig. S3). Avn-C treat-
campal LTP and the activity of GSK3β and caspase-3. LTP ment improved novel object recognition memory in 5XFAD
analysis and immunoblot results revealed that Avn-C has no mice comparable with the wild-type counterpart while the
effect on WT mouse hippocampal LTP (Additional file 2: Fig. vehicle-treated 5XFAD mice scored poorly (Fig. 2f). Next
S2a, b). we employed Morris water maze (MWM) test—a spatial
To assess the effects of Avn-C on synaptic function in a memory task that depends on the hippocampus, to analyze
transgenic mouse model of AD, we treated hippocampal slices whether Avn-C could enhance learning and memory retrieval.
from 5- to 6-month-old 5XFAD mice ex vivo with Avn-C, and The mice were trained on the fixed hidden platform with vi-
performed LTP. LTP generated in the 5XFAD mice slice was sual reference. The platform was removed at 24 h after the last
significantly reduced compared with the WT controls. training and the mice were tested to visit the location of re-
Remarkably, Avn-C treatment significantly increased the moved platform. Compared to WT mice, vehicle-treated
fEPSP slope of the 5XFAD mice hippocampal slices com- 5XFAD mice were severely impaired in the probe test
pared with that of the untreated controls (Fig. 1c). Consistent displaying significant loss of the memory for the platform
with previous results (Fig. 1b), Avn-C treatment (50 μM) re- location whereas Avn-C treated 5XFAD mice showed less
duced cleaved caspase-3 levels and increased p-GSK3β latency time in seconds to reach the hidden platform and sig-
(Ser9) levels, comparable with wild-type controls (Fig. 1d). nificantly increased time spent in the target quadrant with
We also tested the effects of Avn-C on Tg2576 mice, another increased number of platform crossings indicating rescue of
mouse model of amyloid deposition. As expected, tetanus reference memory (Fig. 2g–j). We also tested motor-related
stimulation induced LTP in WT mouse slices, but failed to behavioral phenotypes, including rotarod test, pole test, and
induce LTP in Tg2576 control mouse hippocampal slices. open field test, in 5XFAD mice after Avn-C treatment and
However, Avn-C treatment (50 μM) for 2 h restored LTP in observed no significant effect in basal motor function
hippocampal slices from 7- to 8-month-old Tg2576 mice (Fig. (Additional file 4: Fig. S4a–c). Collectively, these data suggest
1e). We also observed reduced cleaved caspase-3 and in- that Avn-C treatment restores impaired synaptic function
creased p-GSK3β (Ser9) levels, in Avn-C-treated samples in vivo in two different mouse models of AD pathology and
(Fig. 1f). Thus, Avn-C restores hippocampal LTP irrespective also indicate that at least 2 weeks of Avn-C (daily dosage,
of Aβ burden in two different mouse models of AD. 6 mg/kg) treatment is necessary to reverse the impaired cog-
nition in AD mice.
Avn-C Oral Administration Improves Spatial
and Recognition Memory in AD Transgenic Mice Avn-C Administration Suppresses Neuroinflammation
But Had No Effect on Aβ Pathology
Next, we examined the effects of Avn-C in vivo. 5XFAD mice
aged 5–6 months were treated with Avn-C (2, 4, and Following these results, we determined to check whether
6.0 mg/kg, oral administration) or vehicle once daily for 1 or this behavioral improvement could be the effect of Avn-C
2 weeks and brain slices were prepared from these mice and on Aβ pathology in AD mice. For that, we assessed the
analyzed for hippocampal LTP ex vivo (Fig. 2a). One week Aβ burden in these Avn-C-treated AD transgenic mice
Avn-C treatment did not improve impaired LTP at any given hippocampi. Immunoblot analysis showed that 2 weeks
dosages in 5XFAD mice (data not shown). In contrast, 2 weeks Avn-C treatment did not have any effect on hippocampal
Avn-C treatment at 6 mg/kg was associated with increased Aβ-related protein expression. No significant difference
LTP (Fig. 2d), suppressed cleaved caspase-3, and increased was found in the protein levels of BACE1, APP, RIPA-
p-GSK3β (Ser9) levels comparable to the vehicle-treated con- soluble Aβ fractions, and phospho-tau between vehicle-
trols (Fig. 2e), whereas 2 mg/kg Avn-C treatment showed no a n d Av n - C - t r e a t e d t r a n s g en i c m i c e h i p p o c a m p i
effect in LTP while 4 mg/kg showed little, but not significant (Fig. 3a, b). 5XFAD mice begin accumulating amyloid
enhancement in impaired LTP in 5XFAD mice (data not depositions as early as 2 months of age and increasing
shown). Following this, similar LTP and immunoblot results rapidly with age. We next performed Aβ histopathology
Mol Neurobiol
Mol Neurobiol

ƒFig. 2 Avn-C oral administration improves cognitive deficits in AD Avn-C Improves Memory and Suppresses
mice. a Avn-C or vehicle was orally administered for 2 weeks (6 mg/kg Neuroinflammation in AD Mice Through AMPK
bodyweight per day), followed by analysis of LTP, recognition and spatial
learning, and memory tasks. b–e Two weeks Avn-C oral treatment res-
Activation
cued LTP (Tg2576 + Avn-C—open circles, 154.6 ± 6%, n = 6; 5XFAD +
Avn-C—open circles, 151.8 ± 3%, n = 6) and restored GSK3β (Ser9) and Polyphenolic compounds from natural products exert benefi-
caspase-3 active forms to their basal level in both mice hippocampal cial effects by activating adenosine monophosphate-activated
slices. f Avn-C ameliorates impaired object recognition memory in
5XFAD mice. Bar diagram indicates the object preference index
protein kinase (AMPK) [33], a heterotrimeric serine/threonine
(5XFAD: n = 8; Avn-C-treated 5XFAD: n = 10; WT: n = 8). f (top) kinase that plays a critical role in cell death and survival.
Schematic representation of novel object recognition test. g–j Avn-C also Critically, AMPK inhibits inflammation by regulating several
restored impaired spatial memory during the Morris water maze in pathways [34], and AMPK activation alleviates caspase-3 ac-
5XFAD mice (5XFAD: n = 8; Avn-C-treated 5XFAD: n = 10; WT: n =
8). g (top) The apparatus setting for Morris water maze. The square
tivity by increasing expression of anti-apoptotic proteins Bcl-2
indicated in the southeast quadrant (3) represents the site of the hidden and survivin [35]. Therefore, we investigated whether Avn-C
platform. Mice were placed into the northwest quadrant to start each trial exerts its anti-caspase-3 and anti-inflammatory effects through
(1). g (below) The swim paths of representative individual mice in the last AMPK activation. Immunoblotting (Fig. 4a, b) results showed
training trial. h Latency to hidden platform in the training session. Error
bar indicates the SEM; *P < 0.05, **P < 0.01, ***P < 0.005, significant
that Avn-C oral treatment (6 mg/kg, once daily for 2 weeks)
between wild type and 5XFAD. #P < 0.05, ##P < 0.01, ###P < 0.005, increased phospho-AMPK levels in both 5XFAD and Tg2576
significant between 5XFAD and 5XFAD-Avn-C. i Time spent in the mouse hippocampus. A previous study showed that AMPK
target quadrant in the probe test. j Number of mice which crossed the activation reduces neuropathological deficits and improves
location of (removed) platform in probe test. In all panels, error bars
indicate the SEM; *P < 0.05, **P < 0.01, ***P < 0.001. Black box sym-
plasticity in a mouse model of AD [36]. Our results also
bol in LTP data indicates tetanus stimulation showed that Avn-C restored LTP in mouse models of AD
through AMPK activation. To confirm this, we pretreated
5XFAD and Tg2576 mouse acute hippocampal slices with
the AMPK inhibitor Compound C and analyzed LTP.
analyses in Avn-C-treated 5XFAD mice. IHC quantitative Pretreatment with Compound C followed by Avn-C co-treat-
analysis revealed no significant differences in the total ment inhibited the effects of Avn-C on hippocampal LTP in
amyloid burden in the hippocampus between vehicle- both 5XFAD (Fig. 4c) and Tg2576 mice (Fig. 4e), and re-
and Avn-C-treated 5XFAD mice (Fig. 3c). Growing evi- versed Avn-C-mediated increases in p-GSK3β (Ser9) levels,
dence from various studies has implicated the role of in- along with reduced cleaved caspase-3 levels, in both models
flammation in AD progression. Increased levels of (Fig. 4d–f). Compound C alone did not have any impact on
microglial activation and pro-inflammatory markers have control LTP in both AD mice (Additional file 5: Fig. S5a, b).
been observed in brain samples from both AD patients
and AD transgenic mice models. In Tg2576 and 5XFAD Avn-C Induces AMPK Activation Through α-1A
mice, a 2-week treatment with Avn-C significantly re- Adrenergic Receptors
duced the inflammation which was evident from reduced
levels of ionized calcium binding adapter molecule 1 Brain AMPK activation is mainly regulated by α-adrenergic
(Iba1), a marker of activated microglia and inflammation. receptors (α-AR). On ligand binding, α-ARs activate liver ki-
Increased Iba1-positive microglia in the hippocampus of nase B1 or calcium/calmodulin-dependent kinase kinase or in-
vehicle-treated Tg2576 and 5XFAD mice compared to crease AMP levels, leading to subsequent phosphorylation and
vehicle-treated WT mice were normalized in Avn-C- activation of AMPK [37, 38]. α-1A adrenergic receptors are
treated Tg2576 and 5XFAD mice, indicating that Avn-C generally stimulatory, upon their ligand binding showed to pro-
reversed inflammation (Fig. 3d, e). In addition, Avn-C mote hippocampal synaptic plasticity [39]. Phenylephrine is a
treatment significantly suppressed the levels of pro- commercially available selective agonist of α1A adrenergic re-
inflammatory cytokines TNFα and IL6 in 5XFAD and ceptor (α-1A AR) and proven to regulate AMPK by phosphor-
Tg2576 mice hippocampi whereas increasing the levels ylating at two different sites (Thr 172-activation or Ser 485/491-
of anti-inflammatory marker IL-10 as measured by inhibition) [40]. Apart from these two ligands, Leptin is a hor-
ELISA (Fig. 3f, h). Avn-C treatment also decreased the mone that can bind and activate α1A AR. Leptin binding of α-
levels of phospho-forms of IKK and P65 which are the 1A AR leads to AMPK phosphorylation at Thr 172 and activa-
major downstream molecules of cytokine mediated in- tion [41, 42]. Moreover, leptin was also strongly reported to
flammatory pathways in 5XFAD and Tg2576 mice hippo- enhance hippocampal LTP [43] and AMPK activation [44].
campi (Fig. 3g, i). Taken together, these findings indicated Based on these observations from different studies, we speculate
that Avn-C-induced memory enhancement in these mice the possibility that Avn-C also may execute its LTP-restoring
was independent of Aβ pathology whereas reducing pro- effect through interacting with α-1A AR similar to that of leptin.
inflammatory microglial activation. To test this, we used prazosin, an irreversible α-1A AR inhibitor
Mol Neurobiol

a b c d
Tg2576 5XFAD
Tg2576 - Vehicle 5XFAD - Vehicle 5XFAD - Avn-C
Veh Avn-C Veh Avn-C 5XFAD - Vehicle

Normalized O.D. (% of control)


Tg2576 - Avn-C 5XFAD - Avn-C Vehicle

Flourescence intesnsity (%)


Normalized O.D. (% of Control)
BACE1 CA1 CA1 Avn-C
BACE1 n.s. n.s. n.s.
120 n.s n.s n.s n.s n.s.
120 100 n.s
APP 100 APP 100
80

6E10
80 80
60
60 60
40 40
40
p - Tau p-Tau 20 20
20
0 0
0

p-
BA

AP
p-
-actin
AP
BA

-actin

Ta
CE

P
Ta
P

u
CE

1
1

e f WT - Vehicle g h WT - Vehicle
Tg - Vehicle 5X - Vehicle
5X - Avn-C

Fluorescence intesnsity (%)


Tg - Avn-C

Fluorescence intesnsity (%)


WT - Vehicle Tg2576 - Vehicle Tg2576- Avn-C WT - Vehicle 5XFAD - Vehicle 5XFAD - Avn-C
150 150 ** ***
* **
Iba-1

100

Iba-1
100

Iba-1
50 50

0 0

i WT - Vehicle j k WT - Vehicle l
200
Normalized O.D. (% of control)

Tg2576 - Vehicle 5XFAD - Vehicle


150

Normalized O.D. (% of control)


Tg2576 - Avn-C ** 5XFAD - Avn-C 200
Tg2576 100 5XFAD
150 ***
Veh Avn-C Veh Avn-C
50 100
Conccentration (pg/ml)
100 150 ** ** 120 *** *** 150
Conccentration (pg/ml)

100 *** * 100


0 *** *** 50
80 * * 80 125 80 100 125 0
100 200 80 100
60 60 * * p65 60 200
75 150 60 75 p65
40 40 *** 40 150 ***
50 p-p65 100 40 50 p-p65 100
20 20 25 50 20 20
-actin 25 50
-actin
0 0 0 0 0 0 0 0
IL-6 IL-10 p65 p-p65 IL-6 IL-10 p65 p-p65

Fig. 3 Avn-C did not reduce amyloid burden but reduced than in those from vehicle-treated controls (scale bar; 20 μm). h
neuroinflammation in AD mice. a, b Western blot and quantitative Quantitative analyses of fluoroscence intensities are shown in graphs. i
analysis of BACE1, APP, Aβ, and phopho-tau levels in hippocampal Enzyme-linked immunosorbent assay (ELISA) histograms (relative) of
extracts from vehicle (Veh) or Avn-C treated (2 weeks) Tg2576 and IL-6, TNFα, and IL-10 in hippocampal homogenates of WT and Tg2576
5XFAD mice. The error bars indicate the mean ± s.e.m. (n = 4). c mice treated with either Avn-C or vehicle. All of the methods and proce-
Fluorescence images of hippocampal regions of vehicle- or Avn-C treated dures recommended by the manufacturer were followed, and experiments
(2 weeks) 5XFAD mice, indicating the localization of Aβ (6E10) aggre- were performed in triplicate. j Western blot and qauntitative analysis of
gates (scale bar; 200 μm). d Quantitative analyses of fluoroscence inten- IKK, p65, and their active form levels in hippocampal extracts from
sities are shown in graphs. e Immunohistological analysis shows reduced vehicle- (Veh) or Avn-C-treated (2 weeks) Tg2576 mice. k ELISA histo-
levels of microglial activation comparable with WT mouse hippocampus grams (relative) of IL-6, TNFα, and IL-10 in hippocampal homogenates
in Avn-C-treated Tg2576 mouse hippocampal slices than in those from of WT and 5XFAD mice treated with Avn-C or vehicle. l Western blot and
vehicle-treated controls (scale bar; 20 μm). f Quantitative analyses of quantitative analysis of IKK, p65, and their active form levels in hippo-
fluoroscence intensities are shown in graphs. g Immunohistological anal- campal extracts from vehicle- (Veh) or Avn-C-treated (2 weeks) 5XFAD
ysis shows reduced levels of microglial activation comparable with WT mice. In all panels, error bars indicate the SEM; *P < 0.05, **P < 0.01,
mouse hippocampus in Avn-C-treated 5XFAD mouse hippocampal slices ***P < 0.001

[45]. Pretreatment with prazosin blocked the effect of Avn-C on activation of caspase-3 and pro-inflammatory markers while
hippocampal LTP in both 5XFAD (Fig. 5a) and Tg2576 (Fig. increased GSK3β (Ser9) phosphorylation and anti-
5c) mice, and reversed the Avn-C-mediated increases in p- inflammatory marker (Fig. 6).
AMPK and p-GSK3β (Ser9) levels, as well as reduced cleaved
caspase-3 levels (Fig. 5b, d). Prazosin alone did not have any
impact on control LTP in both AD mice (Additional file 5: Fig.
S5c, d). To confirm Avn-C binding to α-ARs, we pretreated Discussion
mouse hippocampal slices with biotinylated Avn-C in the pres-
ence or absence of prazosin. Homogenized hippocampal lysates Here we report for the first time that Avn-C, an oats
were pulled down with streptavidin beads and the bead fractions component, restored impaired memory, a major trait in
immunoblotted for α-1A ARs. The pull-down results confirmed AD condition, in both ex vivo and in vivo studies. Avn-
that biotinylated Avn-C binds to α-1A AR (Fig. 5e). Together, C alleviated AD-related memory impairment by
our results demonstrate that Avn-C rescued Aβ-mediated mem- strengthening synapses which was evident from en-
ory impairment via α-1A AR and AMPK results in reduced hanced LTP, reducing caspase-3 and pro-inflammatory
markers activation, and increasing pS9GSK3β and
Mol Neurobiol

a WT b WT
5XFAD Tg2576
WT 5XFAD 5XFAD + Avn-C WT Tg2576 Tg2576 + Avn-C
__ __ + __ __ +

Normalized O.D.

Normalized O.D.
Avn-C 200 Avn-C 200

(% of control)

(% of control)
AMPK 150 * ** AMPK 150
* **
100 100
p -AMPK p -AMPK
50 50
β-actin 0 β-actin 0
AMPK p-AMPK AMPK p-AMPK

c 1 2 2+3 5 6 5+6 d

1 mV
Con Avn-C
3 4 3+4 Compound C + Avn-C
10 ms 200

Normalized O.D. (% of control)


Compound C (μM) 0 0 10 150
350 ** **
Avn-C (μM) 0 50 50
Normalized fEPSP slope

Control 100
300 Avn-C C-3 50
(%) of baseline)

Comp.C + Avn-C Clv . C -3 0


250 C-3 Clv. C-3
GSK-3ɑβ 200
200 4 ** *
150
pS9GSK-3β
150 6 100
1 5
100 2
E β-actin 50
3 5X FAD 0
50 GSK-3ɑβ pS9GSK-3β
0 10 20 30 40 50 60 70 80 90 100
Time (min)

e 1 2 1+2 5 6 5+6 f
Con Avn-C
1 mV

3 4 3+4 Compound C + Avn-C


200
10 ms
Normalized O.D. (% of control)
Compound C (μM) 0 0 10 150 * *
350 Avn-C (μM) 0 50 50
Control 100
300 Avn-C C-3 50
Normalized fEPSP slope

Comp.C + Avn-C 0
Clv . C -3
(%) of baseline)

250 C-3 Clv. C-3


GSK-3ɑβ 200
200 4 * *
150
pS9GSK-3β
150 6 100
1 5 β-actin
100 2 50
3 Tg2576 0
50 GSK-3ɑβ pS9GSK-3β
0 10 20 30 40 50 60 70 80 90 100
Time (min)
Fig. 4 Avn-C restores impaired LTP by inducing AMPK activation. a, b forms in Compound-C- and Avn-C-treated 5XFAD mouse hippocampi
Immunoblot validation and densitometry analysis of (n = 3) AMPK and (n = 3). e Preincubation of Tg2576 mouse acute hippocampal slices with
phospho-AMPK from WT, vehicle-, or Avn-C-treated 5XFAD and Compound C blocks the effect of Avn-C on LTP (control, 119.5 ± 2.5%,
Tg2576 mouse hippocampal samples. c Acute hippocampal slices from n = 5 closed circles; Avn-C, 160.8 ± 5.8%, n = 4, open circles; Compound
5XFAD mice were pretreated with the AMPK inhibitor Compound C C + Avn-C, 122.7 ± 2.3%, n = 4, triangles; P < 0.05). f Immunoblot vali-
(10 μM) for 1 h prior to Avn-C (2 h) exposure. Preincubation with dation and densitometry analysis (n = 3) of caspase-3, GSK3β (Ser9)
Compound C blocks the effect of Avn-C on LTP (control, n = 5, 117.6 active forms in compound-C- and Avn-C-treated Tg2576 mouse acute
± 3%, closed circles; Avn-C, 163.8 ± 5%, n = 5, open circles; Compound hippocampal samples. Error bars indicate the SEM. *P < 0.05;
C + Avn-C, 124.1 ± 8%, n = 4, triangles; P < 0.05. d Immunoblot valida- **P < 0.01. Black box symbol in LTP data indicates tetanus stimulation
tion and densitometry analysis of GSK3β (Ser9) and caspase-3 active
Mol Neurobiol

a b
1 2 1+2 3 4 3+4 Con Avn-C

1 mV
Prazosin.HCL+ Avn-C

5 6 5+6 10 ms 200 ** * 200


Prazosin.HCL - - +

Normalized O.D. (% of control)


- + + 150 150 ** **
Avn-C
350 100
Normalized fEPSP slope

Control AMPK 100


300 Avn-C 50 50
(%) of baseline)

Prazosin.HCL + Avn-C p-AMPK


0 0
250 AMPK p-AMPK C-3 Clv. C-3
C-3
200 200
4 Clv . C -3 * *
150
150
1 5 6 GSK-3ɑβ 100
100
2 pS9GSK-3β 50
3 5X FAD
50 0
β-actin GSK-3ɑβ pS9GSK-3β
0 10 20 30 40 50 60 70 80 90 100
Time (min)

c d
1 2 1+2 5 6 5+6 Con Avn-C
1 mV

Prazosin.HCL+ Avn-C

10 ms
3 4 3+4 Prazosin.HCL - - + 200 * 200
**

Normalized O.D. (% of control)


Avn-C - + + 150 150
100 100
Control AMPK
300
Normalized fEPSP slope

Avn-C 50 50
Prazosin.HCL + Avn-C p-AMPK
0 0
(%) of baseline)

250
AMPK p-AMPK C-3 Clv. C-3
C-3
200 200
4 Clv . C -3 *
150 150
6
1 5 GSK-3ɑβ 100
100 2
3 pS9GSK-3β 50
Tg2576
50 0
β-actin GSK-3ɑβ pS9GSK-3β
0 10 20 30 40 50 60 70 80 90 100

Time (min)

e Input
Beads
(5%)

α1a- AR

β -actin

Avn-C - + - -
Biotin-Avn-C - - + +
Prazosin+ Avn-C - - - +
Fig. 5 Avn-C restores impaired LTP by inducing AMPK activation 155.5 ± 3.6%, n = 6, triangles; prazosin + Avn-C, 116.8 ± 3.2%, n = 5,
through α-1A adrenergic receptors. a Prazosin pretreatment of hippocam- closed circles; P < 0.05). d Immunoblot validation and densitometry anal-
pal slices from 5XFAD mice blocks the effect of Avn-C on LTP (control, ysis of (n = 3) of AMPK, GSK3β (Ser9), and caspase-3 active forms in
122.3 ± 4%, n = 4, closed circles; Avn-C, 164.0 ± 7%, n = 4, triangles; prazosin- and Avn-C-treated Tg2576 mouse hippocampus. Error bars
Prazosin + Avn-C, 125.8 ± 3%, n = 4, open circles; P < 0.05). b indicate the SEM. *P < 0.05; **P < 0.01. Black box symbol in LTP data
Immunoblot validation and densitometry analysis of western blot bands indicates tetanus stimulation. e Avn-C binds α-1A adrenergic receptors
of (n = 3) AMPK, GSK3β (Ser9), and caspase-3 active forms in prazosin- (α-1A AR). Hippocampal slices were treated with biotinylated Avn-C in
and Avn-C-treated 5XFAD mouse hippocampus. c Prazosin hydrochlo- the presence or absence of prazosin. Tissue lysates were pulled down with
ride pretreatment of Tg2576 mouse hippocampal slices blocks the effect streptavidin beads, followed by western blotting for α-1A AR. In all
of Avn-C on LTP (control, 116.3 ± 0.8%, n = 6, open circles; Avn-C, panels, error bars indicate the SEM; *P < 0.05; **P < 0.01; ***P < 0.001

IL10 levels in two different mouse models of AD. Our molecules in AD and inhibitory effect of Avn-C on
data support the generally adverse role of these these molecules results in memory restoration. The
Mol Neurobiol

significantly reduced the inflammatory processes in these


mice. Inflammatory processes are thought to have an active
role in AD formation and progression. Multiple lines of evi-
dence showed elevated levels of pro-inflammatory markers
such as IL-1β, IL-6, TNFα, and activated microglia in AD
patients CSF and brain samples [51–54]. Aβ-induced elevated
levels of TNFα showed to impair LTP in mouse hippocampi
[55, 56]. Inhibition of dysregulated inflammatory pathways
showed to improve memory in AD mice [57, 58]. In our study,
we showed that Avn-C treatment significantly reduced the
expression of pro-inflammatory cytokines whereas it in-
creased the expression of anti-inflammatory cytokine in both
Tg2576 and 5XFAD mice hippocampi. Furthermore, our data
Fig. 6 Schematic representation of the signaling mechanisms involved in suggests that Avn-C exerts its effect through the activation of
Avn-C-mediated LTP restoration. Aβ treatment inhibits LTP by activat- AMPK. Activation of AMPK has been linked to regulate and
ing caspase-3/GSK3β signaling and pro-inflammatory markers. Avn-C reduce inflammation and memory enhancement [34, 59, 60].
blocks these signaling pathways and rescues LTP by inducing AMPK
phosphorylation through binding to α-1A AR. Pretreatment with either Orally administered resveratrol mediated AMPK activation
Compound C or α-1A AR irreversible inhibitor prazosin hydrochloride results in improved memory and reduced Aβ burden in
blocks the LTP-restoring effect of Avn-C APP/PS1 mice [61, 62]. In support of this, AMPK activator
metformin treatment ameliorated cognitive impairment and
beneficial effects of Avn-C are correlated with its ability reduced Aβ burden in APP/PS1 mice [63]. Similar to these
to modulate α-1A AR, activate AMPK, and downregu- results, our study also confirms the inhibition of AMPK with
late the factors involved in neuroinflammation. Compound C or prazosin HCl blocked the LTP in acute hip-
Synaptic dysfunction is the early and major contributor to pocampal slices of transgenic mouse models. Furthermore,
learning and memory impairment in AD. Finding therapeutic pull-down assay with biotinylated Avn-C-treated hippocam-
agents that facilitate the synaptic strengthening would be ben- pal slice samples showed the interaction between Avn-C and
eficial for memory retrieval in AD. With that in mind, initially α-adrenergic receptors which was inhibited in the presence of
we checked the effect of Avn-C on hippocampal LTP in prazosin HCl. These results indicate that the LTP-restoring
Tg2576 and 5XFAD mice. Both these mice overexpress mu- property of Avn-C is due to activation of AMPK through α-
tant form of human APP and shown to have early LTP impair- adrenergic receptors which results in reduced IKK, NFkB
ment [46–48]. LTP induction in both transgenic mice was not p65, caspase-3 activation, and increased GSK3β (S9)
altered by Avn-C treatment but the LTP maintenance was phosphorylation.
enhanced in case of Avn-C-treated slices compared to its un- Noradrenergic receptors plays major role in learning and
treated control. LTP maintenance requires strengthening of memory process and their dysfunctions have been reported in
synapses and this data suggests Avn-C promotes it. Caspase- mild and moderate AD conditions. The α-1A ARs are G
3 reported to inhibit LTP and promotes LTD in Aβ-treated protein-coupled receptors highly expressed in cognitive cen-
hippocampi [7, 49, 50]. We also observed increased levels of ters of the central nervous system, including the prefrontal
cleaved caspase-3 in both AD mice hippocampi and Avn-C cortex and hippocampus [64]. These receptors are generally
treatment reduced its levels which correlate with increased stimulatory, and their role in plasticity has been documented;
LTP. Moreover, LTP analyses and immunoblot data from binding of its ligand, norepinephrine, promotes hippocampal
2 weeks orally administered Tg2576 and 5XFAD mice LTP [39]. Furthermore, α-1A AR dysfunction is implicated in
showed similar LTP enhancement and cleaved caspase-3 re- AD. For example, polymorphisms in the α-1A AR gene are
duction levels. In addition, Avn-C oral administration im- associated with AD susceptibility [65]. In addition, long-term
proved hippocampus-dependent recognition and spatial mem- stimulation of α-1A ARs increases neurogenesis and im-
ory in 5XFAD mice. proves synaptic plasticity [66]. These studies suggest that in-
We next investigated whether the Avn-C-induced memory creasing α-1A AR activity may delay or ameliorate AD symp-
retrieval could be the results of its effect on amyloid pathology toms. In contrast to these above observations, a small cohort
in these mice. Thus, we examined the levels of BACE1, APP, study on prazosin shown to alleviate agitation and aggressive
soluble Aβ, and phosphorylated tau using immunoblot and behavior in AD patients as a result of α-1A ARs inhibition
IHC staining. Aβ-related proteins and phosphorylated tau [67]. However, the finding in our study strongly suggests that
remained unchanged by Avn-C treatment in 5XFAD and Avn-C may improve synaptic plasticity and behavioral defi-
Tg2576 mice, suggesting that the protective effects are not cits by modifying neuroinflammation, although further studies
Aβ or tau dependent. Nonetheless, Avn-C administration are required to substantiate this.
Mol Neurobiol

Conclusions networks. Nat Neurosci 13(7):812–818. https://doi.org/10.1038/


nn.2583
2. Querfurth HW, LaFerla FM (2010) Alzheimer’s disease. N Engl J
In conclusion, this study provides the first evidence of the Med 362(4):329–344. https://doi.org/10.1056/NEJMra0909142
anti-AD properties of Avn-C, a natural compound that accu- 3. Thompson S, Lanctôt KL, Herrmann N (2004) The benefits and
mulates and is biologically available when orally administered risks associated with cholinesterase inhibitor therapy in
to rats and humans [68, 69]. Results from our CSF analysis Alzheimer’s disease. Expert Opin Drug Saf 3(5):425–440. https://
doi.org/10.1517/14740338.3.5.425
also confirmed the presence of Avn-C which supports its brain 4. Ellis JM (2005) Cholinesterase inhibitors in the treatment of demen-
entry through blood-brain barrier membrane tia. J Am Osteopath Assoc 105(3):145–158. https://doi.org/10.
(Additional file 3: Fig. S3). No apparent toxicity or adverse 7556/jaoa.2005.105.3.145
effects of Avn-C were reported in these previous studies [68, 5. Lukiw WJ (2012) Amyloid beta (Abeta) peptide modulators and
other current treatment strategies for Alzheimer’s disease (AD).
69]. Our LTP analysis data also suggests that no toxic effects
Expert Opin Emerg Drugs 17:43–60. https://doi.org/10.1517/
were observed even at high concentrations (Additional file 6: 14728214.2012.672559
Fig. S6). Presently available drugs to treat AD cause adverse 6. Chapman PF, White GL, Jones MW, Cooper-Blacketer D, Marshall
effects upon long-term dosage, which restricts their use. By VJ, Irizarry M, Younkin L, Good MA et al (1999) Impaired synap-
contrast, Avn-C has few or no adverse effects, making it a tic plasticity and learning in aged amyloid precursor protein trans-
genic mice. Nat Neurosci 2:271–276. https://doi.org/10.1038/6374
potential alternative therapeutic candidate. Overall, we sug- 7. Jo J, Whitcomb DJ, Olsen KM, Kerrigan TL, Lo SC, Bru-Mercier
gest that this compound warrants development as a potential G, Dickinson B, Scullion S et al (2011) Abeta(1-42) inhibition of
therapeutic for AD. LTP is mediated by a signaling pathway involving caspase-3, Akt1
and GSK-3beta. Nat Neurosci 14(5):545–547. https://doi.org/10.
Author Contributions The study was conceived by H.K. and J.J. The 1038/nn.2785
experiments were designed by H.K., W.C., Y.L., and J.J. and carried out 8. Snyder EM, Nong Y, Almeida CG, Paul S, Moran T, Choi EY,
by V.S.R., M.S., S.Y.Y., H.K.K., and S.H. The manuscript was written by Nairn AC, Salter MW et al (2005) Regulation of NMDA receptor
V.S.R., H.K., W.C., Y.L., and J.J. All authors read and approved the final trafficking by amyloid-beta. Nat Neurosci 8(8):1051–1058. https://
manuscript. doi.org/10.1038/nn1503
9. Hsieh H, Boehm J, Sato C, Iwatsubo T, Tomita T, Sisodia S,
Malinow R (2006) AMPAR removal underlies Abeta-induced syn-
Funding Information This work was funded by the Pioneer Research
aptic depression and dendritic spine loss. Neuron 52(5):831–843.
Center Program through the National Research Foundation of Korea
https://doi.org/10.1016/j.neuron.2006.10.035
funded by the Ministry of Science and ICT (NRF-
10. Griffin WS, Sheng JG, Roberts GW, Mrak RE (1995) Interleukin-1
2014M3C1A3053029), the Cooperative Research Program for
expression in different plaque types in Alzheimer’s disease: signif-
Agriculture Science & Technology Development (PJ010508042014,
icance in plaque evolution. J Neuropathol Exp Neurol 54(2):276–
PJ01255102, and PJ012551042019), the Rural Development
281
Administration, and the Republic of Korea, National Research
Foundation of Korea (NRF) grant funded by the Korea government 11. Huell M, Strauss S, Volk B, Berger M, Bauer J (1995) Interleukin-6
(NRF-2016R1A2B4008316, 2019R1A2C1004575). is present in early stages of plaque formation and is restricted to the
brains of Alzheimer’s disease patients. Acta Neuropathol 89(6):
544–551
Compliance with Ethical Standards 12. Dickson DW, Lee SC, Mattiace LA, Yen SH, Brosnan C (1993)
Microglia and cytokines in neurological disease, with special refer-
Ethics Approval and Consent to Participate Not applicable. ence to AIDS and Alzheimer’s disease. Glia 7(1):75–83. https://doi.
org/10.1002/glia.440070113
Consent for Publication Not applicable. 13. Sokolova A, Hill MD, Rahimi F, Warden LA, Halliday GM,
Shepherd CE (2009) Monocyte chemoattractant protein-1 plays a
dominant role in the chronic inflammation observed in Alzheimer’s
Competing Interests The authors declare that they have no conflict of
disease. Brain Pathol 19(3):392–398. https://doi.org/10.1111/j.
interests.
1750-3639.2008.00188.x
14. McGeer PL, McGeer EG (1996) Anti-inflammatory drugs in the
Abbreviations AD, Alzheimer’s disease; aCSF, artificial cerebrospinal fight against Alzheimer’s disease. Ann N Y Acad Sci 777:213–220
fluid; Aβ42, Amyloid β1-42; AMPK, Adenosine mono phosphate kinase; 15. Yoshiyama Y, Higuchi M, Zhang B, Huang SM, Iwata N, Saido TC,
α-1A AR, alpha 1A adrenergic receptors; CA1, cornu ammonis area 1; Maeda J, Suhara T et al (2007) Synapse loss and microglial activa-
CA3, cornu ammonis area 3; fEPSP, field excitatory postsynaptic poten- tion precede tangles in a P301S tauopathy mouse model. Neuron
tial; LTP, long-term potentiation; GSK3β, glycogen synthase kinase β; 53(3):337–351. https://doi.org/10.1016/j.neuron.2007.01.010
IL, interleukin; TNFα, tumor necrosis factor-α; Tg2576 and 5XFAD,
16. Ittner LM, Ke YD, Delerue F, Bi M, Gladbach A, van Eersel J,
transgenic AD mouse models
Wolfing H, Chieng BC et al (2010) Dendritic function of tau medi-
ates amyloid-beta toxicity in Alzheimer’s disease mouse models.
Cell 142(3):387–397. https://doi.org/10.1016/j.cell.2010.06.036
17. Szekely CA, Zandi PP (2010) Non-steroidal anti-inflammatory
drugs and Alzheimer’s disease: the epidemiological evidence.
References CNS Neurol Disord Drug Targets 9(2):132–139. https://doi.org/
10.2174/187152710791012026
1. Palop JJ, Mucke L (2010) Amyloid-beta-induced neuronal dys- 18. Lakey-Beitia J, Berrocal R, Rao KS, Durant AA (2015)
function in Alzheimer’s disease: from synapses toward neural Polyphenols as therapeutic molecules in Alzheimer’s disease
Mol Neurobiol

through modulating amyloid pathways. Mol Neurobiol 51(2):466– atherosclerosis in diabetic LDL receptor-deficient mice. Diabetes
479. https://doi.org/10.1007/s12035-014-8722-9 55(8):2180–2191. https://doi.org/10.2337/db05-1188
19. Mahler A, Mandel S, Lorenz M, Ruegg U, Wanker EE, Boschmann 34. Salminen A, Hyttinen JM, Kaarniranta K (2011) AMP-activated
M, Paul F (2013) Epigallocatechin-3-gallate: a useful, effective and protein kinase inhibits NF-kappaB signaling and inflammation: im-
safe clinical approach for targeted prevention and individualised pact on healthspan and lifespan. J Mol Med 89(7):667–676. https://
treatment of neurological diseases? EPMA J 4(1):5. https://doi. doi.org/10.1007/s00109-011-0748-0
org/10.1186/1878-5085-4-5 35. Liu C, Liang B, Wang Q, Wu J, Zou MH (2010) Activation of
20. Pasinetti GM, Wang J, Ho L, Zhao W, Dubner L (2015) Roles of AMP-activated protein kinase alpha1 alleviates endothelial cell ap-
resveratrol and other grape-derived polyphenols in Alzheimer’s dis- optosis by increasing the expression of anti-apoptotic proteins Bcl-2
ease prevention and treatment. Biochim Biophys Acta 1852(6): and survivin. J Biol Chem 285(20):15346–15355. https://doi.org/
1202–1208. https://doi.org/10.1016/j.bbadis.2014.10.006 10.1074/jbc.M110.102491
21. Goozee KG, Shah TM, Sohrabi HR, Rainey-Smith SR, Brown B, 36. Shah SA, Yoon GH, Chung SS, Abid MN, Kim TH, Lee HY, Kim
Verdile G, Martins RN (2016) Examining the potential clinical val- MO (2017) Novel osmotin inhibits SREBP2 via the AdipoR1/
ue of curcumin in the prevention and diagnosis of Alzheimer’s AMPK/SIRT1 pathway to improve Alzheimer’s disease neuropath-
disease. Br J Nutr 115(3):449–465. https://doi.org/10.1017/ ological deficits. Mol Psychiatry 22(3):407–416. https://doi.org/10.
S0007114515004687 1038/mp.2016.23
2 2. G u o W, Wis e M L , C o l l i n s FW, M e y da ni M ( 20 0 8 ) 37. Hutchinson DS, Bengtsson T (2006) AMP-activated protein kinase
Avenanthramides, polyphenols from oats, inhibit IL-1beta- activation by adrenoceptors in L6 skeletal muscle cells. Diabetes
induced NF-kappaB activation in endothelial cells. Free Radic 55(3):682–690
B iol Med 44(3 ):4 15– 429 . h ttps:/ /doi.o rg/1 0.10 16/j. 38. Kahn BB, Alquier T, Carling D, Hardie DG (2005) AMP-activated
freeradbiomed.2007.10.036 protein kinase: ancient energy gauge provides clues to modern un-
23. Nie L, Wise ML, Peterson DM, Meydani M (2006) derstanding of metabolism. Cell Metab 1(1):15–25. https://doi.org/
Avenanthramide, a polyphenol from oats, inhibits vascular smooth 10.1016/j.cmet.2004.12.003
muscle cell proliferation and enhances nitric oxide production. 39. Izumi Y, Zorumski CF (1999) Norepinephrine promotes long-term
Atherosclerosis 186(2):260–266. https://doi.org/10.1016/j. potentiation in the adult rat hippocampus in vitro. Synapse 31(3):
atherosclerosis.2005.07.027 196–202. https://doi.org/10.1002/(SICI)1098-2396(19990301)31:
24. Yang J, Ou B, Wise ML, Chu Y (2014) In vitro total antioxidant 3<196::AID-SYN4>3.0.CO;2-K
capacity and anti-inflammatory activity of three common oat- 40. Woods A, Vertommen D, Neumann D, Turk R, Bayliss J, Schlattner
derived avenanthramides. Food Chem 160:338–345. https://doi. U, Wallimann T, Carling D et al (2003) Identification of phosphor-
org/10.1016/j.foodchem.2014.03.059 ylation sites in AMP-activated protein kinase (AMPK) for upstream
AMPK kinases and study of their roles by site-directed mutagene-
25. Ji LL (2003) Effects of avenanthramides on oxidant generation and
sis. J Biol Chem 278(31):28434–28442. https://doi.org/10.1074/
antioxidant enzyme activity in exercised rats. Nutr Res 23(11):1579
jbc.M303946200
1570-2003 v.1523 no.1511. https://doi.org/10.1016/S0271-
41. Miyamoto L, Ebihara K, Kusakabe T, Aotani D, Yamamoto-
5317(03)00165-9
Kataoka S, Sakai T, Aizawa-Abe M, Yamamoto Y et al (2012)
26. Koenig R, Dickman JR, Kang C, Zhang T, Chu YF, Ji LL (2014)
Leptin activates hepatic 5′-AMP-activated protein kinase through
Avenanthramide supplementation attenuates exercise-induced in-
sympathetic nervous system and alpha1-adrenergic receptor: a po-
flammation in postmenopausal women. Nutr J 13:21. https://doi.
tential mechanism for improvement of fatty liver in lipodystrophy
org/10.1186/1475-2891-13-21
by leptin. J Biol Chem 287(48):40441–40447. https://doi.org/10.
27. Hassanein KM, El-Amir YO (2017) Protective effects of 1074/jbc.M112.384545
thymoquinone and avenanthramides on titanium dioxide nanopar- 42. Shi T, Papay RS, Perez DM (2017) The role of alpha1-adrenergic
ticles induced toxicity in Sprague-Dawley rats. Pathol Res Pract receptors in regulating metabolism: increased glucose tolerance,
213(1):13–22. https://doi.org/10.1016/j.prp.2016.08.002 leptin secretion and lipid oxidation. J Recept Signal Transduct
28. Whitcomb DJ, Hogg EL, Regan P, Piers T, Narayan P, Whitehead Res 37(2):124–132. https://doi.org/10.1080/10799893.2016.
G, Winters BL, Kim D-H et al (2015) Intracellular oligomeric 1193522
amyloid-beta rapidly regulates GluA1 subunit of AMPA receptor 43. Oomura Y, Hori N, Shiraishi T, Fukunaga K, Takeda H, Tsuji M,
in the hippocampus. Sci Rep 5:10934. https://doi.org/10.1038/ Matsumiya T, Ishibashi M et al (2006) Leptin facilitates learning
srep10934 and memory performance and enhances hippocampal CA1 long-
29. Choi W-S, Kim H-W, Tronche F, Palmiter RD, Storm DR, Xia Z term potentiation and CaMK II phosphorylation in rats. Peptides
(2017) Conditional deletion of Ndufs4 in dopaminergic neurons 27(11):2738–2749. https://doi.org/10.1016/j.peptides.2006.07.001
promotes Parkinson’s disease-like non-motor symptoms without 44. Suzuki A, Okamoto S, Lee S, Saito K, Shiuchi T, Minokoshi Y
loss of dopamine neurons. Sci Rep 7:44989. https://doi.org/10. (2007) Leptin stimulates fatty acid oxidation and peroxisome
1038/srep44989 proliferator-activated receptor alpha gene expression in mouse
30. Kim S-M, Mun B-R, Lee S-J, Joh Y, Lee H-Y, Ji K-Y, Choi H-R, C2C12 myoblasts by changing the subcellular localization of the
Lee E-H et al (2017) TREM2 promotes Aβ phagocytosis by upreg- alpha2 form of AMP-activated protein kinase. Mol Cell Biol
ulating C/EBPα-dependent CD36 expression in microglia. Sci Rep 27(12):4317–4327. https://doi.org/10.1128/MCB.02222-06
7(1):11118–11118. https://doi.org/10.1038/s41598-017-11634-x 45. Timmermans PB, Thoolen MJ, Mathy MJ, Wilffert B, de Jonge A,
31. McIlwain KL, Merriweather MY, Yuva-Paylor LA, Paylor R (2001) van Zwieten PA (1985) Effects of the irreversible alpha-
The use of behavioral test batteries: effects of training history. adrenoceptor antagonists phenoxybenzamine and benextramine
Physiol Behav 73(5):705–717 on the effectiveness of nifedipine in inhibiting alpha 1- and alpha
32. Shih R-H, Wang C-Y, Yang C-M (2015) NF-kappaB signaling 2-adrenoceptor mediated vasoconstriction in pithed rats. Naunyn
pathways in neurological inflammation: a mini review. Front Mol Schmiedeberg's Arch Pharmacol 329(4):404–413
Neurosci 8:77. https://doi.org/10.3389/fnmol.2015.00077 46. Oakley H, Cole SL, Logan S, Maus E, Shao P, Craft J, Guillozet-
33. Zang M, Xu S, Maitland-Toolan KA, Zuccollo A, Hou X, Jiang B, Bongaarts A, Ohno M et al (2006) Intraneuronal beta-amyloid ag-
Wierzbicki M, Verbeuren TJ et al (2006) Polyphenols stimulate gregates, neurodegeneration, and neuron loss in transgenic mice
AMP-activated protein kinase, lower lipids, and inhibit accelerated with five familial Alzheimer’s disease mutations: potential factors
Mol Neurobiol

in amyloid plaque formation. J Neurosci 26(40):10129–10140. inflammation response in microglia by AMPK activation. Biomed
https://doi.org/10.1523/JNEUROSCI.1202-06.2006 Res Int 2014:9–9. https://doi.org/10.1155/2014/692061
47. Jacobsen JS, Wu C-C, Redwine JM, Comery TA, Arias R, Bowlby 60. Zhu YP, Brown JR, Sag D, Zhang L, Suttles J (2015) Adenosine 5′-
M, Martone R, Morrison JH et al (2006) Early-onset behavioral and monophosphate-activated protein kinase regulates IL-10-mediated
synaptic deficits in a mouse model of Alzheimer’s disease. Proc anti-inflammatory signaling pathways in macrophages. J Immunol
Natl Acad Sci U S A 103(13):5161–5166 194(2):584–594. https://doi.org/10.4049/jimmunol.1401024
48. Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya Y, Younkin S, 61. Vingtdeux V, Giliberto L, Zhao H, Chandakkar P, Wu Q, Simon JE,
Yang F, Cole G (1996) Correlative memory deficits, Aβ elevation, Janle EM, Lobo J et al (2010) AMP-activated protein kinase sig-
and amyloid plaques in transgenic mice. Science 274(5284):99– naling activation by resveratrol modulates amyloid-beta peptide
103 metabolism. J Biol Chem 285(12):9100–9113. https://doi.org/10.
49. Li Z, Jo J, Jia JM, Lo SC, Whitcomb DJ, Jiao S, Cho K, Sheng M 1074/jbc.M109.060061
(2010) Caspase-3 activation via mitochondria is required for long- 62. Porquet D, Grinan-Ferre C, Ferrer I, Camins A, Sanfeliu C, Del
term depression and AMPA receptor internalization. Cell 141(5): Valle J, Pallas M (2014) Neuroprotective role of trans-resveratrol
859–871. https://doi.org/10.1016/j.cell.2010.03.053 in a murine model of familial Alzheimer’s disease. J Alzheimers
50. D’Amelio M, Cavallucci V, Middei S, Marchetti C, Pacioni S, Ferri Dis 42(4):1209–1220. https://doi.org/10.3233/JAD-140444
A, Diamantini A, De Zio D et al (2011) Caspase-3 triggers early 63. Matthes F, Hettich MM, Ryan DP, Ehninger D, Krauss S (2015)
synaptic dysfunction in a mouse model of Alzheimer’s disease. Nat The anti-diabetic drug metformin improves cognitive impairment
Neurosci 14(1):69–76. https://doi.org/10.1038/nn.2709 and reduces amyloid-beta in a mouse model of Alzheimer’s disease.
51. Griffin WST, Sheng JG, Royston MC, Gentleman SM, McKenzie Alzheimers Dement 11(7):P845. https://doi.org/10.1016/j.jalz.
JE, Graham DI, Roberts GW, Mrak RE (2006) Glial-neuronal in- 2015.06.1880
teractions in Alzheimer’s disease: the potential role of a ‘cytokine
64. Papay R, Gaivin R, Jha A, McCune DF, McGrath JC, Rodrigo MC,
cycle’ in disease progression. Brain Pathol 8(1):65–72. https://doi.
Simpson PC, Doze VA et al (2006) Localization of the mouse
org/10.1111/j.1750-3639.1998.tb00136.x
alpha1A-adrenergic receptor (AR) in the brain: alpha1AAR is
52. Brosseron F, Krauthausen M, Kummer M, Heneka MT (2014)
expressed in neurons, GABAergic interneurons, and NG2 oligo-
Body fluid cytokine levels in mild cognitive impairment and
dendrocyte progenitors. J Comp Neurol 497(2):209–222. https://
Alzheimer’s disease: a comparative overview. Mol Neurobiol
doi.org/10.1002/cne.20992
50(2):534–544. https://doi.org/10.1007/s12035-014-8657-1
53. Swardfager W, Lanctot K, Rothenburg L, Wong A, Cappell J, 65. Hong CJ, Wang YC, Liu TY, Liu HC, Tsai SJ (2001) A study of
Herrmann N (2010) A meta-analysis of cytokines in Alzheimer’s alpha-adrenoceptor gene polymorphisms and Alzheimer disease. J
disease. Biol Psychiatry 68(10):930–941. https://doi.org/10.1016/j. Neural Transm (Vienna) 108(4):445–450. https://doi.org/10.1007/
biopsych.2010.06.012 s007020170065
54. Magaki S, Mueller C, Dickson C, Kirsch W (2007) Increased pro- 66. Doze VA, Papay RS, Goldenstein BL, Gupta MK, Collette KM,
duction of inflammatory cytokines in mild cognitive impairment. Nelson BW, Lyons MJ, Davis BA et al (2011) Long-term alpha1A-
Exp Gerontol 42(3):233–240. https://doi.org/10.1016/j.exger.2006. adrenergic receptor stimulation improves synaptic plasticity, cogni-
09.015 tive function, mood, and longevity. Mol Pharmacol 80(4):747–758.
55. Samidurai M, Ramasamy VS, Jo J (2018) Beta-amyloid inhibits https://doi.org/10.1124/mol.111.073734
hippocampal LTP through TNFR/IKK/NF-kappaB pathway. 67. Wang LY, Shofer JB, Rohde K, Hart KL, Hoff DJ, McFall YH,
Neurol Res 40(4):268–276. https://doi.org/10.1080/01616412. Raskind MA, Peskind ER (2009) Prazosin for the treatment of
2018.1436872 behavioral symptoms in patients with Alzheimer disease with agi-
56. Wang Q, Wu J, Rowan MJ, Anwyl R (2005) Beta-amyloid inhibi- tation and aggression. Am J Geriatr Psychiatry 17(9):744–751.
tion of long-term potentiation is mediated via tumor necrosis factor. https://doi.org/10.1097/JGP.0b013e3181ab8c61
Eur J Neurosci 22(11):2827–2832. https://doi.org/10.1111/j.1460- 68. Koenig RT, Dickman JR, Wise ML, Ji LL (2011) Avenanthramides
9568.2005.04457.x are bioavailable and accumulate in hepatic, cardiac, and skeletal
57. Doost Mohammadpour J, Hosseinmardi N, Janahmadi M, muscle tissue following oral gavage in rats. J Agric Food Chem
Fathollahi Y, Motamedi F, Rohampour K (2015) Non-selective 59(12):6438–6443. https://doi.org/10.1021/jf2002427
NSAIDs improve the amyloid-beta-mediated suppression of mem- 69. Chen CY, Milbury PE, Collins FW, Blumberg JB (2007)
ory and synaptic plasticity. Pharmacol Biochem Behav 132:33–41. Avenanthramides are bioavailable and have antioxidant activity in
https://doi.org/10.1016/j.pbb.2015.02.012 humans after acute consumption of an enriched mixture from oats.
58. Kotilinek LA, Westerman MA, Wang Q, Panizzon K, Lim GP, J Nutr 137(6):1375–1382
Simonyi A, Lesne S, Falinska A et al (2008) Cyclooxygenase-2
inhibition improves amyloid-β-mediated suppression of memory Publisher’s Note Springer Nature remains neutral with regard to jurisdic-
and synaptic plasticity. Brain 131(3):651–664. https://doi.org/10. tional claims in published maps and institutional affiliations.
1093/brain/awn008
59. Chen C-C, Lin J-T, Cheng Y-F, Kuo C-Y, Huang C-F, Kao S-H,
Liang Y-J, Cheng C-Y et al (2014) Amelioration of LPS-induced

View publication stats

You might also like