You are on page 1of 15

Hindawi Publishing Corporation

Evidence-Based Complementary and Alternative Medicine


Volume 2016, Article ID 5850739, 14 pages
http://dx.doi.org/10.1155/2016/5850739

Research Article
Xiao Yao San against Corticosterone-Induced Stress
Injury via Upregulating Glucocorticoid Receptor Reaction
Element Transcriptional Activity

Guoping Cao,1 Shenglan Gong,2 Fengxue Zhang,3 and Wenjun Fu3


1
Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
2
Shenzhen Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 51800, China
3
School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China

Correspondence should be addressed to Wenjun Fu; fuwenjun201511@163.com

Received 15 April 2016; Revised 28 June 2016; Accepted 4 September 2016

Academic Editor: Abir El-Alfy

Copyright © 2016 Guoping Cao et al. This is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Previous studies have revealed that uncontrollable stress can impair the synaptic plasticity and firing property of hippocampal
neurons, which influenced various hippocampal-dependent tasks including memory, cognition, behavior, and mood. In this work,
we had investigated the effects and mechanisms of the Chinese herbal medicine Xiao Yao San (XYS) against corticosterone-induced
stress injury in primary hippocampal neurons (PHN) cells. We found that XYS and RU38486 could increase cell viabilities and
decrease cell apoptosis by MTT, immunofluorescence, and flow cytometry assays. In addition, we observed that XYS notably
inhibited the nuclear translocation of GR and upregulated the mRNA and protein expressions levels of Caveolin-1, GR, BDNF,
TrkB, and FKBP4. However, XYS downregulated the FKBP51 expressions. Furthermore, the results of the electrophoretic mobility
shift assay (EMSA) and double luciferase reporter gene detection indicated that FKBP4 promotes the transcriptional activity of
GR reaction element (GRE) by binding with GR, and FKBP51 processed the opposite action. The in vivo experiment also proved
the functions of XYS. These results suggested that XYS showed an efficient neuroprotection against corticosterone-induced stress
injury in PHN cells by upregulating GRE transcriptional activity, which should be developed as a potential candidate for treating
stress injury in the future.

1. Introduction plasticity and firing properties of hippocampal neurons [8].


Therefore, investigations of the underlying mechanisms of
In the last decade, hippocampal neurogenesis in the mature stress cascades are urgently needed for developing effective
brain has attracted considerable attention [1–3]. Up to now, therapies of hippocampal dysfunction.
stress is the earliest identified and the most profound factor The hypothalamic-pituitary-adrenal (HPA) axis and its
that can affect adult neurogenesis both positively and nega- end-effect neuroendocrine hormones, namely, glucocorti-
tively [4, 5]. In brain, hippocampal area was crucially involved coids, are mediators of the myriad stress effects on the
in memory and emotional information processing, which hippocampus [9]. Briefly, HPA axis is activated after uncon-
affects the abilities of generating and differentiating into trollable stress accumulating in brain either internally or
multiple neural cell lineages [6]. A large number of evidences externally, and then glucocorticoid receptors (GRs) are
have suggested that hippocampal neurogenesis contributes to triggered and expressed densely in the hippocampus [10].
some hippocampus-dependent tasks, such as cognition and Corticosterone, the major glucocorticoid, exerts powerful
mood [7]. Uncontrollable stress exerts long-lasting changes feedback effects on the HPA axis. Accumulating reports have
in the brain, and it leads to behavioral, emotional, and proved that nerve cells under persistent exposure to high
cognitive modifications including Alzheimer’s disease (AD), concentration of corticosterone can notably induce DNA
anxiety, and major depression through altering the synaptic damage, some protein activation, and cells apoptosis [11].
2 Evidence-Based Complementary and Alternative Medicine

GR is the main mediator of the stress response in neu- Ltd. (China). Bicinchoninic acid (BCA) Protein Assay Kit,
ronal progenitor cells and hippocampal neurogenesis [12]. bovine serum albumin (BSA), and Firefly Luciferase Reporter
The expression level of GR directly regulates the excitation- Gene Assay Kit were purchased from Beyotime Institute of
inhibition balance, which is key for the positioning and Biotechnology (Shanghai, China). Cell Cycle and Apopto-
functional integration of newborn neurons and may con- sis Analysis Kits were all purchased from Beyotime Insti-
tribute to the chronification of paroxysmal brain diseases tute of Biotechnology (Shanghai, China). Corticosterone,
[13]. The hippocampal area expresses GR in a high level, so 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bro-
it is sensitive to the fluctuating levels of corticosterone in mide (MTT), and 4,6-diamidino-2-phenylindole (DAPI)
particular [14]. were provided by Sigma (St. Louis, MO, USA). Anti-
Traditional Chinese medicines (TCMs) have been used bodies against microtubule-associated protein 2 (MAP-2),
in China for thousands of years. Due to the advantage of Caveolin-1, GR, BDNF, TrkB, FKBP4, FKBP51, and GAPDH-
high efficiency and low toxicity, TCMs have attracted great conjugated affinipure goat anti-rabbit IgG and horseradish
interest in recent years [15]. Some active natural components peroxidase-conjugated goat anti-rabbit IgG were supplied
from TCMs, including icariin from Epimedium brevicornum by Abcam (Cambridge, UK). Electrophoretic mobility shift
Maxim, baicalin from Scutellaria baicalensis Georgi, and assay (EMSA) kit was obtained from Invitrogen (Carlsbad,
Shugan Lifei Recipe (SGLFR), have already been proved USA).
with excellent activities against corticosterone-induced stress
injury [16, 17]. Icariin was able to prevent corticosterone- 2.2. High Performance Liquid Chromatography (HPLC) Anal-
induced hypothalamic cell apoptosis via activation of the ysis of XYS. HPLC analysis of XYS was performed on
PI3K/Akt pathway [18]. Baicalin exhibits a beneficial effect on Agilent 1100 HPLC system (Agilent Co., USA) using C18
adult neurogenesis [19]. SGLFR is beneficial to asthma under column COSMOSIL (4.6 mm × 250 mm ID; particle size
stress in rats by regulating the hyperfunction of HPA axis and 5 𝜇m, Japan). The mobile phases comprised eluent A (0.1%
the disorder of immunosystem [20]. Therefore, it is reason- trifluoroacetic acid) and eluent B (acetonitrile). The gradient
able to exploit effective herbal products for the treatment of flow was as follows: 0.00–15.00 min, 17% B; 15–40 min, 17%–
corticosterone-induced stress injury in primary hippocampal 45% B; 40–60 min, 45%–60% B; 60–70 min, 60%–80% B;
neurons (PHN) cells. Xiao Yao San (XYS) is a Chinese herbal 70–85 min, 80% B. The analysis was performed at a flow
formula that has been widely used to treat mental disorders rate of 1.0 mL/min with DAD detection at 235 nm. The
for thousands of years in China [21]. It was reported that XYS injection volume was 20 𝜇L. Furthermore, the professional
had anxiolytic-like effects on the elevated plus-maze (EPM) software Similarity Evaluation System for Chromatographic
test by decreasing brain-derived neurotrophic factor (BDNF) Fingerprint of TCM (version 2004A), which was used for
and increasing tyrosine hydroxylase (TrkB) [22]. In addition, data analysis, was recommended by the State Food and Drug
previous research has proved that treatments with XYS could Administration. We calculated the correlation coefficients of
cause significant reversal of stress-induced deficits in learning entire chromatographic profiles of samples for the simulated
and memory in a spatial memory task [23]. However, no mean chromatogram.
papers have been published to report the effects of XYS
against corticosterone-induced stress injury in PHN cells. 2.3. Animals. Male Wistar rats (180–220 g) were purchased
Therefore, in this paper, we decided to investigate whether from the Experimental Animal Center in Guangzhou Uni-
XYS ameliorated corticosterone-induced stress injury in versity of Chinese Medicine. All experimental procedures
PHN cells and whether the change of GR level was involved were approved by the Animal Care and Use Committee of
in underlying molecular mechanism. the Experimental Animal Center in Guangzhou University
of Chinese Medicine and performed in strict accordance
2. Materials and Methods with the PR China Legislation Regarding the Use and Care
of Laboratory Animals. The rats were allowed to adapt to
2.1. Chemicals and Materials. Compounds including paeoni- the new environment for one week before the experiments,
florin, liquiritin, ferulic acid, and glycyrrhizic acid were which were housed in a room under 12 h light/dark cycles, a
purchased from Chengdu Pufeide Biological Technology Co., relative humidity of 60 ± 10%, and a controlled temperature
Ltd. (Chengdu, China). RU-38486 used for the positive drug of 22 ± 3∘ C. The rats were allowed ad libitum access to water
was obtained from the Medchem Express (Princeton, USA). and a standard pellet diet throughout the experiment.
Corticosterone is one kind of adrenal cortical hormone,
which has the strongest effect on carbohydrate metabolism. 2.4. Preparation of XYS Cerebrospinal Fluid. The herbal for-
As a potent GR antagonist, RU-38486 (mifepristone) can mula of XYS comprises the following traditional Chinese
decrease the injury caused by high level of adrenal corti- medicines: Angelica sinensis (Oliv.) Diels, Paeonia lactiflora
cal hormone [24]. Fetal bovine serum (FBS), penicillin, and Pall., Bupleurum chinense DC., Atractylodes macrocephala
streptomycin were obtained from Hyclone Laboratories, Koidz, Glycyrrhiza uralensis Fisch., Poria cocos (Schw.) Wolf,
Inc. (Massachusetts, USA). Dulbecco’s minimum essential Zingiber officinale Rosc., and Mentha haplocalyx Briq., and
medium (DMEM), Neurobasal-A medium, B27 NeuroMix, the ratio is 6 : 6 : 6 : 6 : 6 : 2 : 2 : 3 [24]. The herbal formula was
cytosine arabinoside, poly-L-lysine, and trypsin were pur- immersed in 10 times volume of distilled water and boiled
chased from Gibco (California, USA). Tissue Protein Ex- at 80∘ C for 1 h. The water extracts were collected and then
traction Kit was purchased from KEYGEN Biotech. Co., concentrated by rotary evaporator. The rats were randomly
Evidence-Based Complementary and Alternative Medicine 3

divided into four groups: animals in Control groups were which they were administered corticosterone plus RU-38486
treated with physiological saline; XYS groups were treated (2 𝜇mol/L 0.1% DMSO). The PHN cells in different groups
with 20.106 (High), 10.053 (Medium), and 5.256 (Low) were incubated for 7 days. Finally, the cell viabilities of
g/kg of XYS water extracts. Physiological saline and XYS hippocampal neurons were detected according to the MTT
water extracts were administered intragastrically (i.g.) twice assay. For examination of morphological changes, the cells
daily for five consecutive days. Rats were anesthetized using were photographed by using an inverted microscope (Nikon,
intraperitoneal (i.p.) chloral hydrate (350 mg/kg) 1 hour after Japan).
the last administration [25]. The skin, fascia, and muscle in
the back side of the neck were exposed successively, subse- 2.8. MAP-2 Detection in PHN Cells. The PHN cells were
quently piercing the ring pillow membrane and extracting fixed using methanol for 30 min and permeabilized using
cerebrospinal fluid (CSF) from the cisterna magna. The CSF 0.3% Triton X-100 for 10 min after XYS pretreatment and
was centrifuged (13,000 r/min) at 4∘ C for 10 min and then corticosterone injury. The cells were incubated with 5% goat
sterilized by 0.22 𝜇m microporous membrane. serum albumin for 1 h at 37∘ C to block nonspecific binding.
The cells were incubated with the anti-MAP-2 antibody
2.5. PHN Cells Isolation and Culture. PHN cells were isolated (1 : 100 dilution) for 16 h at 4∘ C and then incubated with Alexa
from fetal Sprague-Dawley (SD) rats at embryonic day Fluor 488 Goat Anti-Mouse IgG (H+L) (Proteintech Group,
18 (E18) based on the previously described method [26]. Chicago, USA) for 1 h at 37∘ C. The images were obtained
Briefly, the embryos were removed by caesarian section and using a fluorescence microscope (OLYMPUS, Japan).
hippocampi were rapidly isolated under sterile conditions,
cut into 1 mm pieces, and digested with 0.125% trypsin at 2.9. Cell Survival Rate and Cycle Analysis. The PHN cells
37∘ C for 15 min. Trypsin activity was then stopped by planting were incubated in 6-well plates and treated by XYS and
DMEM containing 10% FBS, 100 U/mL of penicillin, and corticosterone for 7 days. Then, the cells were collected,
100 U/mL streptomycin. The cells (5×105 cells/mL) were then washed three with PBS, centrifuged at 600 ×g for 5 min, and
planted on poly-l-lysine coated plastic dishes in a constant fixed with 75% ethanol at 4∘ C for 24 h. After that, the cells
temperature and humidity incubator. 24 hours after plant- were stained with PI staining solution for 30 min at 37∘ C
ing, the DMEM was replaced with Neurobasal-A medium and then analyzed by flow cytometry (FACSCalibur Becton
containing 2% B27 NeuroMix, 100 U/mL of penicillin, and Dickson, San Diego, CA, USA).
100 U/mL streptomycin. Cultures were treated with cytosine
arabinoside (2 𝜇g/mL) to inhibit glial proliferation after 48 2.10. Cell Apoptosis Analysis. For detecting the PHN cells
hours. The hippocampal neurons were used for experiments apoptosis, Annexin and PI staining solution were added
after 7–10 days of culture. into cell suspension medium for 20 min in 37∘ C prior to
assessment by flow cytometry.
2.6. Corticosterone Injury. The PHN cells were incubated in
corticosterone (Cort) at different concentrations (1280, 640, 2.11. Immunofluorescent Examination. The PHN cells were
320, 160, 80, 40, 20, 10, and 0 𝜇mol/L) for 7 days. The cell treated with methanol for 30 min and 0.3% Triton X-100 for
viabilities were detected according to the MTT assay. Briefly, 10 min. The cells were then incubated in a moist box overnight
10 𝜇L MTT was added to each well at 37∘ C for 1 h, and at 4∘ C with the rabbit anti-GR, at a 1 : 100 dilution after the
the formative formazan crystals were dissolved by DMSO blocking of nonspecific protein binding by 5% goat serum
(100 𝜇L) at 37∘ C for 4 h. The absorbance of samples then albumin, then incubated with Alexa Fluor 488 Goat Anti-
was measured at 492 nm using a spectrophotometer (Huayi Mouse IgG (H+L) (Proteintech Group, Chicago, USA) for 1 h
Science and Technology Co. Ltd., China). at 37∘ C, and counterstained by DAPI (1 𝜇g/mL) for 15 min.
Images of the cells were obtained using a laser scanning
2.7. Cell Viabilities and Morphological Changes Assay. We confocal microscope (Leica, TCS SP5, Germany).
used different concentrations of corticosterone pretreating
PHN cells to mimic stress-induced damage and found the 2.12. Quantitative Real-Time PCR Assay. The total RNA
IC50 of corticosterone was 223 𝜇mol/L. Thus, we chose samples of PHN cells were extracted by using RNAiso Plus
223 𝜇mol/L as the corticosterone dose. The PHN cells were reagent (TaKaRa Biotechnology Co., Ltd., China) following
allocated to seven groups: (1) the Control group, in which the manufacturer’s protocol. The purity of the extracted
they were treated by 0.1% DMSO; (2) the CSF group, in RNA was determined; then reverse transcription polymerase
which they were treated by control cerebrospinal fluid; (3) chain reaction (RT-PCR) was performed using PrimeScript
the Cort group, in which they received administration of RT reagent kit (TaKaRa Biotechnology Co., Ltd., China)
corticosterone at a dose of 223 𝜇mol/L; (4) the Cort + following the manufacturer’s instructions with a TC-512 PCR
Low group, in which they received administration of both system (TECHNE, UK). Subsequently, the levels of mRNA
corticosterone and XYS cerebrospinal fluid (low dose); (5) the expression were quantified by real-time PCR with SYBR
Cort + Medium group, in which they received administration PremixEx TaqII (Tli RNaseH Plus) (TaKaRa Biotechnology
of both corticosterone and XYS cerebrospinal fluid (medium Co., Ltd., China) and ABI 7500 Real-Time PCR System
dose); (6) the Cort + High group, in which they received (Applied Biosystems, USA). The sequences of the primers
administration of both corticosterone and XYS cerebrospinal are shown in Table 1. A no-template control was analyzed in
fluid (high dose); and (7) the Cort + RU-38486 group, in parallel for each gene, and GAPDH gene was selected as the
4 Evidence-Based Complementary and Alternative Medicine

Table 1: The primer sequences used for real-time PCR assay in rats.

Gene Full name Primer (5󸀠 -3󸀠 )


F: GACTTTGAAGATGTGATTGC
Caveolin-1 Caveolin-1
R: AGATGGAATAGACACGGCTG
F: TGTATCCCACAGACCAAAGCA
GR Glucocorticoid receptor
R: AATCCTCATTCGTGTTCCCTTC
F: ATCCCATGGGTTACACGAAGGAAG
BDNF Brain-derived neurotrophic factor
R: AGTAAGGGCCCGAACATACGATTG
F: GATCCTGGTGGCCGTGAA
TrkB Neurotrophic tyrosine kinase, receptor, type 2
R: GCTTCGCGATGAAAGTCCTT
F: AGATGACAGCCGAGGAGATG
FKBP4 FK506 binding protein 4
R: AATTTGTCCTTGCGATCCAG
F: GAGCCGTTTGTCTTTAGCCTTG
FKBP51 FK506 binding protein 4
R: GCCAGCAGAGCCGTAAGC
F: TGCACCACCAACTGCTTAG
GAPDH Glyceraldehyde-3-phosphate dehydrogenase
R: GATGCAGGGATGATGTTC

house-keeping gene in our study. Eventually, the unknown and then transfected into PHN cells. After the cells were
template was calculated through the standard curve for cultured for 21 h, FKBP4 (10 𝜇g/L) or FKBP51 (10 𝜇g/L) was
quantitative analysis. Briefly, all gene expression levels were added for 1 h culturation. Finally, the cell lysis solutions
normalized to GAPDH, and the fold changes between the were collected and the effects of FKBP4 and FKBP51 against
different groups were calculated using the 2−ΔΔCt method. GRE transcriptional activity were determined using the single
photon fluorescence detector (Promega, USA).
2.13. Western Blotting Assay. The PHN cells were plated in 6-
well plates and treated; then the cell proteins were collected 2.16. In Vivo Experiments. The rats were randomly allocated
using appropriate cold lysis buffer containing 1 mM phenyl- to six groups: (1) the Control group (𝑛 = 6), in which they
methylsulfonyl fluoride (PMSF) based on the manufacturer’s were given an equal volume of 0.5% CMC-Na; (2) the Cort
instructions. Finally, the protein samples were loaded onto group (𝑛 = 6), in which they received administration of
the SDS-PAGE gel (10%–15%), separated electrophoretically, corticosterone at a dose of 40 mg/kg per day; (3) the Cort
and transferred onto a PVDF membrane (Millipore, USA). + Low group (𝑛 = 6), in which they were given both
After blocking nonspecific binding sites for 1 h with 5%
corticosterone and XYS water extracts at 5.256 g/kg per day;
dried skim milk in TTBS at 37∘ C, the membrane was
(4) the Cort + Medium group (𝑛 = 6), in which both
individually incubated for overnight at 4∘ C with primary
corticosterone and XYS water extracts at 10.053 g/kg per day
anti-Caveolin-1, anti-GR, anti-BDNF, anti-TrkB, anti-FKBP4,
and anti-FKBP51. Then the membrane was incubated at room were administered to them; (5) the Cort + High group (𝑛 =
temperature for 2 h with horseradish peroxidase-conjugated 6), in which both corticosterone and XYS water extracts at
antibodies at a 1 : 5,000 dilution. Protein expression was 20.106 g/kg per day were administered to them; and (6) the
detected by an enhanced chemiluminescence (ECL) method Cort + RU-38486 group (𝑛 = 6), in which they received
and imaged using ChemiDoc XRS (BIO-RAD, USA). To administration of corticosterone plus RU-38486 (20 mg/kg
eliminate the variations of protein expression, the data were per day). Corticosterone was subcutaneously injected once
adjusted to correspond to internal reference expression (IOD a day, RU-38486 was subcutaneously injected once a day,
value of target protein versus IOD of corresponding internal and XYS (suspended in 0.5% CMC-Na) was administered
reference). orally via a gastric tube once a day, with all drugs being given
to the animals for 21 days. Finally, rats were anesthetized
2.14. Electrophoretic Mobility Shift Assay (EMSA). For deter- using intraperitoneal (i.p.) chloral hydrate (350 mg/kg), and
mining the bonding abilities of GR with FKBP4 and FKBP51, hippocampus was quickly dissected out on a cold plate
EMSA was carried out. 1 nM Cy5-labeled DNA, 10 𝜇mol/L and immediately frozen in liquid nitrogen and stored at
FKBP4, and 10 𝜇mol/L FKBP51 were incubated in 10 mM −80∘ C until assayed. Caveolin-1, GR, BDNF, TrkB, FKBP4,
Tris-HCl (pH = 8.0) buffer including 10 𝜇mol/L Histone H1 and FKBP51 mRNA levels of tissue were detected as above
and 10 𝜇mol/L BSA at 4∘ C for 1 h. Reactions were run on 20% method.
Native-PAGE with 1x Tris-glycine buffer (pH = 8.3) at 4∘ C
under 200 V voltage for 15 min. The gels were scanned using 2.17. Statistical Analysis. All the data were analyzed using
ChemiDoc XRS (BIO-RAD, USA). statistical software SPSS 18.0 and expressed as means ± SEM.
Differences among groups were determined using the one-
2.15. Dual-Luciferase Reporter Assay. The GRE-tk-Luc and way ANOVA, followed by a post hoc Newman-Keuls test.
pRL-SV40 plasmids were mixed in the Eppendorf tube Comparisons between two groups were performed using
Evidence-Based Complementary and Alternative Medicine 5

100 1.2

80

Absorbance (490 nm)


Inhibition rate (%) 60
0.8 ∗∗
∗∗
∗∗ ∗∗
40 ##
0.4
IC50 = 223 𝜇mol/L
20

0 0.0
0 320 640 960 1280

Control

CSF

Cort

Medium

High

RU-38486
Low
Cort (𝜇mol/L)

(a) (b)
Control CSF Cort Low Medium High RU-38486
Morphology
MAP-2

(c)
Figure 1: The effects of XYS on the viabilities of corticosterone treated PHN cells. (a) The inhibition rate of corticosterone against PHN cells.
(b) The effect of XYS on the cell viabilities of PHN cells after corticosterone-induced stress injury. (c) The effects of XYS on the morphological
changes and MAP-2-immunopositive neurons (green) of PHN cells (200x magnification) after corticosterone-induced stress injury (𝑛 = 3;
## ∗∗
𝑃 < 0.01 versus CSF; 𝑃 < 0.01 versus Cort).

unpaired Student’s 𝑡-test. 𝑃 < 0.05 and 𝑃 < 0.01 were con- viabilities. The MTT results also indicated that the viability
sidered to be significant. of PHN cells notably decreased after corticosterone treatment
compared with CSF group (𝑃 < 0.01). However, the cell
viability was significantly restored by XYS cerebrospinal
3. Results fluid of different concentrations, low (5.256 g/kg), middle
3.1. High Performance Liquid Chromatography (HPLC) Anal- (10.053 g/kg), and high (20.106 g/kg), and the positive drug
ysis of XYS. Excellent separations were achieved and the RU-38486 (2 𝜇mol/L). Furthermore, Figure 1(c) showed that
chromatograms are shown in Supplementary Figures 1(a) PHN cells were exposed to corticosterone exhibiting char-
and 1(b) in Supplementary Material available online at acteristic of neurodegeneration including a loss of synapses
http://dx.doi.org/10.1155/2016/5850739, in which peaks 1– by using the inverted microscope. However, XYS could
4 represent paeoniflorin, liquiritin, ferulic acid, and gly- decrease the synapses injury. In addition, the control and
cyrrhizic acid, respectively. And the results also showed CSF treated cells exhibited higher fluorescence intensity of
that the contents of paeoniflorin, liquiritin, ferulic acid, and MAP-2 than that of the Cort group by the observation using
glycyrrhizic acid in total of XYS crude drugs were 21.53 mg/g, fluorescence microscope. XYS pretreatment and RU-38486
5.32 mg/g, 4.56 mg/g, and 6.41 mg/g, respectively. Similarity increased the fluorescence intensity (Figure 1(c)). However,
analysis of chromatographic XYS samples was shown in there was no remarkable difference between CSF and Control
Supplementary Figure 2. The results showed that all the group, which showed that the CSF had no significant effect on
similarity values of the 9 samples were greater than 0.95. PHN cells morphology.

3.2. The Effects of XYS on the Viabilities of Corticosterone 3.3. The Effects of XYS on Cell Survival Rate, Cycle, and
Treated PHN Cells. We used different concentrations of Apoptosis. As shown in Figure 2(a), the first peak represented
corticosterone pretreating PHN cells to mimic stress-induced live cells, and the second peak represented the dead cell. The
damage and found the IC50 of corticosterone was 223 𝜇mol/L results of flow cytometry indicated that there was no signif-
(Figure 1(a)). As shown in Figure 1(b), there was no remark- icant difference between CSF and Control group. However,
able difference between CSF and Control group, which corticosterone significantly decreased the cell survival rate,
showed that the CSF had no significant effect on PHN cells while XYS and RU-38486 could raise the rate compared to
6 Evidence-Based Complementary and Alternative Medicine

Control CSF Cort Low Medium High RU-38486

Counts

0 0 0 0 0 0 0
PI intensity PI intensity PI intensity PI intensity PI intensity PI intensity PI intensity
(a)
Control CSF Cort Low Medium High RU-38486
1200

900
Counts

600

300

0
0 200 400 0 200 400 0 200 400 0 200 400 0 200 400 0 200 400 0 200 400
DNA content DNA content DNA content DNA content DNA content DNA content DNA content
(b)
80

##
Percentage of cells (%)

60 ∗
∗∗
∗∗
40
∗∗ #
∗∗ ∗ ∗
∗∗ ∗ ∗
20
∗∗
##
0
G0/G1 S G2/M

Control Medium
CSF High
Cort RU-38486
Low
(c)

Figure 2: The effects of XYS on cell survival rate and cycle. (a) The effect of XYS on cell survival rate using flow cytometry detection. (b) The
effect of XYS on cell cycle using flow cytometry detection. (c) Data analysis of the effect of XYS on cell cycle (𝑛 = 3; # 𝑃 < 0.05 versus CSF;
## ∗ ∗∗
𝑃 < 0.01 versus CSF; 𝑃 < 0.05 versus Cort; 𝑃 < 0.01 versus Cort).

the Cort group. The results of Figures 2(b) and 2(c) showed corticosterone notably increased the cell apoptosis; however,
that the content of cells DNA at G0/G1 and G2/M phase XYS and RU-38486 could decrease the rate of apoptotic
was increased, whereas the cells DNA content at S phase cells compared to Cort group. Furthermore, there was no
was decreased after corticosterone treatment. Furthermore, significant difference of the effects produced by CSF and
XYS decreased the cells DNA content at G0/G1 and G2/M control vehicle (𝑃 > 0.05).
phase and increased the cells DNA content at S phase. In the
same way, there was no significant difference of the effects 3.4. The Effect of XYS on GR Nuclear Transfer in PHN Cells.
produced by CSF and control vehicle. As shown in Figures The results of immunofluorescent examination shown in
3(a) and 3(b), the results of flow cytometry showed that Figure 4 indicated that the nuclear to cytosolic translocation
Evidence-Based Complementary and Alternative Medicine 7

Control CSF Cort


104 1.19 2.88 104 6.23 6.59 104 2.87 30.60
FL3-H:: 7-AAD

FL3-H:: 7-AAD
FL3-H:: 7-AAD
3 3 3
10 10 10

102 102 102

101 101 101


91.27 4.66 84.88 2.30 41.65 24.88
100 100 100
0 1 2 3 4 0 1 2 3 4 0 1 2 3
10 10 10 10 10 10 10 10 10 10 10 10 10 10 104
FL4-H:: Annexin APC FL4-H:: Annexin APC FL4-H:: Annexin APC
Low Medium High
104 4.32% 28.9% 104 2.49 15.10 104 4.38% 12.3%
FL3-H:: 7-AAD

FL3-H:: 7-AAD
FL3-H:: 7-AAD

3 3 3
10 10 10

102 102 102

101 101 101


59.4% 7.44% 70.15 12.27 80.2% 3.14%
100 100 100
0 1 2 3
10 0
101
10 2
10 3
10 4
10 10 10 10 104 10 0
101
102
103 104
FL4-H:: Annexin APC FL4-H:: Annexin APC FL4-H:: Annexin APC
RU-38486
104
7.22 8.07
FL3-H:: 7-AAD

3
10

102

101

82.28 2.43
100
0 1 2 3
10 10 10 10 104
FL4-H:: Annexin APC
(a)

80

##
Apoptosis rate (%)

60

40 ∗∗
## ∗∗
## ∗
20 ∗∗∗∗ ∗∗
∗∗ ∗∗
∗∗∗∗∗∗∗∗
0
Early Later Total

Control Medium
CSF High
Cort RU-38486
Low
(b)

Figure 3: The effect of XYS on cell apoptosis using flow cytometry detection. (a) The effect of XYS on cell apoptosis using flow cytometry
detection. (b) Data analysis of the effect of XYS on cell apoptosis (𝑛 = 3; ## 𝑃 < 0.01 versus CSF; ∗ 𝑃 < 0.05 versus Cort; ∗∗ 𝑃 < 0.01 versus
Cort).
8 Evidence-Based Complementary and Alternative Medicine

Control CSF Cort Low Medium High RU-38486


GR
DAPI
Merge

Figure 4: Immunofluorescence of GR nuclear transfer in PHN cells. FITC-labeled secondary antibodies were used (green fluorescence), and
nuclei were stained by DAPI (400x magnification).

of GR (green fluorescence) has no significant difference 3.7. The Effects of FKBP4 and FKBP51 against GR and GRE
between CSF and Control group (𝑃 > 0.05). However, it Transcriptional Activity. Notably, a new slow-migrating band
was increased after corticosterone injury compared to CSF was observed by the EMSA, suggesting that FKBP4-GR
group. Compared with Cort treated group, XYS and RU- DNA complexes were present in the reaction (Figure 7(a)).
38486 decreased the nuclear translocation of GR, and there However, as shown in Figure 7(b), the single band indi-
was no significant difference between the high dose of XYS cated that FKBP51 could not combine with GR DNA. As
and RU-38486. shown in Figure 7(c), FKBP4 could notably increase the
GRE transcriptional activity in a dose-dependent manner
3.5. The Effects of XYS on Caveolin-1, GR, BDNF, TrkB, FKBP4, (𝑃 < 0.05 or 𝑃 < 0.01). Furthermore, the result of Dual-
and FKBP51 mRNA Levels In Vitro. As shown in Figures Luciferase Reporter Assay suggested that FKBP51 signif-
5(a)–5(f), the results of quantitative real-time PCR assay icantly decreased the GRE transcriptional activity when
revealed that Caveolin-1, GR, BDNF, TrkB, and FKBP4 compared with FKBP4 for the same concentration (𝑃 < 0.01)
mRNA levels were significantly decreased and the FKBP51 (Figure 7(d)).
mRNA level was markedly increased in corticosterone treated
hippocampal neurons compared to CSF treated cells. How- 3.8. The Effects of XYS on Caveolin-1, GR, BDNF, TrkB, FKBP4,
ever, there was no significant difference of the effects pro- and FKBP51 mRNA Levels In Vivo. As shown in Figures 8(a)–
duced by CSF and control vehicle (𝑃 > 0.05). Furthermore, in 8(f), the results of quantitative real-time PCR assay revealed
XYS and RU-38486 group, the Caveolin-1, GR, BDNF, TrkB, that Caveolin-1, GR, BDNF, TrkB, and FKBP4 mRNA levels
and FKBP4 mRNA levels were significantly increased, and were significantly decreased and the FKBP51 mRNA level
the FKBP51 mRNA level was notably decreased compared to was markedly increased in Cort treated rats compared to
Cort group. There was no significant difference of the effects control rats. In addition, XYS significantly upregulated the
produced by high dose of XYS and RU-38486 (𝑃 < 0.05). Caveolin-1, GR, BDNF, TrkB, and FKBP4 mRNA levels and
downregulated the FKBP51 mRNA level compared to Cort
3.6. The Effects of XYS on Caveolin-1, GR, BDNF, TrkB, FKBP4, group. There was no significant difference of the effects
and FKBP51 Protein Levels. As shown in Figures 6(a)–6(f), produced by high dose of XYS and RU-38486 (𝑃 < 0.05).
the Western blotting results revealed that the protein levels
of Caveolin-1, GR, BDNF, TrkB, and FKBP4 were notably 4. Discussion
downregulated, and the FKBP51 protein expression was
significantly upregulated after treatment with corticosterone A number of neurodegenerative and neurodevelopmental
in hippocampal neurons when compared with the CSF group. diseases involving cognitive injuries are probably linked to
Furthermore, there was no significant difference of the effects hippocampal dysfunction, also leading to dysregulation of
produced by CSF and control vehicle (𝑃 > 0.05). However, neurogenesis [25, 27]. Plenty of researches about hippocam-
compared to the Cort group, the protein levels of Caveolin- pal dysfunction have been carried out in recent decades,
1, GR, BDNF, TrkB, FKBP4, and FKBP51 were reversed by but the pathogenesis of hippocampal dysfunction has not
XYS and RU-38486. Furthermore, there was no significant been fully clarified, and few medicines are available. Previous
difference of the effects produced by high dose of XYS and research has shown that stress is an integral part of the
RU-38486 (𝑃 < 0.05). pathogenesis of hippocampal dysfunction [28]. Briefly, stress
Evidence-Based Complementary and Alternative Medicine 9

Caveolin-1 GR
1.2 1.5
∗∗ ∗∗ ∗∗

Relative mRNA expression

Relative mRNA expression


0.9 ∗∗
∗∗ 1.0
∗∗
0.6

0.5
0.3 ##
##

0.0 0.0
Control

CSF

Cort

Medium

High

RU-38486

Control

CSF

Cort

Medium

High

RU-38486
Low

Low
(a) (b)

1.5 BDNF 1.2 TrkB

∗∗
Relative mRNA expression

Relative mRNA expression


∗∗
∗∗
∗∗
1.0 0.8
∗∗
##
0.5 0.4
##

0.0 0.0
Control

CSF

Cort

Medium

High

RU-38486

Control

CSF

Cort

Medium

High

RU-38486
Low

Low
(c) (d)

1.2 FKBP4 ∗∗ 3.5 FKBP51


∗∗
Relative mRNA expression

Relative mRNA expression

##
2.8
∗∗
0.8
2.1
∗∗ ∗∗
1.4 ∗∗
∗∗
0.4
##
0.7

0.0 0.0
Control

CSF

Cort

Medium

High

RU-38486

Control

CSF

Cort

Medium

High

RU-38486
Low

Low

(e) (f)
Figure 5: The effects of XYS on the gene levels of Caveolin-1 (a), GR (b), BDNF (c), TrkB (d), FKBP4 (e), and FKBP51 (f) in PHN cells after
corticosterone-induced stress injury (𝑛 = 3; ## 𝑃 < 0.01 versus CSF; ∗ 𝑃 < 0.05 versus Cort; ∗∗ 𝑃 < 0.01 versus Cort).

triggers important neuroendocrine responses including the In this paper, we used different concentrations of cor-
rapid activation of HPA axis and sympathoadrenal system, ticosterone to pretreat PHN cells to mimic stress-induced
which leads to the release of catecholamines, adrenocorti- damage and found that the IC50 of corticosterone was
cotropic hormone, and glucocorticoids [29]. Moreover, hip- 223 𝜇mol/L. There is accumulating evidence that corticos-
pocampal dysfunction is induced by glucocorticoids, which terone could induce the apoptosis of PHN cells. XYS is a Chi-
are negatively regulated via GR [30]. GR especially plays an nese herbal formula that has been widely used to treat mental
important role in the process of corticosteroids induced stress disorders for thousands of years in China, and we found
[28]. Therefore, the decreasing of stress through upregulation that XYS mainly included paeoniflorin, liquiritin, ferulic acid,
of GRs represents a potential therapeutic approach to allevi- and glycyrrhizic acid by the HPLC analysis. Further studies
ating hippocampal dysfunction. proved that treatment of XYS presented neuroprotective
10

Relative protein expression Relative protein expression


Relative protein expression

0.0
0.4
0.8
1.2
1.6

0.0
0.4
0.8
1.2
GAPDH
BDNF

GAPDH
FKBP4
0.0
0.4
0.8
1.2
GAPDH
Caveolin-1

Control Control Control Control Control Control

CSF CSF CSF CSF CSF CSF


Cort

##

##
Cort Cort Cort Cort

##
Cort

(c)
(a)

(e)
Low Low

∗∗
Low Low Low Low

Medium Medium

∗∗
Medium Medium Medium
∗∗

Medium

High High

∗∗

∗∗
High High High
∗∗

High

RU-38486 RU-38486

∗∗

∗∗
RU-38486 RU-38486 RU-38486
∗∗

RU-38486

Relative protein expression Relative protein expression


Relative protein expression

0
1
2
3
0.0
0.4
0.8
1.2
0.0
0.4
0.8
1.2

GAPDH
TrkB

GAPDH
FKBP51
GR
GAPDH

Control Control Control Control Control Control

CSF CSF CSF CSF CSF CSF

Cort

##
Cort Cort

##
Cort Cort Cort
##

(f)
(d)
(b)


Low Low Low ∗∗ Low Low
∗∗

Low

Medium Medium

∗∗
∗∗

Medium Medium Medium


∗∗

Medium

High High
∗∗

∗∗
High High High
∗∗

High

after corticosterone-induced stress injury (𝑛 = 3, ## 𝑃 < 0.01 versus CSF; ∗ 𝑃 < 0.05 versus Cort; ∗∗ 𝑃 < 0.01 versus Cort).
RU-38486

∗∗
RU-38486
∗∗

RU-38486 RU-38486
∗∗

RU-38486 RU-38486

Figure 6: The effects of XYS on the protein levels of Caveolin-1 (a), GR (b), BDNF (c), TrkB (d), FKBP4 (e), and FKBP51 (f) in PHN cells
Evidence-Based Complementary and Alternative Medicine
Evidence-Based Complementary and Alternative Medicine 11

FKBP4
FKBP51

GR GR

(a) (b)
120 120
∗∗
90 90
∗∗
Stimulation (%)

Stimulation (%)
∗∗
60 ∗∗ 60
∗∗
∗ ∗∗
30 30 ∗∗
∗∗
0 0
0.00 0.25 0.50 1.00 2.00 3.00 0.00 0.25 0.50 1.00 2.00 3.00
FKBP4 (𝜇g) FKBP51 (𝜇g)
(c) (d)
Figure 7: The bonding abilities of GR with FKBP4 (a) and FKBP51 (b) and the effects of FKBP4 (c) and FKBP51 (d) on GRE transcriptional
activity (𝑛 = 3, ∗ 𝑃 < 0.05 versus FKBP4 or FKBP51 = 0 𝜇g, ∗∗ 𝑃 < 0.01 versus FKBP4 or FKBP51 = 40 𝜇g).

effects against corticosterone-induced insult in PHN cells, GR, the steroid-activated nuclear receptor, translocates to the
which was confirmed by morphological observation and nucleus to bind its target DNA sequences when it is triggered,
MTT assay, as well as the MAP-2 staining that is known to and it is a positive or negative transcription factor to mediate
promote the extension of the dendritic tree and the plasticity mRNA production of genes related to neuronal plasticity,
of neuronal morphogenesis [31]. activation, and metabolism [34, 36]. BDNF is a neurotrophic
Furthermore, the present study demonstrated that cor- factor playing an important role in the central nervous system
ticosterone resulted in reduction of survival rate of PHN (CNS) and can prevent neuronal cells death induced by
cells; however XYS and RU-38486 reversed this change. Cell corticosterone stress [37]. In this study, we demonstrated that
cycle is a close mechanism that controls cell reproduction the underlying mechanism of XYS against corticosterone-
and proliferation. It is representatively divided into several induced stress in the PHN cells involved the upregulation of
phases: the periods associated with DNA synthesis phase (S the BDNF-TrkB pathway [35].
phase) and mitosis phase (M phase) that are separated by The FK506 binding proteins (FKBPs) are members of
gaps called G1 and G2 phases [32, 33]. In addition, the cells at a large superfamily of peptidyl-prolyl isomerases (PPIases)
G0 phase do not proliferate temporarily. The results showed [38, 39]. Particularly, FKBP4 and FKBP51 that possess C-
that XYS decreased the cells at G0/G1 and G2/M phases and terminal tetratricopeptide repeat (TPR) domains can bind to
increased the cells at S phase, which indicated that XYS raised GR [40, 41], and they play key roles in regulating the activities
the DNA synthesis of PHN cells. Furthermore, we found that of transient receptor potential channels and cell microtubule
XYS decreased the rate of apoptotic cells by flow cytometry function [41]. In our work, we found that GR DNA could
analysis. Our results showed that XYS exhibited potent ability bind with FKBP4 instead of FKBP51 by EMSA. Moreover,
to improve stress injury. the results of Dual-Luciferase Reporter Assay indicated that
Increased corticosterone level is a hallmark of HPA axis FKBP4 notably increased the GRE transcriptional activity
feedback inhibition, which is mediated by GR in the brain but FKBP51 significantly decreased the GRE transcriptional
[34]. The inhibition of BDNF-TrkB signaling pathway is asso- activity. XYS upregulated the FKBP4 mRNA and protein
ciated with memory impairment in corticosterone treatment levels and downregulated the FKBP51 mRNA and protein
animals [35]. Caveolin-1 is localised to the growth cones and levels. These results proved that XYS promoted the GRE
dendrites of hippocampal neurons and it promotes neuronal transcriptional activity.
differentiation and maturation [36]. FK506 binding protein of In summary, XYS conferred direct protective effects on
molecular weight 51 (FKBP51) and the related protein FKBP4 the corticosterone-induced PHN cells by promoting the
are thought to be involved in the regulation of GR signaling GRE transcriptional activity and the subsequent BDNF-TrkB
[37]. Our results showed that corticosterone resulted in the pathway in vitro. Accordingly, XYS represents a novel and
downregulation of Caveolin-1, which was reversed by XYS. potent candidate that should be developed as one potent
12 Evidence-Based Complementary and Alternative Medicine

Caveolin-1 GR
2.1 2.0

Relative mRNA expression

Relative mRNA expression


1.6 ∗∗

1.4 ∗ ∗∗
# ∗
1.2
##
0.8
0.7
0.4

0.0 0.0
Control

Cort

Medium

High

RU-38486

Control

Cort

Medium

High

RU-38486
Low

Low
(a) (b)
BDNF TrkB
1.8 2.0
Relative mRNA expression

Relative mRNA expression


∗∗
∗∗ 1.5 ∗∗
1.2 ∗∗
∗∗ ∗∗
∗ 1.0

0.6 ## ##
0.5

0.0 0.0
Control

Cort

Medium

High

RU-38486

Control

Cort

Medium

High

RU-38486
Low

Low
(c) (d)
FKBP4 FKBP51
1.8 1.6
##
Relative mRNA expression

Relative mRNA expression

∗∗
∗∗ 1.2
1.2 ∗∗ ∗∗

0.8
∗∗ ∗∗
0.6 ##
0.4

0.0 0.0
Control

Cort

Medium

High

RU-38486

Control

Cort

Medium

High

RU-38486
Low

Low

(e) (f)
Figure 8: The effects of XYS on the gene levels of Caveolin-1 (a), GR (b), BDNF (c), TrkB (d), FKBP4 (e), and FKBP51 (f) in brain tissue of
rats after corticosterone-induced stress injury (𝑛 = 3; ## 𝑃 < 0.01 versus Control; ∗ 𝑃 < 0.05 versus Cort; ∗∗ 𝑃 < 0.01 versus Cort).

candidate for the treatment of hippocampal dysfunction. Of EMSA: Electrophoretic mobility shift assay
course, further investigations are needed to investigate the GRE: GR reaction element
clinical applications of XYS. FKBP51: FK506 binding protein of molecular
weight 51
AD: Alzheimer’s disease
Abbreviations HPA: Hypothalamic-pituitary-adrenal
XYS: Xiao Yao San TCMs: Traditional Chinese medicines
PHN: Primary hippocampal neurons SGLFR: Shugan Lifei Recipe
GR: Glucocorticoid receptor EPM: Elevated plus-maze
BDNF: Brain derived neurotrophic factor FBS: Fetal bovine serum
Evidence-Based Complementary and Alternative Medicine 13

DMEM: Dulbecco’s minimum essential medium memory ability in mice,” Behavioral and Brain Functions, vol.
BCA: Bicinchoninic acid 11, no. 1, article 30, 2015.
MTT: Corticosterone, 3-(4,5-dimethylthiazol- [4] L. D. LaDage, “Environmental change, the stress response, and
2-yl)-2,5-diphenyl tetrazolium neurogenesis,” Integrative and Comparative Biology, vol. 55, no.
bromide 3, pp. 373–383, 2015.
DAPI: 4,6-Diamidino-2-phenylindole [5] D. Suri and V. A. Vaidya, “The adaptive and maladaptive contin-
MAP-2: Microtubule-associated protein 2 uum of stress responses—a hippocampal perspective,” Reviews
CSF: Cerebrospinal fluid in the Neurosciences, vol. 26, no. 4, pp. 415–442, 2015.
HPLC: High performance liquid [6] S. H. Takada, L. C. Motta-Teixeira, A. V. Machado-Nils et al.,
chromatography “Impact of neonatal anoxia on adult rat hippocampal volume,
SD: Sprague-Dawley neurogenesis and behavior,” Behavioural Brain Research, vol.
Cort: Corticosterone 296, pp. 331–338, 2016.
CNS: Central nervous system [7] E. Okun, K. Griffioen, B. Barak et al., “Toll-like receptor 3
PMSF: Phenylmethylsulfonyl fluoride inhibits memory retention and constrains adult hippocampal
ECL: Chemiluminescence. neurogenesis,” Proceedings of the National Academy of Sciences
of the United States of America, vol. 107, no. 35, pp. 15625–15630,
2010.
Additional Points [8] H. Xiong and H. J. Krugers, “Tuning hippocampal synapses by
stress-hormones: relevance for emotional memory formation,”
Availability of Data and Materials. Information on the per-
Brain Research, vol. 1621, pp. 114–120, 2015.
formance of antibodies for immunofluorescent staining and
Western blot was deposited in the open access database [9] T. Kino, “Stress, glucocorticoid hormones, and hippocampal
abcam (http://www.abm.cn/). The datasets were processed by neural progenitor cells: implications to mood disorders,” Fron-
tiers in Physiology, vol. 6, article 230, 2015.
the software GraphPad Prism 6 and Adobe Photoshop CS5.
[10] Y. Izumi, K. A. O’Dell, and C. F. Zorumski, “Corticosterone
enhances the potency of ethanol against hippocampal long-
Competing Interests term potentiation via local neurosteroid synthesis,” Frontiers in
Cellular Neuroscience, vol. 9, article 254, 2015.
The authors declare that there is no conflict of interests [11] D.-J. Saaltink and E. Vreugdenhil, “Stress, glucocorticoid recep-
regarding the publication of this paper. tors, and adult neurogenesis: a balance between excitation and
inhibition?” Cellular and Molecular Life Sciences, vol. 71, no. 13,
pp. 2499–2515, 2014.
Authors’ Contributions
[12] S. Chetty, A. R. Friedman, K. Taravosh-Lahn et al., “Stress and
Guoping Cao and Wenjun Fu designed the study and the glucocorticoids promote oligodendrogenesis in the adult hip-
experiments. Shenglan Gong carried out the PCR. Fengxue pocampus,” Molecular Psychiatry, vol. 19, no. 12, pp. 1275–1283,
Zhang and Wenjun Fu participated in the statistical analysis. 2014.
Guoping Cao and Wenjun Fu drafted the manuscript. All [13] I. Zalachoras, R. Houtman, E. Atucha et al., “Differential tar-
authors read and approved the final manuscript. geting of brain stress circuits with a selective glucocorticoid
receptor modulator,” Proceedings of the National Academy of
Sciences of the United States of America, vol. 110, no. 19, pp. 7910–
Acknowledgments 7915, 2013.
[14] T. B. Santos, I. C. Céspedes, and M. B. Viana, “Chronic corti-
This study is supported by National Natural Science Foun- costerone administration facilitates aversive memory retrieval
dation of China (no. 81403304) and Science and Tech- and increases GR/NOS immunoreactivity,” Behavioural Brain
nology Planning Project of Guangdong Province (no. Research, vol. 267, pp. 46–54, 2014.
2014A020221075). [15] D. Normile, “Asian medicine. The new face of traditional Chi-
nese medicine,” Science, vol. 299, no. 5604, pp. 188–190, 2003.
References [16] B. Lee, B. Sur, I. Shim, H. Lee, and D.-H. Hahm, “Baicalin im-
proves chronic corticosterone-induced learning and memory
[1] S. K. Tiwari, B. Seth, S. Agarwal et al., “Ethosuximide induces deficits via the enhancement of impaired hippocampal brain-
hippocampal neurogenesis and reverses cognitive deficits in an derived neurotrophic factor and cAMP response element-
amyloid-𝛽 toxin-induced Alzheimer rat model via the phos- binding protein expression in the rat,” Journal of Natural
phatidylinositol 3-kinase (PI3K)/Akt/Wnt/𝛽-catenin pathway,” Medicines, vol. 68, no. 1, pp. 132–143, 2014.
Journal of Biological Chemistry, vol. 290, no. 47, pp. 28540– [17] X. Wu, J. Wu, S. Xia, B. Li, and J. Dong, “Icaritin opposes the
28558, 2015. development of social aversion after defeat stress via increases
[2] C.-Y. Tsai, C.-Y. Tsai, S. J. Arnold, and G.-J. Huang, “Ablation of GR mRNA and BDNF mRNA in mice,” Behavioural Brain
of hippocampal neurogenesis in mice impairs the response to Research, vol. 256, pp. 602–608, 2013.
stress during the dark cycle,” Nature Communications, vol. 6, [18] H. Zhang, B. Liu, J. Wu et al., “Icariin inhibits corticosterone-
article 8373, 2015. induced apoptosis in hypothalamic neurons via the PI3-K/Akt
[3] Y. Yang, M. Zhang, X. Kang et al., “Thrombin-induced microgl- signaling pathway,” Molecular Medicine Reports, vol. 6, no. 5, pp.
ial activation impairs hippocampal neurogenesis and spatial 967–972, 2012.
14 Evidence-Based Complementary and Alternative Medicine

[19] P.-W. Zhuang, G.-Z. Cui, Y.-J. Zhang et al., “Baicalin regu- DNA damage and activating mitochondrial signal pathway,”
lates neuronal fate decision in neural stem/progenitor cells Food and Chemical Toxicology, vol. 59, pp. 118–128, 2013.
and stimulates hippocampal neurogenesis in adult rats,” CNS [34] Z.-J. Wang, X.-Q. Zhang, X.-Y. Cui et al., “Glucocorticoid re-
Neuroscience and Therapeutics, vol. 19, no. 3, pp. 154–162, 2013. ceptors in the locus coeruleus mediate sleep disorders caused
[20] Y. Tong, N.-X. Zhang, H.-J. Chen, and L.-Q. Ni, “Protective and by repeated corticosterone treatment,” Scientific Reports, vol. 5,
therapeutic effects of Shugan Lifei Recipe on rats with asthma article 9442, 2015.
under stress and the mechanisms,” Zhong Xi Yi Jie He Xue Bao, [35] H.-J. Wei, J.-H. Xu, M.-H. Li et al., “Hydrogen sulfide inhibits
vol. 3, no. 5, pp. 391–396, 2005. homocysteine-induced endoplasmic reticulum stress and neu-
[21] Y. Zhang, M. Han, Z. Liu, J. Wang, Q. He, and J. Liu, “Chinese ronal apoptosis in rat hippocampus via upregulation of the
herbal formula Xiao Yao San for treatment of depression: a BDNF-TrkB pathway,” Acta Pharmacologica Sinica, vol. 35, no.
systematic review of randomized controlled trials,” Evidence- 6, pp. 707–715, 2014.
Based Complementary and Alternative Medicine, vol. 2012, [36] Y. Li, W.-M. Lau, K.-F. So, Y. Tong, and J. Shen, “Caveolin-
Article ID 931636, 13 pages, 2012. 1 inhibits oligodendroglial differentiation of neural stem/pro-
[22] J.-X. Chen, W. Li, X. Zhao, and J.-X. Yang, “Effects of the genitor cells through modulating 𝛽-catenin expression,” Neuro-
Chinese traditional prescription Xiaoyaosan decoction on chemistry International, vol. 59, no. 2, pp. 114–121, 2011.
chronic immobilization stress-induced changes in behavior and [37] Z.-H. Zhu, R. Yang, X. Fu, Y.-Q. Wang, and G.-C. Wu, “Astro-
brain BDNF, TrkB, and NT-3 in rats,” Cellular and Molecular cyte-conditioned medium protecting hippocampal neurons in
Neurobiology, vol. 28, no. 5, pp. 745–755, 2008. primary cultures against corticosterone-induced damages via
[23] X.-F. Ding, X.-H. Zhao, Y. Tao et al., “Xiao yao san improves PI3-K/Akt signal pathway,” Brain Research, vol. 1114, no. 1, pp.
depressive-like behaviors in rats with chronic immobilization 1–10, 2006.
stress through modulation of locus coeruleus-norepinephrine [38] P. Chmielarz, G. Kreiner, M. Kot et al., “Disruption of gluco-
system,” Evidence-Based Complementary and Alternative corticoid receptors in the noradrenergic system leads to BDNF
Medicine, vol. 2014, Article ID 605914, 10 pages, 2014. up-regulation and altered serotonergic transmission associated
[24] X. Zhu, O. Xia, W. Han et al., “Xiao Yao San improves with a depressive-like phenotype in female GR(DBHCre) mice,”
depressive-like behavior in rats through modulation of 𝛽- Pharmacology Biochemistry and Behavior, vol. 137, pp. 69–77,
arrestin 2-mediated pathways in hippocampus,” Evidence-Based 2015.
Complementary and Alternative Medicine, vol. 2014, Article ID [39] A. Galat, “Functional drift of sequence attributes in the FK506-
902516, 13 pages, 2014. binding proteins (FKBPs),” Journal of Chemical Information and
[25] S.-Y. Yau, A. Li, and K.-F. So, “Involvement of adult hippocam- Modeling, vol. 48, no. 5, pp. 1118–1130, 2008.
pal neurogenesis in learning and forgetting,” Neural Plasticity, [40] J. G. Scammell, T. R. Hubler, W. B. Denny, and D. L. Valentine,
vol. 2015, Article ID 717958, 13 pages, 2015. “Organization of the human FK506-binding immunophilin
[26] L. Xiang, Y. Ren, H. Cai, W. Zhao, and Y. Song, “MicroRNA- FKBP52 protein gene (FKBP4),” Genomics, vol. 81, no. 6, pp.
132 aggravates epileptiform discharges via suppression of 640–643, 2003.
BDNF/TrkB signaling in cultured hippocampal neurons,” Brain [41] D. L. Cioffi, T. R. Hubler, and J. G. Scammell, “Organization and
Research, vol. 1622, pp. 484–495, 2015. function of the FKBP52 and FKBP51 genes,” Current Opinion in
[27] E. J. Kim, B. Pellman, and J. J. Kim, “Stress effects on the hip- Pharmacology, vol. 11, no. 4, pp. 308–313, 2011.
pocampus: a critical review,” Learning and Memory, vol. 22, no.
9, pp. 411–416, 2015.
[28] S. Paul, W. K. Jeon, J. L. Bizon, and J.-S. Han, “Interaction of
basal forebrain cholinergic neurons with the glucocorticoid
system in stress regulation and cognitive impairment,” Frontiers
in Aging Neuroscience, vol. 7, article 43, 2015.
[29] Y. Xu, B. Ku, L. Tie et al., “Curcumin reverses the effects of
chronic stress on behavior, the HPA axis, BDNF expression and
phosphorylation of CREB,” Brain Research, vol. 1122, no. 1, pp.
56–64, 2006.
[30] T. Kino and G. P. Chrousos, “Glucocorticoid and mineralocor-
ticoid receptors and associated diseases,” Essays in Biochemistry,
vol. 40, pp. 137–155, 2004.
[31] M. Buddle, E. Eberhardt, L. H. Ciminello et al., “Microtubule-
associated protein 2 (MAP2) associates with the NMDA
receptor and is spatially redistributed within rat hippocampal
neurons after oxygen-glucose deprivation,” Brain Research, vol.
978, no. 1-2, pp. 38–50, 2003.
[32] L. Lv, L. Zheng, D. Dong et al., “Dioscin, a natural steroid
saponin, induces apoptosis and DNA damage through reactive
oxygen species: a potential new drug for treatment of glioblas-
toma multiforme,” Food and Chemical Toxicology, vol. 59, pp.
657–669, 2013.
[33] Y. Wei, Y. Xu, X. Han et al., “Anti-cancer effects of dioscin on
three kinds of human lung cancer cell lines through inducing
MEDIATORS of

INFLAMMATION

The Scientific Gastroenterology Journal of


World Journal
Hindawi Publishing Corporation
Research and Practice
Hindawi Publishing Corporation
Hindawi Publishing Corporation
Diabetes Research
Hindawi Publishing Corporation
Disease Markers
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Journal of International Journal of


Immunology Research
Hindawi Publishing Corporation
Endocrinology
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Submit your manuscripts at


http://www.hindawi.com

BioMed
PPAR Research
Hindawi Publishing Corporation
Research International
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Journal of
Obesity

Evidence-Based
Journal of Stem Cells Complementary and Journal of
Ophthalmology
Hindawi Publishing Corporation
International
Hindawi Publishing Corporation
Alternative Medicine
Hindawi Publishing Corporation Hindawi Publishing Corporation
Oncology
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Parkinson’s
Disease

Computational and
Mathematical Methods
in Medicine
Behavioural
Neurology
AIDS
Research and Treatment
Oxidative Medicine and
Cellular Longevity
Hindawi Publishing Corporation Hindawi Publishing Corporation Hindawi Publishing Corporation Hindawi Publishing Corporation Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

You might also like