ArticlePDF Available

Epigallocatechin-3-gallate rescues LPS-impaired adult hippocampal neurogenesis through suppressing the TLR4-NF-κB signaling pathway in mice

Authors:

Abstract and Figures

Adult hippocampal dentate granule neurons are generated from neural stem cells (NSCs) in the mammalian brain, and the fate specification of adult NSCs is precisely controlled by the local niches and environment, such as the subventricular zone (SVZ), dentate gyrus (DG), and Toll-like receptors (TLRs). Epigallocatechin-3-gallate (EGCG) is the main polyphenolic flavonoid in green tea that has neuroprotective activities, but there is no clear understanding of the role of EGCG in adult neurogenesis in the DG after neuroinflammation. Here, we investigate the effect and the mechanism of EGCG on adult neurogenesis impaired by lipopolysaccharides (LPS). LPS-induced neuroinflammation inhibited adult neurogenesis by suppressing the proliferation and differentiation of neural stem cells in the DG, which was indicated by the decreased number of Bromodeoxyuridine (BrdU)-, Doublecortin (DCX)- and Neuronal Nuclei (NeuN)-positive cells. In addition, microglia were recruited with activatingTLR4-NF-κB signaling in the adult hippocampus by LPS injection. Treating LPS-injured mice with EGCG restored the proliferation and differentiation of NSCs in the DG, which were decreased by LPS, and EGCG treatment also ameliorated the apoptosis of NSCs. Moreover, pro-inflammatory cytokine production induced by LPS was attenuated by EGCG treatment through modulating the TLR4-NF-κB pathway. These results illustrate that EGCG has a beneficial effect on impaired adult neurogenesis caused by LPSinduced neuroinflammation, and it may be applicable as a therapeutic agent against neurodegenerative disorders caused by inflammation.
Content may be subject to copyright.
www.kjpp.net Korean J Physiol Pharmacol 2016;20(1):41-51
41
Korean J Physiol Pharmacol 2016;20(1):41-51
http://dx.doi.org/10.4196/kjpp.2016.20.1.41
Author contributions: K.J.S., H.G.L., J.Y.J. and W.J.K. designed the experi-
ments. Cell cultures, immunohistochemistry and molecular works was
performed by S.H.C., H.G.L., H.M.K. and M.S.K. K.J.S. and H.G.L. took care
all animals. K.J.S., H.G.L., J.Y.J. and W.J.K. analyzed the data and wrote the
manuscript.
This is an Open Access article distributed under the terms
of the Creative Commons Attribution Non-Commercial
License, which permits unrestricted non-commercial use, distribution, and
reproduction in any medium, provided the original work is properly cited.
Copyright © Korean J Physiol Pharmacol, pISSN 1226-4512, eISSN 2093-3827
INTRODUCTION
Neuroinflammation with microglial activation plays a role
in neurodegenerative diseases, such as Alzheimer's disease and
multiple sclerosis [1], and microglia involved in the inflammation
of the central nervous system have neuroprotective effects [2-
4]. Recent
in vivo
and
in vitro
studies have suggested that
microglia are essential in neural development, synapse formation,
and neuroinflammation involved in the impairment of adult
neurogenesis [5-7]. This evidence was supported by the Gram-
negative bacterial immune stimulation lipopolysaccharide (LPS)-
induced neuroinflammatory model, which showed that LPS-
induced activation of the Toll-like receptor 4 (TLR4) in microglia
led to the production of the pro-inflammatory cytokines, IL-
1
b
, IL-6, and TNF-
a
through the MAPK and NF
k
B pathways
in BV2 cell lines [8,9]. In addition to the immune response by
activated microglia, the neuroprotective cytokines, IL-4 and IL-
25, are also produced [10,11]. Moreover, other studies showed that
Original Article
Epigallocatechin3gallate rescues LPSimpaired adult
hippocampal neurogenesis through suppressing the TLR4NF
k
B
signaling pathway in mice
Kyung-Joo Seong
1,2,3,#
, Hyun-Gwan Lee
1,2,3,#
, Min Suk Kook
1,2,4
, Hyun-Mi Ko
5
, Ji-Yeon Jung
1,2,3,
*, and Won-Jae Kim
1,2,3,
*
1
Dental Science Research Institute,
2
Medical Research Center for Biomineralization Disorders,
3
Department of Oral Physiology,
4
Department of Oral and
Maxillofacial Surgery, School of Dentistry, Chonnam National University, Gwangju 61186,
5
Department of Microbiology, Collage of Medicine, Seonam
Universtity, Namwon 55724, Korea
ARTICLE INFO
Received July 9, 2015
Revised
August 21, 2015
Accepted
September 1, 2015
*Correspondence
Won-Jae Kim
E-mail: wjkim@jnu.ac.kr
Ji-Yeon Jung
E-mail: jjy@jnu.ac.kr
Key Words
Adult Neurogenesis
Epigallocatechin-3-gallate
Neural stem cells
Neuronal Inammation
NF-
k
B signaling
TLR4
#
These authors contributed equally to
this work.
ABSTRACT Adult hippocampal dentate granule neurons are generated from
neural stem cells (NSCs) in the mammalian brain, and the fate specification of
adult NSCs is precisely controlled by the local niches and environment, such as
the subventricular zone (SVZ), dentate gyrus (DG), and Toll-like receptors (TLRs).
Epigallocatechin-3-gallate (EGCG) is the main polyphenolic flavonoid in green
tea that has neuroprotective activities, but there is no clear understanding of the
role of EGCG in adult neurogenesis in the DG after neuroinflammation. Here, we
investigate the eect and the mechanism of EGCG on adult neurogenesis impaired
by lipopolysaccharides (LPS). LPS-induced neuroinflammation inhibited adult
neurogenesis by suppressing the proliferation and dierentiation of neural stem cells
in the DG, which was indicated by the decreased number of Bromodeoxyuridine
(BrdU)-, Doublecortin (DCX)- and Neuronal Nuclei (NeuN)-positive cells. In
addition, microglia were recruited with activatingTLR4-NF-
k
B signaling in the
adult hippocampus by LPS injection. Treating LPS-injured mice with EGCG restored
the proliferation and differentiation of NSCs in the DG, which were decreased
by LPS, and EGCG treatment also ameliorated the apoptosis of NSCs. Moreover,
pro-inflammatory cytokine production induced by LPS was attenuated by EGCG
treatment through modulating the TLR4-NF-
k
B pathway. These results illustrate
that EGCG has a beneficial effect on impaired adult neurogenesis caused by LPS-
induced neuroinammation, and it may be applicable as a therapeutic agent against
neurodegenerative disorders caused by inammation.
42
http://dx.doi.org/10.4196/kjpp.2016.20.1.41Korean J Physiol Pharmacol 2016;20(1):41-51
Seong KJ et al
adult neurogenesis in the hippocampus was affected through the
TLR and NF-
k
B pathway in microglia [12 ,13].
Adult neurogenesis is a process in which new neurons are
continuously generated throughout adult life from the neural
stem cells (NSCs) in the mammalian brain. Small populations
of neurons continue to be born in the adult subventricular
zone, the olfactory system, and the DG of the hippocampus.
The neurogenic niches are important for the NSCs to have
self-renewal and multipotent properties that can give rise
to neurons, astrocytes, and oligodendrocytes [14-16]. The
neuronal fate specification (neurogenesis) and glial fate
specification (gliogenesis) of the NSCs is determined by cellular
communication within the microenvironment [17]. The newborn
neurons are critical to cognitive functions including learning
and memory. For instance, physical activity and an enriched
environment, which are associated with improved memory
function and synaptic plasticity, enhance adult neurogenesis
in the DG [18,19]. Furthermore, adult neurogenesis in the DG
declines with aging and neurodegenerative diseases, which in
turn causes cognitive deficits [20-22].
Green tea has been shown to function as an anticancer
reagent that inhibits the development and progression of skin,
lung, mammary gland, and gastrointestinal tract tumors
in animal models [23,24]. Among the various polyphenols
in green tea including epigallocatechin-3-gallate (EGCG),
()-epigallocatechin, ()-epicatechin, (+)-gallocatechin, and other
catechins, EGCG is the most abundant and the most biologically
active component [25-27]. However, the recovery effects of EGCG
on adult neurogenesis following neuroinflammation at the DG
in the hippocampus are still unknown. In the present study, we
investigate the effect of EGCG on impaired adult neurogenesis in
a mouse model of LPS-induced neuroinflammation.
Here, we present the effects of EGCG on impaired adult
hippocampal neurogenesis
in vivo
caused by neuroinflammation.
We induced inflammation by administering LPS in a
mouse model and explored the positive effects of EGCG
in NSC proliferation and neural differentiation using an
immunohistochemical approach to detect bromodeoxyuridine
(BrdU, an S-phase marker of proliferating cells), doublecortin
(DCX, a marker of immature newborn neurons), neuronal nuclei
(NeuN, a marker of mature neurons), ionized calcium binding
adapter molecule 1 (Iba-1, a marker of microglia), and the levels
of cytokines.
METHODS
Animals & chemicals
7-week-old male C57BL/6 mice were purchased from Daehan
Biolink (Chungbuk, Korea). The animals were kept under
standard conditions of 22
o
C and a humidity of 55% with a
12 hour light-12 hour dark cycle, and allowed free access to
food and water. Mice were divided into the following three
groups: saline-treated group (sham control), saline-treated
with lipopolysaccharide (LPS,
E
.
coli
, serotype 055:B5, St. Louis,
MO, USA) intracerebroventricular (I.C.V.) injection group,
and epigallocatechin-3-gallate (EGCG)-treated with LPS I.C.V.
injection group (n=5 mice/ each group). Supplementary Figure 1
demonstrated the schematic experimental design of time courses
with LPS, EGCG and BrdU injection followed by sampling the
brain for immunohistochemistry. All experiments were approved
by the Animal Care and Use Committee of Chonnam National
University. All chemicals used in the experiments were supplied
by Sigma (St. Louis, MO, USA).
LPS-induced neuroinflammation in the brain
Experimental mice were anesthetized with zoletil (VIRBAC,
Milperra, Australia) (25 mg/kg) and Rompun (Bayer HealthCare,
Mississauga, Canada) (10 mg/kg). LPS for a single unilateral
stereotaxic injection was prepared in phosphate-buffered saline
(PBS, pH 7.4) at a final concentration of 1.0 mg/ml and was
injected into the brain (0.6 mm caudal to the bregma, 1.5 mm to
the right of the bregma, 2.0 mm ventral to the bregma). Mice were
injected with vehicle (sterile saline) or LPS (3
m
g in 3
m
l sterile
saline) following brain surgery at a dose of 1
m
g/min.
EGCG treatment
EGCG (0.5 mg/kg) prepared in physiological saline (PBS
containing 0.9% NaCl) was injected intraperitoneally (i.p.) for
three times with 6 hours interval at 3 hours after recovered from
anesthesia for LPS-injection.
BrdU injection
BrdU (Sigma, St. Louis, MO, USA) was dissolved in
physiological saline and injected i.p. (50 mg/kg). To examine
the proliferation of NSCs in the DG, BrdU was injected into the
animals once, 3 hours prior to sacrifice. To study the survival rate
of newborn cells generated from NSCs and the differentiation rate
of NSCs, BrdU was injected once daily for 5 consecutive days and
the mice were sacrificed on 28
th
day after the final BrdU injection.
Tissue preparation
Animals were perfused transcardially with 0.1 M PBS, followed
by fresh cold 4% paraformaldehyde (PFA) in 0.1 M PBS. The
brains were removed and fixed overnight in 4% PFA in 0.1 M
PBS at 4
o
C, and then washed for 6 hours in PBS at 4
o
C. Sucrose-
saturated brains were then embedded in freezing media (O.C.T
compound, Leica Biosystems, Richmond, USA), frozen in chilled
isopentane (25
o
C), and stored at –80
o
C until sectioning. Brains
EGCG in adult neurogenesis impaired neuroinflammation in mouse
Korean J Physiol Pharmacol 2016;20(1):41-51www.kjpp.net
43
were cryocut coronally at a thickness of 40
m
m using a Cryostat
(Model CM3050; Leica Microsystems, Richmond, USA) and
stored in cryoprotectant solution (25% ethylene glycol, 25%
glycerol, and 0.05 M sodium phosphate buffer, Na-PB) at –20
o
C
until immunohistochemical (IHC) processing.
Fluorescent immunohistochemical staining (IHC)
Sections were washed in Na-PB and mounted on charged
slide glasses for IHC. The sections immunostained for BrdU
were pretreated with 2 N HCl for 30 minutes at 37
o
C and
neutralized with PBS before incubation with primary antibodies.
Sections were incubated for 60 minutes with 5% normal horse
serum in 0.4% Triton X-100 in PBS (PBST). The sections were
incubated overnight at 4
o
C with primary antibodies in the same
buffer solution. Primary antibodies were used at the following
concentrations: rat anti-BrdU (1:200; Abcam, Cambridge, UK),
goat anti-doublecortin (DCX, 1:100; Cell Signaling, Danvers,
MA), mouse anti-neuronal nuclear antigen (NeuN, 1:100;
Chemicon, Temecula, CA), and rabbit anti-ionized calcium-
binding adapter molecule 1 (Iba-1, 1:100; Wako Chemicals USA,
Inc., Richmond, VA). Sections were washed three times with
PBST for 10 minutes at room temperature and blocked in PBST
containing 5% horse serum for 30 minutes. Sections were then
incubated for 2 hours with secondary antibodies conjugated
to FITC (Jackson Immuno-Research, West Grove, PA) or CY3
(Jackson Immuno-Research, West Grove, PA). The sections
were washed three times with PBST, and stained with 10 mg/ml
4’6’-diamidino-2-phenylindole (DAPI) (Sigma-Aldrich, St. Louis,
USA) for 30 minutes before mounted.
Quantification and image analysis
IHC images were acquired with a confocal microscope (Zeiss,
Thornwood, NY) equipped with an argon/krypton laser (488
nm), two helium/neon lasers (543 and 633 nm), and a coherent
laser (Santa Clara, CA), using the 20x objective lens. Images
were analyzed with the Image J (Ver.1.4, NIH, USA). Cells were
counted in a defined frame size of 20~50 with 35
m
m×35
m
m
in every selected section. Colocalizations were investigated
by performing z-stack acquisitions and three-dimensional
reconstructions with the LSM software (Zeiss, Thornwood, NY).
Adobe Photoshop (Version CS6, Adobe Systems, San Jose, CA)
was used to adjust the contrast and brightness.
Western blot analysis
Brain was homogenized in RIPA buffer (150 mM NaCl, 1.0%
IGEPAL
CA-630, 0.5% sodium deoxycholate and 0.1% SDS in
50 mM Tris, Sigma, St. Louis, MO, USA). Proteins were separated
by SDS-PAGE and transferred to a nitrocellulose membrane. The
membranes were incubated with primary anti-TLR4, anti-RelA,
anti-pRelA, and anti-Actin antibodies (Cell Signaling, Danvers,
MA or Abcam, Cambridge, UK). The expression levels of protein
were visualized and analyzed with Fusion FX (Vilber Lourmat,
Eberhardzell, Germany).
Cytokine measurements
The levels of IL-1
b
, IL-6, and TNF-
a
in the DG were measured
using a commercial ELISA kit (R&D Systems, Minneapolis,
MN, USA) as per the manufacturer's instructions, and using
quantitative real-time PCR (Bio-Rad Lab. Inc., CA, USA). In
detail, the total RNAs were prepared using Trizol Reagent (Gibco
BRL) and reverse transcription was performed with a RT system
containing Moloney Murine Leukemia Virus reverse transcriptase
(Promega, Madison, WI) in accordance with the manufacturer’s
instructions. PCR was performed in a Palm-Cycler thermocycler
(Corbett Life Science, Sydney, Australia) and the product was
resolved in a 1.2% agarose gel. Real time amplification of cDNA
was conducted in a Rotor-Gene 3000 System (Corbett Research,
Morklake, Australia) using the SYBR Green PCR Master Mix
Reagent Kit (Qiagen, Valencia, CA). The PCR conditions were as
follows: incubation for 5 minutes at 95
o
C, followed by 30 cycles of
denaturation for 15 seconds at 95
o
C, annealing for 15 seconds at
62
o
C and extension for 15 seconds at 72
o
C. The primers were as
follows: mouse IL-1
b
; 5'-AGG AGA ACC AAG CAA CGA CA-3'
and 5'-CTT GGG ATC CAC ACT CTC CAG-3', mouse IL-6; 5'-
GCC TTC TTG GGA CTG ATG CT-3' and 5'-GCC TTC TTG
GGA CTG ATG CT-3', mouse TNF-
a
; 5'-ATG GCC TCC CTC
TCA TCA GT-3' and 5'-CTT GGT GGT TTG CTA CGA CG-
3' and
b
-actin; 5'-GAT CTG GCA CCA CAC CTT CT-3' and 5'-
GGG GTG TTG AAG GTC TCA AA-3'. The relative levels of
mRNA were calculated using the standard curve generated from
the cDNA dilutions. The mean cycle threshold (Ct) values from
quadruplicate measurements were employed in the calculation
of gene expression, with normalization to β-actin as an internal
control. Calculation of the relative levels of gene expression was
performed using Corbett Robotics Rotor-Gene software (Rotor-
Gene 6 version 6.1, Build 90).
Data analysis
A complete series of 1 in 10 sections of the DG was analyzed.
For each experiment, 9 sections per mouse were selected for
analysis. The immunolabeled cells were counted and multiplied
by 10 to obtain the total number of labeled cells throughout
the DG. The data are expressed as the mean±SEM. Statistical
analyses were performed using a one-way analysis of variance
(ANOVA) followed by a Student-Newman-Keuls test for multiple
comparisons. Results were considered statistically significant
when the p value was less than 0.05.
44
http://dx.doi.org/10.4196/kjpp.2016.20.1.41Korean J Physiol Pharmacol 2016;20(1):41-51
Seong KJ et al
RESULTS
EGCG recovered the proliferation of adult NSCs in the
LPS-injured hippocampal DG
To study the effect of EGCG in the DG related to neurogenesis,
the proliferation of adult NSCs in the DG was initially examined
in an LPS-induced neuroinflammation mouse model by
administering LPS (3
m
g/animal with 1 mg/ml stock) via
intracerebroventricular (I.C.V.) injection (n=5 each). The number
of BrdU (S-phase marker)-positive cells were compared at 3 hours
following a single BrdU injection (i.p. 50 mg/kg), which indicates
proliferating cells, and were found to be significantly decreased
at 1 day (0.7066±0.044010
3
, 38% of decreased compared to
sham control,
F
(4.4)=1.076, p=0.0002, two-tailed unpaired one-
way ANOVA) and day 3 (0.8292±0.034610
3
, 29% of decreased
compared to sham control,
F
(4.4)=1.577, p=0.0003, two-tailed
unpaired one-way ANOVA) post-LPS injection in mice treated
with either vehicle or EGCG compared with the sham controls.
However, EGCG significantly improved the number of BrdU-
positive cells affected by LPS in the hippocampal DG (day1;
0.8242±0.01520×10
3
, 16% of increased compared with LPS only,
F
(4.4)=8.406, p=0.0357, day 3; 0.9462±0.03309×10
3
, 14% of
increased compared with LPS only,
F
(4.4)=1.096, p=0.0404, two-
tailed unpaired one-way ANOVA) (Fig. 1). These results show
that treatment with EGCG recovered NSC proliferation, which
was impaired by LPS-induced neuroinflammation.
EGCG ameliorated the immature neuronal
differentiation of adult NSCs in the LPS-injured
hippocampal DG
DCX (immature neuronal marker) is transiently expressed
in the soma and dendrites of immature newborn neurons.
The newborn NSCs are differentiated into neurons to serve
as functional units in the nervous system following their
proliferation. Thus, we studied whether EGCG ameliorated
the differentiation of adult NSCs after LPS-induced injury by
counting the number of BrdU- and DCX double-positive cells
and the ratio of BrdU
+
DCX
+
cells/BrdU
+
cells in the hippocampal
DG 5 days after injection with BrdU (n=5 each). The number of
double positive cells was decreased post LPS injection compared
to that of sham controls (1.070±0.06641×10
3
, 32% of decreased
compared to sham control,
F
(4.4)=1.287, p=0.0013), but it was
significantly improved after injection with EGCG in the LPS-
injured group compared with the LPS-injured group with no
treatment (1.390.07632×10
3
, 30% of increased compared to LPS
only,
F
(4.4)=1.321, p=0.0118) (Fig. 2A and B). Similarly, the ratio
of the total number of BrdU- and DCX-double positive cells to
the total number of BrdU-positive cells was also decreased in the
LPS-treated group compared with the sham controls (43.60±1.887
×10
3
, 11.4% of decreased compared to sham control,
F
(4.4)=1.017,
p=0.0026), but the ratio was recovered in the EGCG-treated
LPS-injured group compared with the LPS-injured group with
no treatment (50.80±1.985×10
3
, 7.2% of increased compared to
LPS only,
F
(4.4)=1.107, p=0.0302, two-tailed unpaired one-way
A NOVA) (Fig. 2A and C). These results suggest that EGCG had
a recovery effect on the neuronal differentiation of adult NSCs,
which was impaired by LPS in the early stage.
Fig. 1. The effect of EGCG on the prolife-
ration of adult NSCs in the DG impaired
by LPS-induced neuroinflammation.
(A) Representative images show BrdU-
positive cells in the adult dentate gyrus
(DG) on day 1 and day 3 after BrdU in-
jection following LPS injection. Newly
proliferated cells in the subgranular zone
(SGZ) of the DG were immunolabeled
using anti-BrdU (green). (B, C) Quantitative
analysis of the number of BrdU-positive
cells in the DG of the hippocampus
on day 1 and day 3 after LPS injection.
(n=5 each). BrdU-positive cells were
decreased in the LPS-injured group at
1
st
, and 3
rd
day post brain inflammation,
which was improved significantly
by EGCG (p=0.0357 and p=0.0404
respectively, one-way ANOVA) (*p<0.05,
***p<0.001). Data are expressed as the
mean±SEM. Scale bar, 200
m
m.
EGCG in adult neurogenesis impaired neuroinflammation in mouse
Korean J Physiol Pharmacol 2016;20(1):41-51www.kjpp.net
45
EGCG amended the mature neuronal differentiation
of adult NSCs in the LPS-injured hippocampal DG
To investigate the effect of EGCG on the maturation of
newborn neurons following LPS-induced neuroinflammation,
immunohistochemical analysis was performed using antibodies
against the mature neuronal marker, NeuN, and BrdU. The
results show that the number of cells co-localized with NeuN and
BrdU are decreased in the LPS-injured group (0.6280±0.03967×
10
3
, 27% of decreased compared to sham control,
F
(4.4)=1.623,
p=0.0021), but the number of these cells are increased in the
EGCG-treated LPS-injured group compared with the vehicle-
treated LPS-injured group (0.7476±0.02307×10
3
, 19% of increased
compared to LPS only,
F
(4.4)=2.958, p=0.0313) (Fig. 3A and B).
Furthermore, the ratio of NeuN/BrdU double-positive cells to
the total BrdU-positive cells in the EGCG-treated LPS-injured
group was significantly increased compared with that of the
vehicle-treated LPS-injured group (52.80±1.158, 5.6% of increased
compared to LPS only, F(4.4)=2.791, p=0.0378, two-tailed
unpaired one-way ANOVA), which was decreased compared
with the sham control group (Fig. 3C). These results support
the notion of a beneficial effect of EGCG on mature neuronal
differentiation of adult NSCs, which was impaired by LPS-
induced neuroinflammation.
EGCG improved adult NSC survival in the LPS-injured
hippocampal DG
Following NSC proliferation in the DG, the newborn NSCs
survive and differentiate into neurons. To determine the survival
rate of newborn cells derived from NSCs in the DG, the number
of BrdU-positive cells was quantified 3 hours and 28 days after
the last injection of BrdU, which was injected once a day for 5
consecutive days. The total number of BrdU-positive cells was
decreased in the LPS-injured group, and was recovered in the
EGCG-treated LPS-injured group up to the level of the sham
controls (n=5 each). Subsequently, we quantified the survival rate
of newborn cells by counting the total BrdU-positive cells, 28
days after the final of 5 consecutive days of BrdU injections, to
BrdU-positive cells, 3 hours after the final of 5 consecutive days
of BrdU injections. The rate was decreased in all groups, but the
EGCG-treated LPS-injured group showed an improved survival
rate of newborn cells compared with the LPS-injured group
with no treatment (43.20±0.8602, 3.8% of increased compared
to LPS only,
F
(4.4)=1.162, p=0.0170, two-tailed unpaired one-
way ANOVA) (Fig. 4A and C). Further, to examine the effect of
Fig. 2. The effect of EGCG on the im-
mature neuronal differentiation in
the DG after LPS-induced neuroin-
flammation. (A) Representative images
show BrdU- and DCX-positive cells in the
DG 5 days after BrdU injection following
LPS injection. BrdU (green) and DCX
(red) double stained cells (yellow)
represent newly generated immature
neurons (arrows). (B, C) Quantitative
analysis of the number of BrdU-po-
sitive cells and/or DCX-positive cells
(n=5 each). The differentiation of the
NSCs in the hippocampal DG affected
by LPS-induced neuroinflammation
was recovered by EGCG (0.5 mg/kg)
(p=0.0302, one-way ANOVA) (*p<0.05,
**p<0.01). Data are expressed as the
mean±SEM. Scale bar, 200
m
m.
46
http://dx.doi.org/10.4196/kjpp.2016.20.1.41Korean J Physiol Pharmacol 2016;20(1):41-51
Seong KJ et al
EGCG on newborn cell survival related to apoptosis, the number
of cleaved caspase 3-positive cells was counted. The total number
of cleaved caspase 3-positive cells was significantly decreased in
the EGCG-treated LPS-injured group compared with the vehicle-
treated LPS-injured group (0.4880.01393×10
2
, 12% of decreased
compared to LPS only, F (4.4)=1.062, p=0.0109, two-tailed
unpaired one-way ANOVA) (Fig. 4B and D). The results show
that neuroinflammation decreased the survival of dividing cells,
whereas EGCG recovered the survival of dividing cells, which
was impaired by LPS-induced neuroinflammation.
EGCG downregulated the expression of
proinflammatory cytokines through the TLR4/NF-
k
B
pathway in LPS-induced neuroinflammation
To study the effect of EGCG on neuroinflammation in
detail, the activity of microglia following LPS I.C.V. injection
was quantified by the immunoreactivity of Iba-1, a specific
marker of microglia (n=5 each). The number of Iba-1-positive
cells in the DG was increased (day 1; 4.876±0.2238×10
3
, 54% of
increased compared to sham control,
F
(4.4)=2.181, p=0.0002,
day 3; 4.528±0.2567×10
3
, 39% of increased compared to sham
control,
F
(4.4)=2.917, p=0.0025, two-tailed unpaired one-way
ANOVA ) by LPS injection, which was suppressed at day 1
and day 3 of EGCG treatment (day 1; 3.393±0.2497×10
3
, 7% of
decreased compared to LPS only,
F
(4.4)=1.245, p=0.0022, day
3; 3.402±0.1609×10
3
, 4% of decreased compared to LPS only,
F
(4.4)=2.544, p=0.0059, two-tailed unpaired one-way ANOVA )
(Fig. 5A and B). Since LPS acted as a TLR4 agonist, the expression
of TLR4, NF-
k
B, and cytokines was examined as molecular
targets of EGCG in adult neurogenesis impaired by LPS-induced
neuroinflammation. The expression of TLR4 and phospho-
RelA (activated form) was upregulated in the LPS-injured group
compared with the sham controls, but the expression of TLR4
and phospho-RelA was suppressed in the EGCG-treated LPS-
injured group compared with the LPS-injured group. Therefore,
LPS activated the expression of TLR4 and stimulated NF-
k
B, but
EGCG attenuated the LPS-induced TLR4 and NF-
k
B (Fig. 5C).
In addition, LPS upregulated the production of IL-1
b
, IL-6, and
TNF-
a
by microglia, however, the increased mRNA and protein
levels of cytokines induced by LPS injection were compromised
by EGCG up to 53.75%, 46.67%, and 30.71% for IL-1
b
, IL-6 and
TNF-
a
transcripts, respectively, and up to 72.61%, 47.59% and
59.57% for proteins, respectively, compared with the expression
levels of the LPS-injured group (p<0.05) (Fig. 5D~G). The results
demonstrate that EGCG suppressed the inflammatory activities
Fig. 3. The effect of EGCG on the mature
neuronal differentiation in the DG after
LPS- induced neuroinflammation. (A)
Representative images show BrdU and
NeuN double immunostained cells in
the DG 28 days after BrdU injection
following LPS injection. BrdU (green)
and NeuN (red) double immunostained
cells (yellow) represent newly generated
mature neurons (arrows). (B, C) Quantitative
analysis of the number of BrdU- and/or
NeuN-positive cells (n=5 each). BrdU and
NeuN double positive cells represent
matured neurons from adult NSCs that
are affected by neuroinflammation, and
the damage was rescued by EGCG (0.5
mg/kg) (p=0.0378, one-way ANOVA) (*p
<0.05, **p<0.01). Data are expressed as
the mean±SEM. Scale bar, 200 µm.
EGCG in adult neurogenesis impaired neuroinflammation in mouse
Korean J Physiol Pharmacol 2016;20(1):41-51www.kjpp.net
47
of microglia, resulting in the attenuation of proinflammatory
cytokine production via the TLR4/NF-
k
B signaling pathway.
DISCUSSION
Adult NSCs in the hippocampal DG have the ability to generate
self-renewing neural stem cells possessing multipotency [28].
Adult neurogenesis occurs in three discrete stages, proliferation
of NSCs, cell survival, and neuronal differentiation, which are
affected by various factors such as hormones, growth factors, enriched
environment, stress, drugs, and pathological stimulation [29,30].
The process in the DG affected by these factors is associated
with cognitive functions including learning and memory [31].
Neuroinflammation in the brain following injuries causes neural
development-related diseases, which modulate adult hippocampal
neurogenesis [32,33]. During the inflammatory response
following injury to the central nervous system (CNS), microglial
cells and a population of glial cells of the CNS as immune cells
produce proinflammatory cytokines. In particular, IL-6 and NO
affect adult hippocampal neurogenesis, which are involved in the
pathogenesis of neurological diseases such as Alzheimer's disease
(AD) and Parkinson's disease (PD) [34,35].
EGCG, among the catechins, a major subgroup of poly-
phenolic flavonoids in green tea, is well-known for its anti-
carcinogenic effects via the suppression of the proliferation and
angiogenesis of cancer cells [36]. Even though the instability
of catechins, previous studies were showing that EGCG was
effectively delivered over blood brain barrier and was founded
physiologically activated forms in the brain [37,38]. In addition,
it may be used for the prevention and treatment of AD and PD
based on the evidence that EGCG promotes adult hippocampal
neurogenesis [39-41]. Moreover, recent studies have demonstrated
that EGCG had a neuroprotective effect through the reduction of
neuroinflammation [42-45]. Oxidative stress has been implicated
in the pathophysiology of the majority of neurodegenerative
diseases, and most of the researches in this field have focused on
EGCG as a natural pharmacological compound [46]. However,
there have been no studies assessing the effect of EGCG on
neurogenesis in the DG of the adult hippocampus impaired by
neuroinflammation. Therefore, here, we investigated whether
EGCG improves adult neurogenesis via the modulation of
neuroinflammation
in vivo
.
In the present study, the effect of EGCG on the proliferation of
adult NSCs in the DG following LPS injection was investigated.
Proliferation of adult NSCs in the DG was inhibited by LPS
Fig. 4. The effect of EGCG on the survival
of newborn cells in the DG after LPS-
induced neuroinflammation. (A) Re-
presentative images show BrdU-positive
cells (green) in the DG at 3 hours (upper)
or 28 days (lower) after consecutive
BrdU injection for 5 days, following LPS
injection. (B) Representative images
show cleaved caspase-3 immunoreactive
cells (red) in the DG 28 days after
consecutive BrdU injection for 5 days.
(C) The survival rate of newborn cells
in LPS-induced injures was improved
significantly by EGCG (0.5 mg/kg) which
was analyzed by counting BrdU-positive
cells in the DG of the hippocampus at 5
days over 28 days (n =5 each, p=0.0170,
one-way ANOVA) (*p<0.05, **p<0.01).
(D) Quantitative analysis of the number
of cleaved caspase-3-positive cells in the
DG of the hippocampus (n=5 each). The
number of cleaved caspase-3-positive
cells in the DG of the hippocampus
was decreased in the EGCG-treated
LPS-injured group (p=0.0109, one-way
ANOVA) (*p<0.05). Data are expressed as
the mean±SEM; Scale bar in A and B, 200
m
m.
48
http://dx.doi.org/10.4196/kjpp.2016.20.1.41Korean J Physiol Pharmacol 2016;20(1):41-51
Seong KJ et al
injection, however, the number of BrdU-positive cells indicating
the rate of adult NSC proliferation was improved within one day
in the EGCG-treated LPS-injured group (Fig. 1A~C). Moreover,
the survival rate of newborn cells, represented by the ratio of
the total number of BrdU-positive cells 28 days after the final
BrdU injection following LPS injection, was amended by EGCG
treatment (Fig. 2A~C). These results suggest that EGCG rescued
the proliferation and survival of adult NSCs in the DG impaired
by LPS-induced neuroinflammation, which is consistent with
that of previous reports showing the effective role of EGCG in
increasing cell proliferation in mouse hippocampal DG and
neural progenitor cell proliferation during adult hippocampal
neurogenesis [47, 4 8].
To investigate the effect of EGCG on immature neuronal
differentiation in the DG following LPS-induced neuroin-
flammation, immunohistochemical analysis was performed using
a marker of immature neural differentiation, DCX, and BrdU, 5
days after BrdU injection following LPS with or without EGCG
injection. The number of double positive cells (DCX
+
and BrdU
+
)
was decreased post LPS injection compared with that of the sham
controls, however, it was significantly improved in the EGCG-
treated LPS-injured group compared with the LPS-injured group
with no treatment (Fig. 3A~C). The results indicate that EGCG
ameliorated the neuronal differentiation of NSCs impaired by LPS
in the adult hippocampal DG at an early stage. In addition, NeuN
(a mature neuronal marker) and BrdU- positive cells at 28 days
were decreased in the LPS-injured group, whereas the number
of double positive cells was increased in the EGCG-treated LPS-
Fig. 5. The effect of EGCG on microglia and proinflammatory cytokines after LPS- induced neuroinflammation. (A) Representative images show
Iba-1-positive cells (green) in the DG on day 1 (upper) and day 3 (lower) after LPS injection. (B) The positive effect of EGCG was quantified by counting
the number of Iba-1-positive cells that was significantly reduced by EGCG (0.5 mg/kg) compared with LPS only (n=5 each, day 1; p=0.0002, day 3;
p=0.0025 respectively, one-way ANOVA). (C) Western blotting analysis was performed to quantify the TLR4, Rel A, active form of Rel A (pRel A), and
b
-actin as a control in the hippocampal DG. The TLR4-NF
k
B pathway was increased by LPS, which was rescued by EGCG (0.5 mg/kg) (n=4 each). (D~G)
Quantitative real-time PCR and ELISA was performed to measure IL-1β, IL-6, and TNF-α in the hippocampus after LPS-induced neuroinflammation (n=4
each). The levels of IL-1
b
, IL-6, and TNF-
a
were suppressed by EGCG treatment in LPS-induced (n=4 each) p<0.05, **p<0.01 compared with the control,
p<0.05 and
★★
p< 0.01 compared with the LPS-injured group, ***p<0.001). Data are expressed as the mean±SEM. Scale bar in A, 200
m
m (upper) and
in an inset, 100
m
m (lower).
EGCG in adult neurogenesis impaired neuroinflammation in mouse
Korean J Physiol Pharmacol 2016;20(1):41-51www.kjpp.net
49
injured group compared with the vehicle-treated LPS-injured
group (Fig. 4A~C). It is indicating that EGCG had a recovery
effect on the neuronal differentiation of adult NSCs at a late
stage, which was impaired by LPS induced neuroinflammation.
These results demonstrate that the neuronal differentiation of
adult NSCs in the DG is rescued by the injection of EGCG into
mice with LPS-induced neuroinflammation. Our data also was
supported the beneficial effect of EGCG on adult neurogenesis
by showing improved proliferation and differentiation of adult
mouse hippocampal NSCs in the DG under normal conditions
[48].
TLRs are expressed in immune and non-immune cells and
are crucial for innate immune responses. For instance, TLR4 is
activated by LPS-mediated inflammation in immune cells [49],
and is also found in the central nervous system for regulating
neurogenesis [50]. A previous report showed that EGCG has
anti-inflammatory activity via the downregulation of TLR4
expression [51]. Thus, the recruitment of activated microglia
was evaluated in LPS-induced neuroinflammation by showing
the number of Iba-1-positive microglial cells. LPS activated the
concentrating microglia in the brain, however, the activated
microglia were suppressed by EGCG treatment, which modified
the TLR4-mediated NF-
k
B pathway and proinflammatory
cytokines (Fig. 5A~G). The results suggest a potential role of
EGCG in significantly improving adult neurogenesis impaired
by neuroinflammation, and show that the beneficial effect
was associated with the downregulation of proinflammatory
cytokines through the modulation of microglial activity.
It is still unclear whether EGCG decreases the level of oxidative
stress in neuroinflammation for its improvement of adult
neurogenesis. Published reports have demonstrated that microglia
produce reactive oxygen species (ROS) in PD [52,53], and
in vivo
and
in vitro
models of AD have shown that EGCG modulates the
cellular mechanisms of neuroprotection and neurorestoration
through the activation of the protein kinase C pathway for
improving cell survival and apoptosis and antioxidant function
against ROS [54,55]. These works support that EGCG may be
involved in the regulation of ROS for rescuing adult neurogenesis
affected by neuroinflammation. To effectively address these
questions, studies regarding the molecular mechanisms of EGCG
in relation to ROS using primary cultures of adult NSCs are
required in the future.
In conclusion, we suggest that the administration of EGCG
was beneficial for improving the proliferation, survival rate, and
neuronal differentiation of adult NSCs in the DG by suppressing
the activity of microglia and the TLR4-mediated NF-
k
B path-
way in impaired neurogenesis caused by neuroinflammation.
Therefore, EGCG may be a potential therapeutic agent for
neuroinflammatory diseases.
ACKNOWLEDGEMENTS
This work was supported by the National Research Foundation
of Korea (NRF) grant funded by the Korea government (MSIP)
(2011-0030121), (2014R1A2A2A01007582) and by a grant
(CRI 12052-22) from Chonnam National University Hospital
Biomedical Research Institute.
SUPPLEMENTARY MATERIALS
Supplementary data including one figure can be found with
this article online at http://pdf.medrang.co.kr/paper/pdf/Kjpp/
Kjpp020-01-06-s001.pdf.
REFERENCES
1. Ekdahl CT, Kokaia Z, Lindvall O. Brain inflammation and
adult neurogenesis: the dual role of microglia.
Neuroscience.
2009;158:1021-1029.
2. Polazzi E, Monti B. Microglia and neuroprotection: from in vitro
studies to therapeutic applications.
Prog Neurobiol.
2010;92:293-
315.
3. Dheen ST, Kaur C, Ling EA. Microglial activation and its
implications in the brain diseases.
Curr Med Chem.
2007;14:1189-
1197.
4. Lourbopoulos A, Errk A, Hellal F. Microglia in action: how aging
and injury can change the brain's guardians.
Front Cell Neurosci.
2015;9:54.
5. Casano AM, Peri F. Microglia: multitasking specialists of the
brain.
Dev Cell.
2015;32:4 69- 477.
6. Delpech JC, Madore C, Nadjar A, Joffre C, Wohleb ES, La
S. Microglia in neuronal plasticity: Influence of stress.
Neu-
ropharmacology.
2015;96:19-28.
7. Bilimoria PM, Stevens B. Microglia function during brain de-
velopment: New insights from animal models.
Brain Res.
2015;
1617:7-17.
8. Jeong JW, Lee HH, Han MH, Kim GY, Kim WJ, Choi YH. Anti-
inflammatory effects of genistein via suppression of the toll-like
receptor 4-mediated signaling pathway in lipopolysaccharide-
stimulated BV2 microglia.
Chem Biol Interact.
2014;212:30-39.
9. Su X, Chen Q, Chen W, Chen T, Li W, Li Y, Dou X, Zhang Y, Shen Y,
Wu H, Yu C. Mycoepoxydiene inhibits activation of BV2 microglia
stimulated by lipopolysaccharide through suppressing NF-κB,
ERK 1/2 and toll-like receptor pathways.
Int Immunopharmacol.
2014;19:88-93.
10. Park KW, Lee DY, Joe EH, Kim SU, Jin BK. Neuroprotective role
of microglia expressing interleukin-4.
J Neurosci Res.
2005;81:397-
402.
11. Zhang Q, Yuan L, Liu D, Wang J, Wang S, Zhang Q, Gong Y, Liu
H, Hao A, Wang Z. Hydrogen sulfide attenuates hypoxia-induced
neurotoxicity through inhibiting microglial activation.
Pharmacol
Res.
2014;84:32-44.
12. Lehnardt S. Innate immunity and neuroinflammation in the
50
http://dx.doi.org/10.4196/kjpp.2016.20.1.41Korean J Physiol Pharmacol 2016;20(1):41-51
Seong KJ et al
CNS: the role of microglia in Toll-like receptor-mediated neuronal
injury.
Glia.
2010;58:253-263.
13. Matsuda T, Murao N, Katano Y, Juliandi B, Kohyama J, Akira
S, Kawai T, Nakashima K. TLR9 signalling in microglia atte-
nuates seizure-induced aberrant neurogenesis in the adult hip-
pocampus.
Nat Commun.
2015;6:6514.
14. Suhonen JO, Peterson DA, Ray J, Gage FH. Differentiation of
adult hippocampus-derived progenitors into olfactory neurons in
vivo.
Nature.
1996;383:624-627.
15. Gage FH. Mammalian neural stem cells.
Science.
2000;287:1433-
1438.
16. Suh H, Deng W, Gage FH. Signaling in adult neurogenesis.
Annu
Rev Cell Dev Biol.
2009;25:253-275.
17. Wen S, Li H, Liu J. Dynamic signaling for neural stem cell fate
determination.
Cell Adh Migr.
20 09;3:107-117.
18. Brown J, Cooper-Kuhn CM, Kempermann G, Van Praag H, Winkler
J, Gage FH, Kuhn HG. Enriched environment and physical activity
stimulate hippocampal but not olfactory bulb neurogenesis.
Eur J
Neurosci.
2003;17:2042-2046.
19. van Praag H, Kempermann G, Gage FH. Running increases
cell proliferation and neurogenesis in the adult mouse dentate
gyrus.
Nat Neurosci.
1999;2:266-270.
20. Drapeau E, Mayo W, Aurousseau C, Le Moal M, Piazza PV, Abrous
DN. Spatial memory performances of aged rats in the water maze
predict levels of hippocampal neurogenesis.
Proc Natl Acad Sci U S
A.
2003;100:14385-14390.
21. Tatebayashi Y, Lee MH, Li L, Iqbal K, Grundke-Iqbal I. The
dentate gyrus neurogenesis: a therapeutic target for Alzheimer's
disease.
Acta Neuropathol.
2003;105:225-232.
22. Abrous DN, Koehl M, Le Moal M. Adult neurogenesis: from
precursors to network and physiology.
Physiol Rev.
2005;85:523-569.
23. Rogers AE, Hafer LJ, Iskander YS, Yang S. Black tea and mammary
gland carcinogenesis by 7,12-dimethylbenz[a]anthracene in rats fed
control or high fat diets.
Carcinogenesis.
1998;19:1269-1273.
24. Zhou H, Chen JX, Yang CS, Yang MQ, Deng Y, Wang H. Gene
regulation mediated by microRNAs in response to green tea
polyphenol EGCG in mouse lung cancer.
BMC Genomics.
2014;15
Suppl 11:S3.
25. Singh BN, Shankar S, Srivastava RK. Green tea catechin, epigallo-
catechin-3-gallate (EGCG): mechanisms, perspectives and clinical
applications.
Biochem Pharmacol.
2011;82:1807-1821.
26. Lecumberri E, Dupertuis YM, Miralbell R, Pichard C. Green tea
polyphenol epigallocatechin-3-gallate (EGCG) as adjuvant in cancer
therapy.
Clin Nutr.
2013;32:894-903.
27. Steinmann J, Buer J, Pietschmann T, Steinmann E. Anti-infective
properties of epigallocatechin-3-gallate (EGCG), a component of
green tea.
Br J Pharmacol.
2013;168:1059-1073.
28. Gould E. How widespread is adult neurogenesis in mammals?
Nat
Rev Neurosci.
2007;8:481-488.
29. Lledo PM, Alonso M, Grubb MS. Adult neurogenesis and functional
plasticity in neuronal circuits.
Nat Rev Neurosci.
2006;7:179-193.
30. Kriegstein A, Alvarez-Buylla A. The glial nature of embryonic and
adult neural stem cells.
Annu Rev Neurosci.
2009;32:149-184.
31. Drew LJ, Fusi S, Hen R. Adult neurogenesis in the mammalian
hippocampus: why the dentate gyrus?
Learn Mem.
2013;20:710-729.
32. Singhal G, Jaehne EJ, Corrigan F, Toben C, Baune BT. Inflam-
masomes in neuroinflammation and changes in brain function: a
focused review.
Front Neurosci.
2014;8:315.
33. Monje ML, Toda H, Palmer TD. Inflammatory blockade restores
adult hippocampal neurogenesis.
Science.
2003;302:1760-1765.
34. Filiou MD, Arefin AS, Moscato P, Graeber MB. 'Neuroinflammation'
differs categorically from inflammation: transcriptomes of Al-
zheimer's disease, Parkinson's disease, schizophrenia and in-
flammatory diseases compared.
Neurogenetics.
2014;15:201-212.
35. Song C, Wang H. Cytokines mediated inflammation and
decreased neurogenesis in animal models of depression.
Prog
Neuropsychopharmacol Biol Psychiatry.
2011;35:760-768.
36. Roy AM, Baliga MS, Katiyar SK. Epigallocatechin-3-gallate induces
apoptosis in estrogen receptor-negative human breast carcinoma
cells via modulation in protein expression of p53 and Bax and
caspase-3 activation.
Mol Cancer Ther.
2005;4:81-90.
37. Abd El Mohsen MM, Kuhnle G, Rechner AR, Schroeter H, Rose S,
Jenner P, Rice-Evans CA. Uptake and metabolism of epicatechin
and its access to the brain after oral ingestion.
Free Radic Biol Med.
2002;33:1693-1702.
38. Lin LC, Wang MN, Tseng TY, Sung JS, Tsai TH. Pharmacokinetics
of (-)-epigallocatechin-3-gallate in conscious and freely moving
rats and its brain regional distribution.
J Agric Food Chem.
2007;55:1517-1524.
39. Li J, Ye L, Wang X, Liu J, Wang Y, Zhou Y, Ho W. (-)-Epigallocatechin
gallate inhibits endotoxin-induced expression of inflammatory
cytokines in human cerebral microvascular endothelial cells.
J
Neuroinflammation.
2012;9:161.
40. Mandel SA, Amit T, Weinreb O, Reznichenko L, Youdim MB.
Simultaneous manipulation of multiple brain targets by green tea
catechins: a potential neuroprotective strategy for Alzheimer and
Parkinson diseases.
CNS Neurosci Ther.
2008;14:352-365.
41. Avramovich-Tirosh Y, Reznichenko L, Mit T, Zheng H, Fridkin
M, Weinreb O, Mandel S, Youdim MB. Neurorescue activity, APP
regulation and amyloid-beta peptide reduction by novel multi-
functional brain permeable iron- chelating- antioxidants, M-30 and
green tea polyphenol, EGCG.
Curr Alzheimer Res.
2007;4:403-411.
42. Herges K, Millward JM, Hentschel N, Infante-Duarte C, Aktas O,
Zipp F. Neuroprotective effect of combination therapy of glatiramer
acetate and epigallocatechin-3-gallate in neuroinflammation.
PLoS
One.
2011;6:e25456.
43. Wu KJ, Hsieh MT, Wu CR, Wood WG, Chen YF. Green tea extract
ameliorates learning and memory deficits in ischemic rats via
its active component polyphenol epigallocatechin-3-gallate by
modulation of oxidative stress and neuroinflammation.
Evid Based
Complement Alternat Med.
2012;2012:163106.
44. Weinreb O, Amit T, Youdim MB. A novel approach of proteomics
and transcriptomics to study the mechanism of action of the
antioxidant-iron chelator green tea polyphenol (-)-epigallocatechin-
3-gallate.
Free Radic Biol Med.
2007;43:546-556.
45. Weinreb O, Mandel S, Amit T, Youdim MB. Neurological me-
chanisms of green tea polyphenols in Alzheimer's and Parkinson's
diseases.
J Nutr Biochem.
2004;15:506-516.
46. Gan L, Meng ZJ, Xiong RB, Guo JQ, Lu XC, Zheng ZW, Deng YP,
Luo BD, Zou F, Li H. Green tea polyphenol epigallocatechin-3-
gallate ameliorates insulin resistance in non-alcoholic fatty liver
disease mice.
Acta Pharmacol Sin.
2015;36:597-605.
47. Wang Y, Li M, Xu X, Song M, Tao H, Bai Y. Green tea epigal-
locatechin-3-gallate (EGCG) promotes neural progenitor cell pro-
EGCG in adult neurogenesis impaired neuroinflammation in mouse
Korean J Physiol Pharmacol 2016;20(1):41-51www.kjpp.net
51
liferation and sonic hedgehog pathway activation during adult
hippocampal neurogenesis.
Mol Nutr Food Res.
2012;56:1292-1303.
48. Yoo KY, Choi JH, Hwang IK, Lee CH, Lee SO, Han SM, Shin HC,
Kang IJ, Won MH. (-)-Epigallocatechin-3-gallate increases cell
proliferation and neuroblasts in the subgranular zone of the dentate
gyrus in adult mice.
Phytother Res.
2010;24:1065-1070.
49. Wang W, Deng M, Liu X, Ai W, Tang Q, Hu J. TLR4 activation
induces nontolerant inflammatory response in endothelial
cells.
Inflammation.
2011;34:509-518.
50. Rolls A, Shechter R, London A, Ziv Y, Ronen A, Levy R, Schwartz M.
Toll-like receptors modulate adult hippocampal neurogenesis.
Nat
Cell Biol.
2007;9:1081-1088.
51. Hong Byun E, Fujimura Y, Yamada K, Tachibana H. TLR4 sig-
naling inhibitory pathway induced by green tea polyphenol epi-
gallocatechin-3-gallate through 67-kDa laminin receptor.
J Im-
munol.
2010;185:33-45.
52. Kumar P, Kumar A. Protective effects of epigallocatechin gal-
late following 3-nitropropionic acid-induced brain damage:
possible nitric oxide mechanisms.
Psychopharmacology (Berl).
2009;207:257-270.
53. Mao H, Fang X, Floyd KM, Polcz JE, Zhang P, Liu B. Induction
of microglial reactive oxygen species production by the
organochlorinated pesticide dieldrin.
Brain Res.
2007;1186:267-274.
54. Levites Y, Amit T, Youdim MB, Mandel S. Involvement of protein
kinase C activation and cell survival/ cell cycle genes in green tea
polyphenol (-)-epigallocatechin 3-gallate neuroprotective action.
J
Biol Chem.
2002;277:30574-30580.
55. Weinreb O, Amit T, Mandel S, Youdim MB. Neuroprotective
molecular mechanisms of (-)-epigallocatechin-3-gallate: a reflective
outcome of its antioxidant, iron chelating and neuritogenic
properties.
Genes Nutr.
2009;4:283-296.
Supplementary figure 1. The schematic representation of experimental research design. (A) LPS (black arrow), EGCG (red arrows, 3
times with 6 hours interval), and BrdU (gray arrow) injection were done in the time course and the animals were sacrificed on day 1, 3, and 5
for immunohistochemical analysis to the quantification of NSC proliferation and immature differentiation. (B) LPS (black arrow), EGCG (red
arrows), and 5 consecutive BrdU (gray arrows) injection daily were done in the time course and the animals were sacrificed on day 5 and 28 for
immunohistochemical analysis to the quantification of NSC cell survival and mature differentiation. LPS, lipopolysaccharide, EGCG, epigallocatechin-
3-gallate, BrdU, bromodeoxyuridine, injt, injection, and Sac, scarified.

Supplementary resource (1)

... In a study involving mice with lipopolysaccharide-induced neuroinflammation, EGCG at a concentration of 0.5 mg/kg was found to restore neurogenesis in the DG. Additionally, EGCG treatment attenuated the production of pro-inflammatory cytokines induced by lipopolysaccharides by modulating the TLR4-NF-κB pathway [123]. In another study, a two-week treatment with EGCG at varying doses (0, 0.625, 1.25, 2.5, 5, and 10 mg/kg) dose-dependently increased the survival of NSCs. ...
... Selected bioactive compounds and molecular pathways leading to postnatal neurogenesis stimulation are listed in Table 1. survival and neuronal differentiation of adult hippocampal precursor cells [124] 2.5 mg/kg male Balb/C mice increased cell survival increase in DCX+ cells increase in hippocampal levels of phospho-Akt [124] 0.5 mg/kg male C57BL/6 mice (LPS-induced neuroinflammation model) stimulation of neurogenesis in DG suppression of the activity of microglia and the TLR4-mediated NF-kB pathway (anti-inflammatory activity) [123] quercetin 25 mg/kg 3xTg-AD mice Alzheimer model reduction in taupathy neuronal population protection improvement in spatial learning [133] 40 mg/kg male Wistar rats Alzheimer model increase in neurogenesis increase in the number of proliferating NSCs/progenitor cells increase in DCX+ cells improvement in the number of BrdU/NeuN+ cells promotes the expression of BDNF, NGF, CREB and EGR-1 genes in adult rat DG [134] 100 mg/kg 3xTg-AD mice Alzheimer model increase in neurogenesis enhances the levels of CREB and BDNF β-amyloidosis reduction and tendency to decrease tauopathy [135] atranorin 5 µM Neuro2A neurite outgrowth neurotrophic activity increase in BDNF and NGF [126] 10 mg/kg healthy Wistar rats changed some forms of behavior promotes neurogenesis [125] 10 mg/kg Sprague Dawley rats depression model changed some forms of behavior promotes neurogenesis [136] Rg1 2.5-20 mg/kg C57BL/6J mice depression model antidepressant activity changed some forms of behavior pERK1/2 and pCREB pathway [132] 20 mg/kg Sprague Dawley rats aging model neurogenesis stimulation improvement in cognitive capacity protect NSCs/progenitor cells by increased level of SOX-2 expression reduced astrocytes activation by decrease level of Aeg-1 expression anti-inflammatory activity enhances the activity of antioxidant enzymes GSH-Px and SOD [131] Aeg-1, astrocyte elevated gene-1; AKT, protein kinase B; AMP, AMP-activated protein kinase; ApoE, apolipoprotein E; BDNF, brain-derived neurotrophic factor; BrdU, bromodeoxyuridine; CR, calretinin; CREB, cyclic AMPresponsive element-binding protein 1; DCX, doublecortin; DG, DG; EGR-1, early growth response protein 1; GSH-Px, glutathione peroxidase; Hat1, histone acetyltransferase 1; Hdac4, histone deacetylase 4; Hes5, Hes family BHLH transcription factor 5; JAK/STAT, Janus kinase-signal transducer and activator of transcription; MBP, myelin basic protein; NeuN, neuronal nuclei; NGF, nerve growth factor; NSCs, neural stem cells; pERK1/2, phospho extracellular signal-regulated kinase 1/2; PI3K/AKT, phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT); Sox2, SRY-Box transcription factor 2; SOD, superoxide dismutase; TLR4-NF-κB, Toll-like receptor 4/nuclear factor kappa B; Wnt7a, Wnt family member 7A. ...
Article
Full-text available
Since postnatal neurogenesis was revealed to have significant implications for cognition and neurological health, researchers have been increasingly exploring the impact of natural compounds on this process, aiming to uncover strategies for enhancing brain plasticity. This review provides an overview of postnatal neurogenesis, neurogenic zones, and disorders characterized by suppressed neurogenesis and neurogenesis-stimulating bioactive compounds. Examining recent studies, this review underscores the multifaceted effects of natural compounds on postnatal neurogenesis. In essence, understanding the interplay between postnatal neurogenesis and natural compounds could bring novel insights into brain health interventions. Exploiting the therapeutic abilities of these compounds may unlock innovative approaches to enhance cognitive function, mitigate neurodegenerative diseases, and promote overall brain well-being.
... Besides peripheral immune cells, TLR4 is expressed by all brain cells (Bsibsi et al. 2002;Kigerl et al. 2007;Rolls et al. 2007). Thus, many studies demonstrated that TLR4 activation caused neurogenesis alterations (Rolls et al. 2007;Seong et al. 2016) which could lead to schizophrenia-like behavior in animal's offspring (Zhu et al. 2014;Talukdar et al. 2021). Moreover, a large body of evidence has demonstrated that TLR4 expression and activation are associated with cognitive deficits and modulated by antipsychotic treatment in SCZ and BD patients (McKernan et al. 2011;García-Bueno et al. 2016;Wieck et al. 2016;Kéri et al. 2017;Kozłowska et al. 2019;Balaji et al. 2020). ...
Article
Full-text available
Immune dysregulation has been widely described in the pathophysiology of schizophrenia (SCZ) and bipolar disorder (BD). Particularly, TLR4-altered activation was proposed as one of the underlying processes of psychosis onset. Since TLR4 activation was altered by T399I and D299G polymorphisms, we hypothesized that those variants could present common genetic factors of SCZ and BD. A total of 293 healthy volunteers and 335 psychotic patients were genotyped using PCR–RFLP. Genotype, allele, and haplotype distribution between controls and patients were evaluated according to clinical parameters. Statistical analyses were adjusted by logistic regression. In dominant model, T399I CT + TT and allele frequency were significantly higher in controls compared to psychotic population (p = 0.004, p = 0.002, respectively), SCZ (p = 0.02, p = 0.01, respectively), and BD (p = 0.03, p = 0.02, respectively). Similarly, D299G AG + GG and allele frequency were significantly higher in controls compared to psychotic population (p = 0.04, p = 0.04, respectively) and SCZ (p = 0.04, p = 0.03, respectively). T399I CT + TT and T allele were overrepresented in controls compared to paranoid subgroup (Padjusted = 0.04, p = 0.04, respectively) and type I BD (p = 0.04). Moreover, T399I and D299G were less prevalent in SCZ late-onset age (p = 0.03, p = 0.02, respectively). TA haplotype was associated with protection from psychoses (p = 0.02) and particularly from schizophrenia (p = 0.04). In conclusion, TLR4 polymorphisms could present a preventive genetic background against psychoses onset in a Tunisian population. While T399I could be associated with protection against SCZ and BD, presenting an overlapping genetic factor between those psychoses, D299G was suggested to be associated with protection only from schizophrenia.
... Moreover, TLR4 is one of the cell membrane receptors of LPS [26]. It could transmit signaling to myeloid differentiation factor 88 (MyD88) and then activate the downstream NF-κB or MAPKs [27]. Thus, TLR4/NF-κB signaling axis is the main inflammatory cascade involved in our researches for illuminating the functions of TEC in SCI. ...
Article
Full-text available
Since tectorigenin has been reported to possess anti-inflammation, redox balance restoration, and anti-apoptosis properties, we determine to unravel whether tectorigenin has potential in alleviating spinal cord injury (SCI). Herein, PC12 cells were induced by lipopolysaccharide (LPS) to establish in vitro SCI models. The cell viability and apoptosis were detected through cell counting kit-8 and flow cytometry assays. The caspase-3/8/9 content was measured by colorimetric method. Western blot was conducted to quantify the expressions of cleaved caspse-3/8/9, IGFBP6, TLR4, IκBα, p-IκBα, RELA proto-oncogene, p65, and p-p65. Enzyme-linked immunosorbent assay and real-time quantitative polymerase chain reaction were carried out to quantitate expressions of IGFBP6, interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α). SwissTargetPrediction and GSE21497 database were utilized to predict the potential therapeutic targets of tectorigenin. Comparison of IGFBP6 expression in SCI tissues and normal tissues was analyzed by GEO2R. Our study found that LPS induced the declined cell viability, elevated cell apoptosis, upregulation of caspase-3/8/9, cleaved caspase-3/8/9, IL-1β, IL-6, TNF-α, IGFBP6, and TLR4, and the activation of IκBα and p65 in PC12 cells. Tectorigenin reversed the above effects of LPS. IGFBP6 was predicted to be the potential therapeutic target of tectorigenin and was overexpressed in SCI tissues. Notably, IGFBP6 overexpression offset the effects of tectorigenin on PC12 cells. In conclusion, tectorigenin could alleviate the LPS-induced apoptosis, inflammation, and activation of NF-κB signaling in SCI cell models via inhibiting IGFBP6.
Article
Full-text available
Aging significantly impacts several age-related neurological problems, such as stroke, brain tumors, oxidative stress, neurodegenerative diseases (Alzheimer's, Parkinson's, and dementia), neuroinflammation, and neurotoxicity. Current treatments for these conditions often come with side effects like hallucinations, dyskinesia, nausea, diarrhea, and gastrointestinal distress. Given the widespread availability and cultural acceptance of natural remedies, research is exploring the potential effectiveness of plants in common medicines. The ancient medical system used many botanical drugs and medicinal plants to treat a wide range of diseases, including age-related neurological problems. According to current clinical investigations, berries improve motor and cognitive functions and protect against age-related neurodegenerative diseases. Additionally, berries may influence signaling pathways critical to neurotransmission, cell survival, inflammation regulation, and neuroplasticity. The abundance of phytochemicals in berries is believed to contribute to these potentially
Article
Full-text available
Aging significantly impacts several age-related neurological problems, such as stroke, brain tumors, oxidative stress, neurodegenerative diseases (Alzheimer’s, Parkinson’s, and dementia), neuroinflammation, and neurotoxicity. Current treatments for these conditions often come with side effects like hallucinations, dyskinesia, nausea, diarrhea, and gastrointestinal distress. Given the widespread availability and cultural acceptance of natural remedies, research is exploring the potential effectiveness of plants in common medicines. The ancient medical system used many botanical drugs and medicinal plants to treat a wide range of diseases, including age-related neurological problems. According to current clinical investigations, berries improve motor and cognitive functions and protect against age-related neurodegenerative diseases. Additionally, berries may influence signaling pathways critical to neurotransmission, cell survival, inflammation regulation, and neuroplasticity. The abundance of phytochemicals in berries is believed to contribute to these potentially neuroprotective effects. This review aimed to explore the potential benefits of berries as a source of natural neuroprotective agents for age-related neurological disorders.
Preprint
Full-text available
Sepsis-associated encephalopathy (SAE), one of the common complications of sepsis, is associated with higher ICU mortality, prolonged hospitalization and long-term cognitive decline. Sepsis can induce neuroinflammation, which negatively affects hippocampal neurogenesis. Dexmedetomidine has been shown to protect against SAE. However, the potential mechanism remains unclear. In this study, we added lipopolysaccharide (LPS)-stimulated astrocytes-conditioned media (LPS-CM) to neural stem cells (NSCs) culture, which were pretreated with dexmedetomidine in the presence or absence of the α2-adrenoceptor antagonist yohimbine or the α2A-adrenoceptor antagonist BRL-44408. LPS-CM impaired the neurogenesis of NSCs, characterized by decreased proliferation, enhanced gliogenesis and declined viability. Dexmedetomidine alleviated LPS-CM-induced impairments of neurogenesis in a dose-dependent manner. Yohimbine, as well as BRL-44408, reversed the effects of dexmedetomidine. We established a mouse model of SAE by cecal ligation and perforation (CLP). CLP induced astrocyte-related neuroinflammation and hippocampal neurogenesis deficits, companied with learning and memory decline, which was reversed by dexmedetomidine. The effect of dexmedetomidine was blocked by BRL-44408. Collectively, our findings support the conclusion that dexmedetomidine can protect against SAE, likely mediated by the combination of inhibiting neuroinflammation via astrocytic α2A-adrenoceptor with attenuating neuroinflammation-induced hippocampal neurogenesis deficits via NSCs α2A-adrenoceptor.
Article
Full-text available
Pathological conditions such as epilepsy cause misregulation of adult neural stem/progenitor populations in the adult hippocampus in mice, and the resulting abnormal neurogenesis leads to impairment in learning and memory. However, how animals cope with abnormal neuro-genesis remains unknown. Here we show that microglia in the mouse hippocampus attenuate convulsive seizure-mediated aberrant neurogenesis through the activation of Toll-like receptor 9 (TLR9), an innate immune sensor known to recognize microbial DNA and trigger inflammatory responses. We found that microglia sense self-DNA from degenerating neurons following seizure, and secrete tumour necrosis factor-a, resulting in attenuation of aberrant neurogenesis. Furthermore, TLR9 deficiency exacerbated seizure-induced cognitive decline and recurrent seizure severity. Our findings thus suggest the existence of bidirectional communication between the innate immune and nervous systems for the maintenance of adult brain integrity.
Article
Full-text available
Neuroinflammation, the inflammatory response in the central nervous system (CNS), is a major determinant of neuronal function and survival during aging and disease progression. Microglia, as the resident tissue-macrophages of the brain, provide constant support to surrounding neurons in healthy brain. Upon any stress signal (such as trauma, ischemia, inflammation) they are one of the first cells to react. Local and/or peripheral signals determine microglia stress response, which can vary within a continuum of states from beneficial to detrimental for neuronal survival, and can be shaped by aging and previous insults. In this review, we discuss the roles of microglia upon an ischemic or traumatic injury, and give our perspective how aging may contribute to microglia behavior in the injured brain. We speculate that a deeper understanding of specific microglia identities will pave the way to develop more potent therapeutics to treat the diseases of aging brain.
Article
Full-text available
Epigallocatechin-3-gallate (EGCG) has been demonstrated to inhibit cancer in experimental studies through its antioxidant activity and modulations on cellular functions by binding specific proteins. We demonstrated previously that EGCG upregulates the expression of microRNA (i.e. miR-210) by binding HIF-1α, resulting in reduced cell proliferation and anchorage-independent growth. However, the binding affinities of EGCG to HIF-1α and many other targets are higher than the EGCG plasma peak level in experimental animals administered with high dose of EGCG, raising a concern whether the microRNA regulation by HIF-1α is involved in the anti-cancer activity of EGCG in vivo. We employed functional genomic approaches to elucidate the role of microRNA in the EGCG inhibition of tobacco carcinogen-induced lung tumors in A/J mice. By analysing the microRNA profiles, we found modest changes in the expression levels of 21 microRNAs. By correlating these 21 microRNAs with the mRNA expression profiles using the computation methods, we identified 26 potential targeted genes of the 21 microRNAs. Further exploration using pathway analysis revealed that the most impacted pathways of EGCG treatment are the regulatory networks associated to AKT, NF-κB, MAP kinases, and cell cycle, and the identified miRNA targets are involved in the networks of AKT, MAP kinases and cell cycle regulation These results demonstrate that the miRNA-mediated regulation is actively involved in the major aspects of the anti-cancer activity of EGCG in vivo.
Article
Full-text available
Recent literature has pointed to the existence of inflammasome-mediated inflammatory pathways in central nervous system (CNS) disorders and associated changes in behavior. Neuroinflammation, which is an innate immune response in the CNS against harmful and irritable stimuli such as pathogens and metabolic toxic waste, as well as to chronic mild stress, is mediated by protein complexes known as inflammasomes. Inflammasomes activate pro-inflammatory caspases 1 and 5, which then cleave the precursor forms of pro-inflammatory cytokines IL-1β, IL-18, and IL-33 into their active forms. These pro-inflammatory cytokines have been shown to promote a variety of innate immune processes associated with infection, inflammation, and autoimmunity, and thereby play an instrumental role in the instigation of neuroinflammation during old age and subsequent occurrence of neurodegenerative diseases, cognitive impairment, and dementia. In particular, NLRP inflammasomes may also have a role in the etiologies of depression, Alzheimer's disease (AD) and in metabolic disorders, such as Type II diabetes, obesity and cardiovascular diseases that have been shown to be co-morbid with psychiatric illnesses. It has been reported that while these inflammasomes may be activated through TNF-α dependent pathways, other cytokines, like IFN-γ, may assist in inhibiting their activation and thus delay disease progression. Furthermore, some other cytokines, including IL-6, may not have a direct role in inflammasome-mediated diseases. An array of recent research suggests that NLRP inflammasomes targeted therapies could be used for alleviating neuroinflammation and for treatment of associated psychiatric illnesses, although this still remains a challenge and necessitates further extensive research. This review examines the complex inflammatory signaling pathways involved in the activation of NLRP inflammasomes and the role they play in promoting neuroinflammation and subsequent behavioral changes.
Article
Epidemiological studies suggest that tea may reduce cancer risk, and in laboratory rodents, chemopreventive effects of tea or purified extracts of tea have been demonstrated in lung, gastrointestinal tract and skin. There is some evidence of chemoprevention by tea in the mammary gland, but the data are not conclusive. In order to evaluate more fully the possible influence of black tea on 7,12-dimethylbenz[a]anthracene (DMBA)-induced mammary gland tumors in the female S‐D (Sprague‐Dawley) rat, three large studies were performed: experiment 1, tumorigenesis in rats fed AIN-76A diet and given 25 mg/kg DMBA and 1.25 or 2.5% whole tea extract or water to drink; experiment 2, tumorigenesis in rats given 15 mg/kg DMBA and the same diet and fluids as in experiment 1; experiment 3, tumorigenesis in rats fed control or HF (high fat, corn oil) diet and given 15 mg/kg DMBA and 2% tea or water to drink. Tea was given throughout the experiment; DMBA was given by gastric gavage at 8 weeks of age. There was no consistent effect of tea on tumorigenesis in rats fed AIN-76A diet; there was, however, evidence in experiment 3 of a reduction of tumorigenesis by tea in rats fed the HF diet. In experiment 3, rats fed the HF diet and given water showed the expected increase in tumor burden (number and weight) compared with rats fed control diet. However, rats fed the HF diet and given 2% tea showed no increase in tumor burden; their tumor burden was significantly lower than in rats fed the HF diet and given water (P , 0.01) and was not different from rats fed control diet and given water or tea. In addition, in experiment 3, the number of malignant tumors per tumor-bearing rat was increased by the HF diet in water-drinking rats (P , 0.01) but not in tea-drinking rats. Therefore, it appears that tea partially blocked the promotion of DMBA-induced mammary tumorigenesis by the HF diet.
Article
Epigallocatechin-3-gallate (EGCG) is a major polyphenol in green tea. In this study, we investigated the effects of EGCG on insulin resistance and insulin clearance in non-alcoholic fatty liver disease (NAFLD) mice. Mice were fed on a high-fat diet for 24 weeks. During the last 4 weeks, the mice were injected with EGCG (10, 20 and 40 mg·kg(-1)·d(-1), ip). Glucose tolerance, insulin tolerance and insulin clearance were assessed. After the mice were euthanized, blood samples and tissue specimens were collected. Glucose-stimulated insulin secretion was examined in isolated pancreatic islets. The progression of NAFLD was evaluated histologically and by measuring lipid contents. Insulin-degrading enzyme (IDE) protein expression and enzyme activity were detected using Western blot and immunocapture activity assays, respectively. The high-fat diet significantly increased the body weight and induced grade 2 or 3 liver fatty degeneration (steatosis, lobular inflammation and ballooning) accompanied by severe hyperlipidemia, hyperglycemia, hyperinsulinemia and insulin resistance in the model mice. Administration of EGCG dose-dependently ameliorated the hepatic morphology and function, reduced the body weight, and alleviated hyperlipidemia, hyperglycemia, hyperinsulinemia and insulin resistance in NAFLD mice. Furthermore, EGCG dose-dependently enhanced insulin clearance and upregulated IDE protein expression and enzyme activity in the liver of NAFLD mice. EGCG dose-dependently improves insulin resistance in NAFLD mice not only by reducing body weight but also through enhancing the insulin clearance by hepatic IDE. The results suggest that IDE be a potential drug target for the treatment of NAFLD.
Article
Microglia are macrophages that colonize the brain during development to establish a resident population of professional phagocytes that protect against invading pathogens and contribute to brain development and homeostasis. As such, these cells sit at the interface between immunology and neurobiology. In addition to their key roles in brain physiology, microglia offer a great opportunity to address central questions in biology relating to how migrating cells find their positions in the embryo, adopt a behavior that is appropriate for that position, and interact with their local environment. We aim, in this review, to survey key recent advances in microglial research. Copyright © 2015 Elsevier Inc. All rights reserved.
Article
The role of microglia in healthy brains is just beginning to receive notice. Recent studies have revealed that these phagocytic cells control the patterning and wiring of the developing central nervous system (CNS) by regulating, amongst many other processes, programmed cell death, activity-dependent synaptic pruning and synapse maturation. Microglia also play important roles in the mature brain and have demonstrated effects on behavior. Converging evidence from human and mouse studies together raise questions as to the role of microglia in disorders of brain development such as autism, schizophrenia or obsessive–compulsive disorder. In this review, we summarize a number of major findings regarding the role of microglia in brain development and highlight some key questions and avenues for future study.This article is part of a Special Issue entitled SI: Neuroimmunology in Health And Disease.