hermawati2019

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

Received: 29 January 2019

| Revised: 1 August 2019


| Accepted: 15 August 2019

DOI: 10.1111/ejn.14556

RESEARCH REPORT

Chlorogenic acid ameliorates memory loss and hippocampal cell


death after transient global ischemia

Ery Hermawati1,2 | Nur Arfian3 | Mustofa Mustofa4 | Ginus Partadiredja5

1
Doctoral Program, Faculty of Medicine,
Public Health and Nursing, Universitas
Abstract
Gadjah Mada, Yogyakarta, Indonesia Chlorogenic acid (CGA) is known to have antioxidant potentials, yet the effect of
2
Department of Physiology, Faculty CGA on brain ischemia has not been sufficiently understood. Brain ischemia such as
of Medicine, Tanjungpura University,
transient global ischemia disrupts many areas of the brain of rats, including the hip-
Pontianak, West Kalimantan, Indonesia
3 pocampus. Male Wistar rats were randomly assigned into five groups, that is, sham‐
Department of Anatomy, Faculty
of Medicine, Public Health and operated (SO), bilateral common carotid occlusion (BCCO), and BCCO+ 15, 30,
Nursing, Universitas Gadjah Mada, and 60 mg/kg bw CGA groups (CGA15, CGA30, and CGA60, respectively). Brain
Yogyakarta, Indonesia
4
ischemia was induced in Wistar rats with BCCO for 20 min followed by intraperito-
Department of Pharmacology and Therapy,
Faculty of Medicine, Public Health and neal injection of CGA. The rats were examined for the spatial memory in a Morris
Nursing, Universitas Gadjah Mada, water maze test on the 3rd day and were euthanized on the 10th day after BCCO. The
Yogyakarta, Indonesia
total number of pyramidal cells was estimated, and the mRNA expressions of Bcl2,
5
Department of Physiology, Faculty
Bax, caspase‐3, SOD2, SOD1, GPx, ET‐1, eNOS, CD31, and VEGF‐A were meas-
of Medicine, Public Health and
Nursing, Universitas Gadjah Mada, ured. The BCCO group spent less time and distance in the target quadrant than any
Yogyakarta, Indonesia other group in the spatial memory retention test. The CA1 pyramidal cell numbers in
Correspondence
the BCCO and CGA15 groups were lower than in the CGA30 and CGA60 groups.
Ginus Partadiredja, Department of Physiology, The mRNA expressions of Bcl2, SOD2, and CD31 in the BCCO group were lower
Faculty of Medicine, Public Health and than in the CGA15, CGA30, and CGA60 groups. The ET‐1 expression was higher
Nursing, Universitas Gadjah Mada,
Yogyakarta, Indonesia. in the BCCO and CGA15 groups than in the SO, CGA30, and CGA60 groups. CGA
Emails: gpartadiredja@yahoo.com and improves the spatial memory and prevents the CA1 pyramidal cell death after BCCO
gpartadiredja@ugm.ac.id
by increasing Bcl2, SOD2, and CD31 expressions and decreasing ET‐1 expression.
Funding information
Ministry of Research, Technology, and KEYWORDS
Higher Education, Indonesia, Grant/Award chlorogenic acid, hippocampal pyramidal cells, spatial memory, stereology, transient global ischemia
Number: 0299/E3/2016
The peer review history for this article is
available at https​://publo​ns.com/
publo​n/10.1111/EJN.14556​

Abbreviations: AIF, apoptosis‐inducing factor; BCCO, bilateral common carotid occlusion; CA1, cornu ammonis area 1; CA2, cornu ammonis area 2;
CA3, cornu ammonis area 3; CE, coefficient of error; CGA, chlorogenic acid; CVbiol, biological coefficient of variation; CV, coefficient of variation;
eNOS, endothelial nitric oxide synthase; ET‐1, endothelin‐1; FOV, field of view; GPx, glutathione peroxidase; IR injury, ischemia/reperfusion injury;
MWM, Morris water maze; PBS, phosphate‐buffered saline; ROS, reactive oxygen species; RSEM, rabbit small clot embolic stroke model; SOD, superoxide
dismutase; SO, sham‐operated; TGI, transient global ischemia; VEGF‐A, vascular endothelial growth factor‐A.

Edited by Eilis Dowd.

Eur J Neurosci. 2019;00:1–19. wileyonlinelibrary.com/journal/ejn © 2019 Federation of European Neuroscience Societies | 1


and John Wiley & Sons Ltd
2
|    HERMAWATI et al.

1 | IN TRO D U C T ION 2011). The present study aimed at investigating the possible
beneficial effects of CGA on treating the impairment of spatial
One of the most vulnerable areas of the brain toward isch- memory function as well as deficits of hippocampal neurons
emia is the hippocampus (Sanderson, Reynolds, Kumar, and vascular responses following transient global ischemia.
Przyklenk, & Huttemann, 2013). A brain ischemia condi-
tion such as transient global ischemia (TGI) may lead to
hippocampal pyramidal cell death, especially in the CA1
2 | M ATERIAL S AND M ETHOD S
region. Such a detrimental consequence has been known as
2.1 | Animals, BCCO model, and CGA
delayed neuronal death (Inagaki & Etgen, 2013; Movassaghi
treatment
et al., 2012; Quintard et al., 2011). Hippocampal neuronal
death also impairs the spatial memory function (Andersen, The animal handling and experimental procedures were ap-
Morris, Amaral, Bliss, & O'Keefe, 2007; Bear, Connors, & proved by the Ethical Committee of the Faculty of Medicine,
Paradiso, 2007; Kandel, Schwartz, & Jessel, 2000; Purves Public Health and Nursing, Universitas Gadjah Mada (ap-
et al., 2004). proval number: KE/FK/1239/EC/2015). Fifty‐five male
The underlying culprit of neuronal damage in ischemic Wistar rats, aged 3–5 months and weighing 200–350 g, were
conditions is oxidative stress (Movassaghi et al., 2012; obtained from the Animal House (LPPT) and the Department
Selakovic, Korenic, & Radenovic, 2011), which is due to the of Pharmacology and Therapy, Faculty of Medicine, Public
increased production of free radicals (Buch, Patel, Ranpariya, Health and Nursing, Universitas Gadjah Mada. The rats were
Sheth, & Parmar, 2012). The increased level of free radicals housed in cages under the natural light–dark cycle of 12:12 hr.
activates antioxidant enzymes including superoxide dis- The rats were allowed to access food pellets and water ad libi-
mutase (SOD) and glutathione peroxidase (GPx) (Mehta, tum during the experiment. The room temperature and humid-
Manhas, & Raghubir, 2007). At one stage, an imbalance be- ity were at ranges of 26–31°C and 70%–90%, respectively.
tween oxidant and antioxidant levels may occur and lead to The animals were randomly divided into five groups,
the cellular apoptosis accompanied by a reduction of anti‐ that is, SO group, which was the sham‐operated group;
apoptotic signaling, such as the BCL‐2 family in the mito- BCCO group, which was the group undergoing bilateral
chondria (Sasi, Hwang, Jaboin, Csiki, & Lu, 2009). common carotid occlusion (BCCO); and CGA15, CGA30,
Brain ischemia may also lead to vascular reactions in the and CGA60 groups, which were the groups undergoing
form of vascular remodeling (Gibbons & Dzau, 1994). The BCCO as well as being given intraperitoneal (ip) injec-
vascular reaction depends on vasodilators, such as endothelial tion of 15, 30, and 60 mg/kg bw of CGA, respectively.
nitric oxide synthase (eNOS; Sawada & Liao, 2009), and vaso- Four to five animals of the same group were placed in the
constrictors, such as endothelin‐1 (ET‐1; Yeung, Shen, Chung, same cage. Transient global ischemia (TGI) model was
& Chung, 2013). ET‐1 is a potent vasoconstrictor that causes performed using the bilateral common carotid occlusion
ischemia injury and functional deficits in learning and mem- (BCCO) method based on the protocol of previous stud-
ory (Sheng et al., 2015). In the chronic phase after an ischemia/ ies (Hatakeyama, Matsumoto, Brengman, & Yanagihara,
reperfusion injury, there will be a recovery period involving 1988; Martinez, Musumeci, Loreto, & Carnazza, 2007;
eNOS (Sawada & Liao, 2009) and vascular endothelial growth Sharifi et al., 2012; Yu et al., 2012; Zhao, Zhang, Guo,
factor‐A (VEGF‐A; Wang et al., 2005; Greenberg & Jin, 2013). & Zhang, 2013; Zheng, Liu, Wang, & Xu, 2007). The
Polyphenols are known to exert neuroprotective effects rats were anesthetized using intramuscular injection of
by lowering apoptotic markers (Panickar & Anderson, 2011). 100 mg/kg bw of ketamine (PT Guardian Pharmatama,
Chlorogenic acid (CGA) is the main phenol compound pres- Jakarta, Indonesia). The anterior neck skin was opened,
ent in coffee and known to have a very beneficial antioxi- blunt dissection of salivary gland and muscle was con-
dant effect (Ito, Sun, Watanabe, Okamoto, & Hatano, 2008; ducted to visualize trachea and both common carotid
Lafay, Morand, Manach, Besson, & Scalbert, 2006; Marinova, arteries, and the vagal nerves were separated from the
Toneva, & Yanishlieva, 2009; Olthof, Hollman, & Katan, arteries. The carotid arteries were clamped using non‐
2001; Vauzour, 2012). It has been reported to have neuro- traumatic vascular clamps (Dieffenbach; World Precision
protective effects, and it may improve memory functions in Instruments, USA). Both arteries were occluded for
mice with scopolamine‐induced amnesia (Kwon et al., 2010). 20 min. The same procedure was performed on rats of the
Ahn et al. (2011) reported that CGA alone or in combination SO group except the occlusion of the common carotid ar-
with the ribosomal protein PEP‐1‐rpS3 might have protec- teries. CGA (Sigma‐Aldrich, USA, Cat. #C3878‐1G) was
tive effects on the CA1 pyramidal cells of the hippocampus administered 30 min after the BCCO procedure. The CGA
of gerbils undergoing brain ischemia as expressed by relative was dissolved in phosphate‐buffered saline (PBS) solu-
cell density. However, this study did not conduct quantitative tion (every 10 mg CGA was dissolved in 1 ml PBS). The
examination on the hippocampal pyramidal cells (Ahn et al., control groups (the SO and BCCO groups) received PBS
HERMAWATI et al.   
| 3

injection to ensure that any parameter changes observed recorded. The target quadrant was the quadrant where the plat-
in the CGA treatment groups during the experiment were form was previously located in the learning phase. The time
solely due to the effects of CGA and not the solvent. spent was presented as the percentage of total time spent in the
The animals were kept in the recovery phase for 2 days target quadrant to 60 s, whereas the path length was presented
before being tested for the spatial memory. Four to five rats of as the percentage of total trajectory in the target quadrant to
the same group undergoing recovery were placed in one cage. the total distance traveled within 60 s.
During the recovery phase, the rats did not get any medica- An additional test, which was a visual testing (visible plat-
tion, except antiseptic on the site of surgery. Careful observa- form) or sensorimotor test, was performed 1 day after the re-
tion on the site of surgery was performed regularly to detect tention phase test. This test assesses the sensory, motor, and
any early signs of infection. motivational factors of the rats upon performing the memory
task. In this procedure, the platform was made visible by at-
taching a color flag on top of the platform. This test consisted
2.2 | Morris water maze (MWM) test
of three trials in 1 day with the duration of 60 s for each trial.
The device for the test consisted of a white‐painted rounded It was expected that all rats had an equal sensorimotor ca-
pool (1.5 m in diameter and 0.4 m in height). The pool was di- pability, and thus, any differences between the rats’ perfor-
vided into four virtual quadrants titled A, B, C, and D. There mance during the memory test arose from the differences in
was a rounded white‐painted platform (13 cm in diameter and the spatial memory capability.
16.5 cm in height) located in the middle of a randomly cho-
sen quadrant and kept in the same location throughout the
experiment for each rat. Eight starting points were marked
2.3 | Tissue preparation
on the outside of the tank wall. Several colored pictures were Ten days after BCCO, the rats were euthanized by cervical
placed on the curtain around the pool. These pictures served dislocation and decapitation under deep anesthesia of chloro-
as distal cues for the rats to find the platform. The tank was form (Merck, Germany, Cat. #1024451000). The cerebrums
filled with water and added with milk (1.5–2.5 cm above the of the rats were dissected out from the skulls, and the two cer-
platform) until the platform was barely visible. ebral hemispheres were separated from each other. The right
The spatial memory test, which consisted of an escape hippocampus was isolated from the right hemisphere and
acquisition phase and a memory retention phase, was per- stored in RNAlater® Stabilization Solution (Thermo Fisher
formed 3 days after BCCO using the protocol of Bouet et al. Scientific, USA, Cat. #AM7021).
(2010). The escape acquisition or learning phase began with The left hemisphere was soaked in 10% formaldehyde in
placing a randomly chosen rat at a randomly selected starting phosphate buffer solution (PBS) for 24 hr. On the next day,
point with its head facing the pool wall. The starting point the left hippocampus was isolated from the hemisphere and
was randomly determined between numbers 1 and 8 for each immersed in 70% ethanol solution before being dehydrated
test. The platform was placed in the middle of a randomly se- in ascending concentrations of ethanol, cleared with toluene,
lected quadrant, and this platform was kept at the same place infiltrated, and finally embedded in a paraffin block.
for any given rat during all trials of the escape acquisition
phase. The rat was expected to accidentally find the platform
and climb on to it. This marked the end of the trial. The time
2.4 | Stereological analyses
from the start to the end of the trial was recorded as an escape Six paraffin blocks containing hippocampal tissues from
latency. The rat was left on the platform for 20 s before it was each group were randomly taken and processed further for
taken, dried, and returned to the holding cage for 60 s be- stereological analyses. As most tissues usually shrink dur-
fore the next trial. If the rat failed to find the platform within ing histological processing (Partadiredja, Miller, & Oorschot,
60 s, the rat was taken from the pool and placed on top of 2003), the correction for shrinkage is important in the estima-
the platform for 20 s. In this case, the escape latency was re- tion of volume. In order to calculate the shrinkage of the hip-
corded as 60 s. All trials were recorded with a video camera pocampus, each hippocampus was photographed twice using
placed above the pool and connected to a personal computer. a Deskcam camera (PT. Miconos, Indonesia), that is, shortly
The path length was measured indirectly on the screen of the after being isolated from the cerebrum and after being dehy-
computer using a curvimeter (Comcurve 10; Koizumi Sokki drated. A virtual grid made using ImageJ software program
Mfg, Nagaoka‐shi, Niigata, Japan). Each rat underwent four (NIH Image; National Institutes of Health, Bethesda, MD)
trials per day for four consecutive days. was superimposed on these photographs and used to count the
The retention phase was conducted 24 hr after the last trial number of points falling on the photographs of hippocampus.
of the learning phase. The platform was removed from the pool This number of points was entered into the following formula
during the test. The test was performed only once for 60 s. The to calculate the volume shrinkage factor (Nyengaard, 1999;
total time spent and path length in the target quadrant were Pulungan, Sofro, & Partadiredja, 2018):
4
|    HERMAWATI et al.

Volume shrinkage = 1−
(Σpoints after dehydration∕Σpoints before dehydration)1.5

The average volume shrinkage was 36.58%, with the maxi-


mum and minimum values of 64.64% and 14.61%, respectively.
All paraffin blocks containing hippocampal tissues were
sectioned at 3 μm thickness using a Leica RM2235 micro-
tome (Leica Biosystems, Wetzlar, Germany). The first sec-
tion showing the hippocampal tissue was considered as the
first section, and the next sections were numbered sequen-
tially. For each rat, a number between 1 and 150 was ran-
domly chosen to determine the first section to be taken as
a sample. A pair of consecutive sections was taken, and the
following 148 sections were removed before another pair of
F I G U R E 1 A photograph showing the whole area of
sections was taken. This procedure was repeated until the
hippocampus superimposed by a virtual grid
entire hippocampus was thoroughly sectioned (Howard &
Reed, 2005). This procedure yielded between 7 and 10 pairs Vref = T.ΣP.(a∕p)(1 + volume shrinkage)
of sampled sections showing the CA1 region with an aver-
age of eight pairs of sections and between six and nine pairs
of sections showing the CA2–CA3 region with an average where “Vref” is volume of the hippocampus, “T” is the distance
of eight pairs of sections. All sampled sections were stained between the sampled sections, “(a/p)” is the area represented
with 1% toluidine blue solution. by each point of the grid, “ΣP” is total number of points, and
The total number of pyramidal cells of hippocampus “volume shrinkage” is the shrinkage of the tissue which had
was estimated using the Nv × Vref method (West, 2012a), in been calculated before under the presumption that the shrinkage
which Nv is the number of cells per unit volume and Vref occurred at the same rate for the three axes (x, y, and z).
is the reference volume, or the hippocampal pyramidal cell The numerical density (Nv) of pyramidal cells of hip-
layer volume. The hippocampal volume was estimated using pocampus was calculated using the physical disector probe
the Cavalieri principle (Gundersen & Jensen, 1987; Michel as described in previous studies (Hermawati, Sari, &
& Cruz‐Orive, 1988; Miki, Harris, Wilce, Takeuchi, & Bedi, Partadiredja, 2015; Partadiredja & Bedi, 2010; Sterio, 1984).
2000, 2004; Partadiredja & Bedi, 2010). One section from A pair of consecutive sections were designated as a test sec-
each pair of sampled sections was taken for the estimation. tion and lookup section. The fields of view (FOVs) of each
Pictures of hippocampus from these sections were viewed hippocampal test section to be examined were randomly and
under an Olympus CX21FS1 (Olympus, Singapore PTE systematically selected in a raster fashion of every 5 FOVs
Ltd) light microscope at 40× magnification and captured for the CA1 region and every 3 FOVs for the CA2–CA3 re-
using a digital camera (Optilab; PT. Miconos, Indonesia). gion. The selected FOVs were viewed under the Olympus
The photographs of hippocampal parts were merged using CX21FS1 (Olympus, Singapore PTE Ltd) light microscope
Microstepper (PT. Miconos, Indonesia) to display the whole and captured using a digital camera (Optilab; PT. Miconos,
area of the hippocampus. A virtual grid consisting of reg- Indonesia). The analogue of these FOVs at the lookup sec-
ularly spaced array of points at a distance of 10 mm be- tions was also captured. This strategy yielded between 12 and
tween points was made using ImageJ software (NIH Image; 22 pairs of sampled FOVs of the CA1 region with an average
National Institutes of Health, Bethesda, MD). This grid was of 17 pairs of FOVs and between 18 and 43 pairs of FOVs of
superimposed on the picture of the hippocampus (Figure 1). the CA2–CA3 region with an average of 31 pairs of FOVs.
Using a slide micrometer (Ted Pella, Inc., USA), a final An image of a rectangular counting frame slightly smaller
magnification of the picture was calculated to be 350×, and than an FOV, which was 50 × 50 mm, was placed over the
at this magnification, the area per point was 816,326 μm2. picture of the examined area. The final magnification of this
The total number of points (ΣP) falling on the hippocampal frame was calculated using the slide micrometer and found
area was counted. The total number of points counted for the to be 850×. At this magnification, the area of the counting
CA1 region was between 211 and 841 points with an aver- frame (“a”) was calculated to be 3,460.20 μm2. This count-
age of 561 points, while that for the CA2–CA3 region was ing frame consisted of two adjacent solid lines designated as
between 314 and 1058 points with an average of 732 points. exclusion lines and two adjacent interrupted lines designated
The volume of hippocampus was calculated using the for- as inclusion lines. The pyramidal cell nucleus was used as the
mula (Howard & Reed, 2005; Pulungan et al., 2018; West, counting unit. Following the forbidden line rule (Gundersen,
2012a): 1977), any nuclear profile appearing in the counting frame
HERMAWATI et al.   
| 5

of the test section but not in that of the lookup section pro- The precision estimate of the stereological procedure was
vided that this profile did not touch the exclusion lines or calculated using the formula:
their extensions, was counted (Figure 2). In order to increase
CV2total = CV2biol + CE2
the efficiency in the counting, the test section was afterward
used as the lookup section and vice versa (Gundersen, 1986).
The hippocampal tissue slides examined were coded, and where CV is coefficient of variation, CVbiol is biological co-
therefore, the experimenter was not aware of which experi- efficient of variation which reflects individually biological
mental group any slide belonged. The total number of nuclear variations, and CE is coefficient of error that reflects sampling
profiles (ΣQ−) was between 111 and 281 profiles with an av- error (Boyce, Dorph‐Petersen, Lyck, & Gundersen, 2010;
erage of 218 profiles in the CA1 region, whereas that in the West, 2012b). The sampling strategy is considered to be op-
CA2–CA3 region was between 119 and 296 profiles with an timal if CE2/CV2 is between 0.2 and 0.5 (Agustina, Sofro, &
average of 207 profiles. Partadiredja, 2019; Partadiredja, Karima, Utami, Agustiningsih,
The numerical density of pyramidal cells in hippocampus & Sofro, 2019).
was calculated using the following formula (Howard & Reed,
2005; Pulungan et al., 2018; Sterio, 1984; West, 2013): 2.5 | RNA extraction and real‐time

Nv = (ΣQ ∕a.h.ΣP).(1∕(1 + vol.shrinkage)), quantitative PCR
Six samples from each group were randomly taken and pro-
where “ΣQ−” is the total number of nuclear profiles, “a” is the
cessed for RNA extraction. The right hippocampus was cut
area of counting frame, “h” is the section thickness (3 μm),
into half and homogenized for RNA extraction in RNAiso
“ΣP” is the total number of counting frames, and “vol.shrink-
PLUS (Takara Bio, Tokyo, Japan, Cat. #9108/9109). The
age” is the shrinkage correction. The total number of pyramidal
RNA of the hippocampal tissue was extracted using RNAiso
cells of hippocampus was calculated by multiplying Vref and Nv
PLUS (Takara Bio, Tokyo, Japan, Cat. #9108/9109).
(Hermawati et al., 2015; Partadiredja & Bedi, 2010; Pulungan
RNA quantification was determined using NanoDrop.
et al., 2018).

(a)

(b)

F I G U R E 2 Micrographs showing
a pair of sections (3 μm thickness) with
physical disector probes of the CA1
region (a) and CA2–CA3 region (b) of the
hippocampus. Any nuclear profiles observed
in the counting frame of the test sections but
not in the lookup sections were counted (x)
6
|    HERMAWATI et al.

Subsequently, cDNA was made using 1,000 ng of RNA procedure. Otherwise, Kruskal–Wallis tests were conducted.
according to the manufacturer's instructions (ReverTra The analyses were followed by LSD or Mann–Whitney post
Ace Kit; TOYOBO Co., Japan, Cat. #TRT‐101) in a 20 μl hoc tests where needed. The statistical significance was set at
mixture reaction with random primer. The cycling condi- probability value of p < .05. All statistical analyses were per-
tions were 30°C for 10 min, 42°C for 60 min, and 99°C for formed using SPSS software version 20 (IBM Corp., USA).
5 min. The cDNA was then stored in a −20°C freezer.
Real‐time quantitative PCR was performed using KAPA
SYBR® FAST Master Mix (2×) Universal (Kapa Biosystems,
3 | RESULTS
KK4600, Massachusetts, USA) in a real‐time PCR machine 3.1 | CGA prevents spatial memory
(CFX96; Bio‐Rad, USA). The cDNA was diluted four times deterioration
prior to the PCR. The PCR condition of the denaturation stage
The results of escape latency (Figure 3a) and path length
was 94°C for 2 min. The primers, number of cycles, annealing
(Figure 3b) displayed the same pattern. Beginning with the
temperatures, and time were adjusted for each gene examined
third trial, all groups demonstrated a decrease in the escape la-
(Table 1), followed by an elongation stage with a temperature
tency and began to travel at a shorter path length. After the 14th
of 72°C for 10 min. The relative quantification of target genes’
trial, the graph showed a stable pattern of escape latency and
mRNA expression was calculated using the △△CT method.
path length for all groups. These indicated that on the fourth
day, the rats of all groups have learned the escape procedure.
The BCCO group required the longest latency to find the plat-
2.6 | Statistical analyses
form compared to any other groups in most trials except for
Prior to data analyses, normality and homogeneity tests were trials 1, 9, 10, and 15. The BCCO group also traveled at the far-
performed using the Shapiro–Wilk or Kolmogorov–Smirnov thest distance in most trials except for trials 1, 3, 7, and 8. In the
tests and the Levene tests. The data were transformed into last trial, the BCCO group took the longest escape latency and
log10 whenever necessary. When normally distributed and path length to reach the platform compared to all other groups
homogenous, the data were analyzed using one‐way ANOVA of rats.

TABLE 1 Primer list and PCR conditions

No. Gene Forward Reverse PCR condition


1. GAPDH GGCACAGTCAAGGCTGAGAATG TCTCGCTCCTGGAAGATGGTGA 94°C 10 s, 57°C 20 s
72°C 1 min, 35 cycles
2. SOD2 ATGTTGTGTCGGGCGGCGTGCAGC GCGCCTCGTGGTACTTCTCCTCGGTG 94°C 10 s, 65°C 1 min
72°C 1 min, 35 cycles
3. SOD1 GCGGTGAACCAGTTGTGGTG AGCCACATTGCCCAGGTCTC 94°C 10 s, 60°C 20 s
72°C 1 min, 40 cycles
4. GPx CTCT CGCGGTGGCACAGT CCACCACCGGGTCGGACATA C 94°C 10 s, 62°C 20 s
72°C 1 min, 35 cycles
5. Bcl2 TTGTGGCCTTCTTTGAGTTCG TACTGCTTTAGTGAACCTTTTT 95°C 15 s, 60°C 1 min
72°C 1 min, 40 cycles
6. BAX AGACAGGGGCCTTTTTGTTAC GAGGACTCCAGCCACAAAGAT 95°C 15 s, 60°C 1 min
72°C 1 min, 40 cycles
7. Caspase-3 CCG ACT TCC TGT ATG CTT ACT C CGT ACA GTT TCA GCA TGG C 94°C 10 s, 56°C 1 min
72°C 1 min, 35 cycles
8. ET-1 GCTCCTGCTCCTCCTTGATG CTCGCTCTATGTAAGTCATGG 94°C 10 s, 55°C 20 s
72°C 1 min, 35 cycles
9. eNOS CCGGCGCTACGAAGAATG AGTGCCACGGATGGAAATT 94°C 10 s, 54°C 20 s
72°C 1 min, 35 cycles
10. CD31 CCCAGTGACATTCACAGACA ACCTTGACCCTCAGGATCTC 94°C 10 s, 54°C 20 s
72°C 1 min, 35 cycles
11. VEGF‐A ACCTCCACCATGCCAAGT TTGGTCTGCATTCACATCTG 94°C 10 s, 65°C 1 min
72°C 1 min, 35 cycles
HERMAWATI et al.   
| 7

F I G U R E 3 Spatial memory test using Morris water maze. (a) Means ± SEM of escape latency in the escape acquisition phase of the Morris
water maze test (*p < .05, BCCO compared to CGA15, CGA30, and CGA60). (b) Means ± SEM of path length in the escape acquisition phase of
the Morris water maze test (*p < .05, SO compared to CGA30 and CGA60, and BCCO compared to CGA30 in trial 1; SO and BCCO compared to
CGA15, CGA30, and CGA60 in trial 2). (c) The percentages of time in the target quadrant during the retention phase of the Morris water maze test
(*p < .05, BCCO compared to SO, CGA15, CGA30, and CGA60). (d) The percentages of path length in the target quadrant during the retention
phase of the Morris water maze test (*p < .05, BCCO compared to SO, CGA15, CGA30, and CGA60)

The Kruskal–Wallis test revealed that the BCCO group path length (p = .002; df = 4, 50; F = 4.830). The percentages
spent a significantly longer escape latency than the CGA15, of latency and path length in the target quadrant of the BCCO
CGA30, and CGA60 groups (p = .018) in trial 2. The one‐way group were lower than those of other groups as shown by LSD
ANOVA showed significant differences between groups in the post hoc test (Figure 3c and d). It was concluded that CGA
path length in trial 1 (p = .043; df = 4, 50; F = 2.662) and trial improves memory retention function in the TGI model at all
2 (p = .006; df = 4, 50; F = 4.110). The LSD post hoc test doses. The sensorimotor test with a visible platform showed no
showed that the SO group traveled at a longer swimming trajec- significant differences between groups in all trials. Therefore,
tory than the CGA30 (p = .008) and CGA60 groups (p = .044), the results of the Morris water maze test might truly represent
while the BCCO group had a longer swimming trajectory than the spatial memory function (Hermawati, 2018).
the CGA30 group (p = .019) in trial 1. In addition, the SO and
BCCO groups followed a longer swimming trajectory than the
3.2 | CGA inhibits neuronal loss of CA1
CGA15, CGA30, and CGA60 groups (p = .06) in trial 2. There
pyramidal cells of hippocampus
were no significant differences between groups in the escape
latency and path length in any other trials. The estimated volume, numerical density, and total number
One‐way ANOVA of the memory retention phase data of pyramidal cells in the CA1 area of the hippocampus of
demonstrated significant differences between groups in the rats are presented in Table 2. The one‐way ANOVA proce-
percentages of latency (p = .003; df = 4, 50; F = 4.505) and dure of these data revealed that the estimated total number
8
|    HERMAWATI et al.

of pyramidal cells in the CA1 area of the BCCO and CGA15


groups was significantly lower than that of the CGA30 and

CE2tot ∕CV2tot = 0.221


CGA60 groups (p = .026; df = 4, 25; F = 3.316).

CEtot = 0.122
CE (cells) Table 3 shows the estimated volume, numerical density,

BCCO, bilateral common carotid occlusion; CGA, chlorogenic acid; CGA15, BCCO + CGA15 mg/kg bw ip; CGA30, BCCO + CGA 30 mg/kg bw ip; CGA60, BCCO + CGA 60 mg/kg bw ip; SO, sham‐operated.
0.128 and total number of pyramidal cells in the CA2–CA3 area of
0.129
0.126
0.103
0.122
the hippocampus of rats. The one‐way ANOVA procedure
of these data showed no significant main effect of groups
(p = .522; df = 4, 25; F = 0.825). It was concluded that CGA

CVbiol = 0.052
CVtot = 0.259
inhibits CA1 pyramidal cell death in the hippocampus at
CV (cells)

moderate and high doses.


As can be seen from Tables 2 and 3, the sampling strategy
0.213
0.283
0.390
0.171
0.170

of the stereological procedure was considered optimal since


the CE2total ∕CV2total was 0.221 and 0.292 for the estimation of
Total pyramidal cells

the number of pyramidal cells in the CA1 and CA2–CA3 re-


One‐way ANOVA
a

gions, respectively (Hermawati, 2018).


104.460 ± 16.644
146.522 ± 10.256
125.449 ± 10.916

145.591 ± 10.126
99.568 ± 11.515
Estimated volume, numerical density, and total pyramidal cell number (mean ± SEM) in the CA1 area of the hippocampus of rats

F = 3.316

3.3 | CGA increases Bcl2 expression


df = 4,25

p = .026

The effects of CGA on apoptotic regulators were assessed


by measuring the mRNA expressions of Bcl2, Bax, and cas-
395.953 ± 26.358
482.113 ± 30.077
431.423 ± 13.096
387.991 ± 39.198

553.963 ± 19.095

pase‐3 proteins (Figure 4). One‐way ANOVA of the log10‐


transformed data of mRNA expression of Bcl2 demonstrated
a significant main effect of groups (p = .021; df = 4, 11;
Nv (mm3)

F = 4.506). The LSD post hoc test showed that the mRNA
expression of Bcl2 of the BCCO group was lower than that
of the CGA15 (p = .013), CGA30 (p = .032), and CGA60
CE2tot ∕CV2tot = 0.111

groups (p = .004). The CGA60 group had a higher Bcl2 ex-


CEtot = 0.095

pression than the SO group (p = .018). The Kruskal–Wallis


test showed that there was no significant difference between
CE (vol)

0.080
0.083
0.097
0.116

0.095

groups in Bax mRNA expression (p = .200). However, the


Bax expression tended to be higher in the BCCO group than
in all other groups. There was also no significant difference
between groups in the caspase‐3 expression in one‐way
CVbiol = 0.072
CVtot = 0.286

ANOVA (p = .156; df = 4, 13; F = 1.988). Therefore, CGA


CV (vol)

may affect apoptotic process by increasing Bcl2 expression


0.434
0.187
0.193
0.298

0.244

(Hermawati, 2018).
p < .05 compared to the BCCO and CGA15 groups.

3.4 | CGA increases SOD2 expression


Volume (mm3)

0.268 ± 0.047
0.308 ± 0.023
0.290 ± 0.022
0.265 ± 0.032

0.266 ± 0.026

The effects of CGA on antioxidant enzymes were inves-


tigated by measuring the mRNA expressions of SOD2,
SOD1, and GPx proteins (Figure 5). One‐way ANOVA
of these data showed significant differences between
groups in mRNA expression of SOD2 (p = .003; df = 4,
11; F = 8.227). The LSD post hoc test revealed that
mRNA expression of SOD2 of the SO group was lower
than that of the CGA30 (p = .042) and CGA60 groups
n

6
6
6
6

(p = .045), and that of the BCCO group was lower than


that of the SO (p = .042), CGA15 (p = .001), CGA30
(p = .001), and CGA60 (p = .001) groups. The mRNA
TABLE 2

expressions of SOD1 (p = .844 in Kruskal–Wallis test)


CGA15
CGA30
CGA60
Group

BCCO

and GPx (p = .053; df = 4, 14; F = 3.049 in one‐way


SO

ANOVA) did not differ between groups. Nevertheless,


HERMAWATI et al.   
| 9

there was a tendency of lower GPx expression in the

CE2tot ∕CV2tot = 0.292


BCCO group compared to the SO, CGA15, CGA30,

CEtot = 0.114
and CGA60 groups. Thus, CGA may increase SOD2
CE (cells)
expression without affecting other antioxidant enzymes

BCCO, bilateral common carotid occlusion; CGA, chlorogenic acid; CGA15, BCCO + CGA15 mg/kg bw ip; CGA30, BCCO + CGA 30 mg/kg bw ip; CGA60, BCCO + CGA 60 mg/kg bw ip; SO, sham‐operated.
(Hermawati, 2018).
0.127
0.116
0.112
0.109
0.105
3.5 | CGA affects vascular responses by
decreasing ET-1 expression and increasing

CVbiol = 0.031
CVtot = 0.212
CD31 expression
CV (cells)
0.261
0.158
0.290
0.159
0.146
Figure 6 presents the mRNA expression of ET‐1, eNOS, CD31,
and VEGF‐A. One‐way ANOVA of the log10‐transformed data
of mRNA expression of ET‐1 demonstrated that the mRNA ex-
Total pyramidal cells

pression of ET‐1 was higher in the BCCO and CGA15 groups


Estimated volume, numerical density, and total pyramidal cell number (mean ± SEM) in the CA2–CA3 area of the hippocampus of rats

One‐way ANOVA

than in the SO, CGA30, and CGA60 groups (p = .002; df = 4,


94.081 ± 10.054
81.699 ± 5.281
80.870 ± 9.587
94.463 ± 6.169
83.011 ± 4.978

19; F = 5.773). There was no significant difference between


F = 0.825

groups in the mRNA expression of eNOS (p = .265; df = 4,


df = 4.25

p = .522

15; F = 1.456). However, there was a tendency that the BCCO


group had lower eNOS mRNA expression than the CGA15,
CGA30, and CGA60 groups. CGA may affect vascular remod-
236.923 ± 14.678

226.361 ± 25.054
246.659 ± 14.687

eling in the TGI model by decreasing ET‐1 expression.


227.439 ± 8.620

270.390 ± 7.858

In addition to vasodilatory and vasoconstrictive re-


Nv (mm3)

sponses, this study also assessed CD31 as an endothelial


marker (Figure 6). One‐way ANOVA of the log10‐trans-
formed data of mRNA expression of CD31 demonstrated
significant differences between groups (p = .031; df = 4, 15;
CE2tot ∕CV2tot = 0.131

F = 3.563). The mRNA expression of CD31 in the BCCO


CEtot = 0.081

group was lower than that in the CGA15 (p = .003), CGA30


CE (vol)

(p = .025), and CGA60 groups (p = .049). Additionally, the


0.087
0.053
0.085
0.108
0.061

LSD post hoc test showed that the CD31 expression in the
SO group was lower than in the CGA15 group (p = .036).
It was assumed that CGA protected endothelial cells in the
TGI model, and this might be shown by the increase in CD31
CVbiol = 0.043
CVtot = 0.225

expression.
CV (vol)

The one‐way ANOVA procedure demonstrated that there


0.201
0.136
0.357
0.165
0.196

was no significant difference between groups in the mRNA


expression of VEGF‐A (p = .203; df = 4, 14; F = 1.714).
Nevertheless, there was a tendency of higher VEGF‐A ex-
Volume (mm3)

pression in the CGA15, CGA30, and CGA60 groups than the


0.395 ± 0.032
0.359 ± 0.020
0.376 ± 0.048
0.386 ± 0.026
0.309 ± 0.024

BCCO group (Hermawati, 2018).

4 | DISCUSSION

To the best of our knowledge, our research is the first study


that has investigated the effects of CGA on the spatial mem-
ory and the number of hippocampal pyramidal cells in the
n
6
6
6
6
6

TGI model of rats. It has been found that the spatial memory
retention was decreased in TGI‐treated rats (BCCO group)
but not in any other treated group. Accordingly, the num-
TABLE 3

ber of hippocampal pyramidal cells in the CA1 region of the


CGA15
CGA30
CGA60
Group

BCCO

BCCO and CGA15 groups was lower than that of the CGA30
SO

and CGA60 groups. It might be that CGA protects pyramidal


10
|    HERMAWATI et al.

F I G U R E 4 mRNA expressions
of Bcl2 (a), Bax (b), and caspase-3 (c)
(means ± SEM); *p < .05, CGA15 and
CGA30 compared to the BCCO group, and
CGA60 compared to the SO and BCCO
groups

cells by increasing the mRNA expressions of the apoptotic following artificial brain ischemia. The unbiased stereology
regulator Bcl2, the antioxidant enzyme SOD2, and the en- procedure was used in this study to estimate the number
dothelial marker CD31, as well as by decreasing the mRNA of hippocampal pyramidal cells. The present study found
expression of the vasoconstrictor ET‐1. that the number of pyramidal cells in the CA1 area of the
The present study lends support to previous studies (Auer, SO group was 125,449. This figure was close to that ob-
Jensen, & Whishaw, 1989; Bendel et al., 2005; Hartman, served in another study using the same procedure, which
Lee, Zipfel, & Wozniak, 2005; Quintard et al., 2011; Soares, was 152,700 pyramidal cells in the CA1 region of 30‐day‐
Schiavon, Milani, & de Oliveira, 2013) which have demon- old male Wistar rats (Miki et al., 2005). The present study
strated that BCCO‐induced TGI model disrupts the spatial showed that there was no difference between groups in the
memory. CGA reversed this deficit in the spatial memory number of pyramidal cells of the CA2–CA3 region, whereas
retention as shown in the better performance of the CGA15, there were differences between groups in that of the CA1
CGA30, and CGA60 groups compared to the BCCO group. region. The groups with moderate and high doses of CGA
This finding is in agreement with previous studies which had more pyramidal cell counts than the low‐dose CGA and
showed that CGA improved the spatial memory of scopol- BCCO groups. The number of pyramidal cells in the SO
amine (Kwon et al., 2010)‐ and artificial‐aging (senescence group was not significantly different from that in the me-
accelerated mouse; Han, Miyamae, Shigemori, & Isoda, dium‐ and high‐dose CGA groups. This finding is consistent
2010)‐induced memory deterioration in rodents. The im- with studies suggesting that the hippocampal CA1 region
provement of the spatial memory might be partly contrib- is more susceptible to ischemia than the CA2–CA3 region
uted by the possible protective effects of CGA on the CA1 (Auer et al., 1989; Bendel et al., 2005; Bernaudin, Nouvelot,
hippocampal pyramidal cells from cell death initiated by MacKenzie, & Petit, 1998; Hwang et al., 2005).
the ischemia/reperfusion injury. Other studies reported that The vulnerability of the hippocampal CA1 area is due to
CGA protected neuronal cells in cell culture from gluta- several factors. In this area, the NMDA glutamate receptor
mate (Mikami, Kakizawa, & Yamazawa, 2016; Mikami & density is higher than that in the CA3 area (Butler et al., 2010).
Yamazawa, 2015), H2O2 (Cho et al., 2009; Mikami et al., The NMDA receptors in the CA1 area are more responsive
2016), methyl mercury (Mikami et al., 2016), and aluminum to oxygen–glucose deprivation than those in the CA3 area.
(Wang et al., 2017) toxicity. The NMDA receptors in the CA1 area activate tyrosine kinase
Although there was a study on the effect of CGA on the pathways, while those in the CA3 area activate tyrosine phos-
hippocampal tissue of focal ischemic model mice (Miao, phatase pathways. Tyrosine phosphatase activity prevents the
Cao, Li, Fang, & Miao, 2017), it seems that up to the pres- NMDA receptor upregulation in CA3 cells (Gee et al., 2006).
ent, there is no study that quantitatively assesses the hip- An induction of NMDA receptors in the CA1 area leads to a
pocampal pyramidal cell number of rats treated with CGA greater increase in Ca2+ levels than that in the CA3 area. This
HERMAWATI et al.   
| 11

F I G U R E 5 mRNA expressions
of SOD2 (a), SOD1 (b), and GPx (c)
antioxidant enzymes (means ± SEM).
*p < .05, BCCO compared to the SO,
CGA15, CGA30, and CGA60 groups, and
SO compared to the CGA30 and CGA60
groups

F I G U R E 6 mRNA expressions of
ET-1 (a) (*p < .05, BCCO and CGA15
compared to the SO, CGA15, CGA30,
and CGA60 groups), eNOS (b), CD31 (c)
(*p < .05, SO compared to CGA15, and
BCCO compared to CGA15, CGA30, and
CGA60), and VEGF‐A (d) (means ± SEM)

intracellular Ca2+ increase may cause an intracellular accumu- than in the CA3 region. In hypoxic conditions, neurons in the
lation that will result in mitochondrial damage and induce cel- CA1 region express JunB, p53, and Bax more than those in the
lular apoptosis (Li et al., 2000; Stanika, Winters, Pivovarova, CA3 region. In contrast, neurons in the CA3 region and the
& Andrews, 2010). It has also been found that mitochondria dentate gyrus express Bcl2 and Bcl‐XL more than those in the
isolated from cells of the CA1 region release more ROS than CA1 region (Banasiak, Xia, & Haddad, 2000).
those of the CA3 region (Wang & Michaelis, 2010). This po- Chlorogenic acid is a class of the polyphenol group that
tentially causes greater oxidative damage in the CA1 region has antioxidant properties (Bonita, Mandarano, Shuta, &
12
|    HERMAWATI et al.

Vinson, 2007; Esquivel & Jiménez, 2012). It reaches its blood 2004). It has been reported that damaged endothelial cells
level peak within 15 min after intravenous administration express ET‐1 higher than normal (Schaller, 2006). In the
(Wan et al., 2010), whereas its maximum level in plasma is present study, the BCCO group, which had low CD31 expres-
13.33 min with the half‐life of 59.1 min following ip injection sion, demonstrated a high ET‐1 expression (Figure 6). High
(Yuan, Qiao, Xu, Wang, & Li, 2006). The bioavailability of ET‐1 levels also induce endothelial cell damage by affecting
CGA after oral administration is poor due to the first pass NO metabolic pathways, thus increasing the oxidative stress
metabolism by the liver and the degradation by normal flora (Amiri et al., 2004; Davenport et al., 2016). ET‐1 secreted by
in the digestive tract (Gonthier, Verny, Besson, Remesy, & endothelium induces the growth of vascular smooth muscle
Scalbert, 2003). An ip dose of 30 mg/kg bw of CGA was re- cells and the deposition of extracellular matrix, which in turn
ported to exert positive effects on brain damage due to focal results in vascular remodeling. The ET‐1 change in vascular
ischemia as indicated by the improvement in the infarct vol- structure is shown by an increase in the wall and lumen ratio
ume and behavioral score (Lee et al., 2012). CGA improved of resistance arteries (Amiri et al., 2004).
the behavioral function if administered within 5 min up to 1 h Within hours of the acute phase, many inflammatory me-
after the onset of brain ischemia in the rabbit small clot em- diators are released to the ischemic tissue as sterile inflam-
bolic stroke model (RSEM; Lapchak, 2007). Referring to all matory responses to hypoxia (Shichita, Sakaguchi, Suzuki,
of the above‐mentioned findings, the present study used pure & Yoshimura, 2012). As a consequence of the reperfusion
compound of CGA administered intraperitoneally, 30 min phase, the level of free radicals increases and causes oxida-
after BCCO surgery, at a mid‐dose of 30 mg/kg bw. Multilevel tive stress due to the imbalance between the levels of free
doses of 15, 30, and 60 mg/kg bw were determined for this radicals and antioxidant enzymes (Lakhan, Kirchgessner, &
study in order to observe the dose–effect relationship. Hofer, 2009; Mehta et al., 2007; Nakajima et al., 2012).
The BCCO operation induces ischemia/reperfusion injury Oxidative stress may also play an important role in the
(IR injury; Auer et al., 1989; Bendel et al., 2005; Quintard cell death following the brain ischemia (Lakhan et al., 2009;
et al., 2011; Soares et al., 2013), which consists of acute and Nakajima et al., 2012). An inability of the antioxidant system
chronic phases (Fagan, Hess, Hohnadel, Pollock, & Ergul, to overcome free radicals may result in the damage of cellu-
2004). The acute phase is characterized by the release of lar proteins, DNA, and lipids (Mehta et al., 2007). Most of
apoptotic mediators (Eltzschig & Collard, 2004; Eltzschig & these free radicals are present in the mitochondria as a con-
Eckle, 2011). Our study demonstrated that the CGA‐treated sequence of the electron transport chain. The first‐line de-
groups had higher mRNA expression of Bcl2, an anti‐apop- fense of antioxidant systems in the mitochondria is MnSOD
totic protein, than the BCCO group. Furthermore, the mRNA (SOD2), whereas that in the cytosol is SOD1 (Cu‐Zn SOD;
expression of Bcl2 was higher in the CGA60 than the SO Ruszkiewicz & Albrecht, 2015).
group. The alterations in this apoptosis regulator may inhibit Chlorogenic acid may inhibit oxidative stress‐induced cell
delayed neuronal death. death by capturing free radicals. The phenol ring of CGA do-
Delayed neuronal death usually occurs several days after nates hydrogen atoms to the free radicals and hence neutral-
IR injury (Inagaki & Etgen, 2013; Movassaghi et al., 2012; izes them (Karthikesan, Pari, & Menon, 2010; Leopoldini,
Quintard et al., 2011). Anticipating the occurrence of this cell Russo, & Toscano, 2011; Zang, Cosma, Gardner, Castranova,
death, the present study performed hippocampal pyramidal & Vallyathan, 2003). CGA may also increase the expression
cell counting on the 10th day after BCCO procedure. In addi- of antioxidant enzymes. Indeed, in this study, CGA increased
tion, it took several days for the rats to recover from the sur- the mRNA expression of SOD2. There was no significant dif-
gery and another 6 days to perform the spatial memory test. ference between groups in the GPx enzyme expression, but
A previous study in our laboratory has also found that on the there was a tendency that CGA increased the mRNA expres-
10th day following BCCO operation, changes in the mRNA sion of GPx. These results corroborate the findings of other
expressions of SOD2, Bcl2, eNOS, ET‐1, and CD31 were still studies on neuronal and mesenchymal stem cell culture that
detected (Hermawati, Arfian, Mustofa, & Partadiredja, 2018). CGA increased the expressions of SOD2 (Li et al., 2012) and
Pathological processes emerging in blood vessels follow- GPx (Li et al., 2008; Wang et al., 2017). However, the ad-
ing IR injury can be divided into two phases, that is, acute ministration of CGA did not affect the mRNA expression of
and chronic phases. In the acute phase, which occurs within SOD1. This might be understood in light of the fact that most
a few hours, ET‐1 and VEGF are released and affect the of the emerging free radicals following ischemia are found
blood–brain barrier permeability and vascular tone. During in mitochondria, where SOD2 is the first‐line defense of the
the chronic phase, which starts within several days after IR antioxidant enzymes (Ruszkiewicz & Albrecht, 2015). This
injury, apoptosis and angiogenesis occur (Fagan et al., 2004). study revealed that the administration of CGA at the three
In the acute phase of IR injury, vascular tone is affected dose levels increased the mRNA expression of SOD2 enzyme
by factors including ET‐1, NO, VEGF, and angiopoietin‐1. in the CGA‐treated groups as compared to the BCCO group.
ET‐1, one of these factors, is a vasoconstrictor (Fagan et al., Furthermore, CGA at doses of 30 and 60 mg/kg bw brought
HERMAWATI et al.   
| 13

about the mRNA expression of SOD2 higher in the CGA30 An alternative of caspase‐independent pathway of cell
and CGA60 groups than the SO group. death is a necrosis pathway. During cerebral ischemia, many
Cell deaths induced by ischemia/reperfusion injury occur free radicals are produced, and hence, Ca2+ ions easily enter
through apoptosis, autophagy, or necrosis (Puyal, Ginet, & the neurons. Ischemic conditions lead to energy deficits,
Clarke, 2013). The present study examined cell death path- which bring about the failure of the Ca‐ATPase pump to
ways through the mRNA expressions of Bcl2, Bax, and drive Ca2+ ions out of the cells. This results in the accumu-
caspase‐3. It has been found that CGA protected hippocam- lation of Ca2+ ions in the cells. In turn, increased levels of
pal pyramidal cells by increasing mRNA expression of Bcl2. Ca2+ ions in the cells activate proteases, protein kinases, or
Other studies on neurons, mesenchymal stem cells, and liver phospholipases. One of the proteases that is activated in the
tissue cultures also demonstrated an increase in Bcl2 follow- cytosol is calpain (Hayashi & Abe, 2004; Ostwald, Hagberg,
ing the administration of CGA (Ali et al., 2017; Fang et al., Andine, & Karlsson, 1993). Subsequently, proteolysis oc-
2016; Kim et al., 2012; Li et al., 2012; Shi et al., 2009). Bcl2 curs, cytoskeletons degrade (Hayashi & Abe, 2004; Yokota
plays a role in protecting cells by inhibiting mitochondrial et al., 2003), and the neurons eventually die. It has been re-
permeability transition, forming bonds with pro‐apoptotic ported that the proteolysis occurred within 15 min after the
proteins, inhibiting free radical formation, inhibiting Ca2+ in- ischemia in the putamen, parietal cortex, and hippocampal
flux, and regulating intracellular pH (Banasiak et al., 2000). CA1 region. On the seventh day after ischemia, only the
Another regulator that influences cell death is Bax. Our hippocampus still showed the proteolytic signs. This sug-
study found that there was no significant difference between gests that the proteolysis in the hippocampal CA1 region oc-
groups in the mRNA expression of Bax, but there was a curs slowly (Yokota et al., 2003), and this might explain the
tendency that Bax expressions in the CGA15, CGA30, and phenomenon of delayed neuronal death in the hippocampus
CGA60 groups were decreased. Under normal conditions, after the brain ischemia. The present study found that such
Bax is present in the cytosol. Under stressful conditions, Bax neuronal death of CA1 pyramidal cells of the hippocampus
moves to the mitochondria and alters the stability of mito- occurred 10 days after BCCO surgery as had been predicted
chondrial membranes and causes mitochondrial membrane beforehand.
potential changes (Banasiak et al., 2000). Bax opens mito- Other cells than neurons that are also susceptible to
chondrial permeability transition pore (mPTP) located be- ischemia are glial and endothelial cells (Chen et al., 2011;
tween the inner and the outer membrane of the mitochondria. Hayashi & Abe, 2004; Shichita et al., 2012). Our study found
Further damage to the outer membrane of the mitochondria that the expression of CD31, which is an endothelial cell
brings about the release of various apoptotic factors, such as marker (Burger & Touyz, 2012), of the CGA15, CGA30,
cytochrome c, to the cytosol. This will induce cellular apop- and CGA60 groups was higher than that of the BCCO group.
tosis whether it is caspase‐dependent or caspase‐free apopto- Thus, CGA may protect blood vessels’ endothelium from the
sis (Lipton, 1999; Puyal et al., 2013). deleterious effect of ischemia.
There was no significant difference between groups On the 7th day after ischemia, an angiogenesis pro-
in the mRNA expression of caspase‐3. Caspase is known cess involving angiopoietin‐1 and VEGF begins (Arai, Jin,
to play a role in both intrinsic and extrinsic pathways of Navaratna, & Lo, 2009; Shen et al., 2014). In the acute phase,
apoptosis. However, there are also apoptotic pathways that VEGF increases the permeability of the blood–brain barrier.
are caspase‐independent. The caspase‐independent apop- In the later phase, VEGF triggers the endothelial response to
tosis is triggered by the release of apoptosis‐inducing fac- angiogenesis (Fagan et al., 2004). Considering this pattern
tor (AIF) and endonuclease G (endoG). These factors are of VEGF actions on angiogenesis, this study measured the
released from the mitochondria to the nucleus where they mRNA expression of VEGF‐A on the 10th day.
trigger DNA damage (Puyal et al., 2013). Under cerebral VEGF‐A is considered to play a very important role in an-
ischemia conditions, AIF is released by the activation of giogenesis, which also occurs in ischemia/reperfusion injury
PARP1 (Culmsee et al., 2005). In turn, AIF induces cell (Arai et al., 2009). VEGF induces the migration and prolif-
death (Culmsee et al., 2005; Zhu et al., 2003, 2006). AIF eration of endothelial cells and increases the vascular per-
release occurs earlier than the release of cytochrome c meability during the tissue repair (Bagdas et al., 2014). No
(Zhu et al., 2003). This might be the reason for the ab- significant difference between groups in the VEGF‐A expres-
sence of the differences between groups in the mRNA sion was found in the present study. However, there was a ten-
expression of caspase‐3. It is plausible that the neuronal dency of higher VEGF‐A expression in the CGA15, CGA30,
death occurred through a caspase‐independent pathway, and CGA60 groups than in the BCCO group. Apparently
as has been reported that neurons in the hippocampal CA1 CGA exerts paradoxical effects on physiological and patho-
region suffered from cell death via a caspase‐independent logical conditions (Bagdas et al., 2014; Park, Hwang, Park,
pathway following a global ischemia injury (Hua et al., & Lee, 2015) In pathological conditions such as diabetic ret-
2007). inopathy (Zhou et al., 2016) or adenocarcinoma cell culture
14
|    HERMAWATI et al.

(Park et al., 2015), CGA inhibits VEGF expression. On Perguruan Tinggi scheme of the Ministry of Research,
the other hand, in physiological conditions, for example, in Technology and Higher Education (Grant Number 0299/
wound repair or postischemia, CGA stimulates VEGF ex- E3/2016). We would like to thank Suparno (Department
pression (Bagdas et al., 2014; Fagan et al., 2004). of Physiology, Faculty of Medicine, UGM), Suhardi
In ischemic stroke, however, VEGF shows a biphasic (Department of Histology and Cellular Biology, Faculty of
feature. In the acute phase, VEGF increases the blood–brain Medicine, UGM) and Wiwit Ananda, M.Sc (Department of
barrier permeability. In the subacute and chronic phases, that Anatomy, Faculty of Medicine, Public Health and Nursing,
is, after a few days postischemia, VEGF is protective. It in- UGM), for their technical assistances as well as Erik C.
duces re‐endothelialization of blood vessels. VEGF develops Hookom for language editing.
normal endothelial functions and suppresses endothelial cell
damage via the phosphatidylinositol 3‐kinase pathway. VEGF CONFLICT OF INTEREST
also stimulates neovascularization (Fagan et al., 2004). Our
study may lend support to this notion, since at 10 days after The authors do not have conflicts of interest.
the global ischemia of the brain, there was a slight increase in
VEGF expression following CGA administration. This may AUTHOR CONTRIBUTIONS
signify an active angiogenesis process.
The chronic or recovery phase when angiogenesis occurs Ery Hermawati designed the experiment, conducted the
involves the vasodilator eNOS, which plays a role in the pro- experiment, collected and analyzed data, as well as wrote
liferation and differentiation of vascular smooth muscle cells the manuscript. Nur Arfian designed the experiment and
(Sawada & Liao, 2009). This leads to a vascular remodeling analyzed and interpreted the data. Mustofa designed the ex-
response. This is also largely determined by ET‐1 as a vaso- periment and checked the interpretation of the data. Ginus
constrictor, the level of which increases with ischemia (Hung Partadiredja designed the experiment, interpreted the data,
et al., 2015; Leung, Ho, Lo, Chung, & Chung, 2004). In addi- wrote the manuscript and was responsible for final approval
tion, eNOS as a vasodilator restores the blood flow after the of the version to be published.
ischemia (Sawada & Liao, 2009). Ischemia/reperfusion injury
causes the vascular remodeling in the form of increased ET‐1 DATA AVAILABILIT Y STATEMENT
expression and decreased eNOS expression. This results in
an imbalance between vasoconstrictor and vasodilator levels, Original data have been deposited in Figshare.
as evidenced in the kidneys (Arfian et al., 2012). The present
study revealed that CGA lowered the ET‐1 expression, but
ORCID
not the eNOS expression. Possibly, CGA affects the vascular
response through the vasoconstrictor ET‐1, but not through Ery Hermawati https://orcid.org/0000-0002-4814-6178
the vasodilator eNOS. Nur Arfian https://orcid.org/0000-0003-1694-2054
Chlorogenic acid may prevent the spatial memory reten-
Mustofa Mustofa https://orcid.org/0000-0002-0522-8413
tion capability and the hippocampal CA1 pyramidal cells
from degeneration‐induced transient global ischemia. CGA Ginus Partadiredja https://orcid.
may exert these beneficial effects by increasing Bcl2 and org/0000-0003-0395-4240
SOD2 expressions. Furthermore, CGA may affect the vas-
cular response in the global ischemia model by repairing R E F E R E NC E S
endothelial cell damage by increasing CD31 expression
Agustina, F., Sofro, Z. M., & Partadiredja, G. (2019). Subchronic ad-
and decreasing ET‐1 expression. It is acknowledged that
ministration of high‐dose sodium fluoride causes deficits in cere-
no comparison between CGA and drugs of standard ther- bellar Purkinje cells but not motor coordination of rats. Biological
apy for brain ischemia (e.g., tissue plasminogen activator, Trace Element Research, 188, 424–433. https​ ://doi.org/10.1007/
piracetam, citicoline) was made in this study. Therefore, s12011-018-1420-0
another study may be directed toward such comparison. Ahn, E. H., Kim, D. W., Shin, M. J., Kwon, S. W., Kim, Y. N., Kim, D.
Future studies on the effect of CGA on other mechanisms S., … Choi, S. Y. (2011). Chlorogenic acid improves neuroprotec-
of pyramidal cell death such as necrotic and caspase‐inde- tive effect of PEP‐1‐ribosomal protein S3 against ischemic insult.
Experimental Neurobiology, 20, 169–175. https​://doi.org/10.5607/
pendent pathways may also worth investigating.
en.2011.20.4.169
Ali, N., Rashid, S., Nafees, S., Hasan, S. K., Shahid, A., Majed, F., &
ACKNOWLEDGEMENTS Sultana, S. (2017). Protective effect of Chlorogenic acid against meth-
otrexate induced oxidative stress, inflammation and apoptosis in rat
This study was a part of Ery Hermawati's doctoral thesis. liver: An experimental approach. Chemico‐Biological Interactions,
The present study was funded by the Penelitian Unggulan 272, 80–91. https​://doi.org/10.1016/j.cbi.2017.05.002
HERMAWATI et al.   
| 15

Amiri, F., Virdis, A., Neves, M. F., Iglarz, M., Seidah, N. G., Touyz, Burger, D., & Touyz, R. M. (2012). Cellular biomarkers of endothelial
R. M., … Schiffrin, E. L. (2004). Endothelium‐restricted overex- health: Microparticles, endothelial progenitor cells, and circulating
pression of human endothelin‐1 causes vascular remodeling and endothelial cells. Journal of the American Society of Hypertension,
endothelial dysfunction. Circulation, 110, 2233–2240. https​://doi. 6, 85–99. https​://doi.org/10.1016/j.jash.2011.11.003
org/10.1161/01.CIR.00001​44462.08345.B9 Butler, T. R., Self, R. L., Smith, K. J., Sharrett‐Field, L. J., Berry,
Andersen, P., Morris, R., Amaral, D., Bliss, T., & O'Keefe, J. (Eds.) J. N., Littleton, J. M., … Prendergast, M. A. (2010). Selective
(2007). The hippocampus book. New York, NY: Oxford University vulnerability of hippocampal cornu ammonis 1 pyramidal cells
Press. to excitotoxic insult is associated with the expression of poly-
Arai, K., Jin, G., Navaratna, D., & Lo, E. H. (2009). Brain angiogenesis amine‐sensitive N‐methyl‐D‐asparate‐type glutamate receptors.
in developmental and pathological processes: Neurovascular injury Neuroscience, 165, 525–534. https​://doi.org/10.1016/j.neuro​scien​
and angiogenic recovery after stroke. The FEBS Journal, 276, 4644– ce.2009.10.018
4652. https​://doi.org/10.1111/j.1742-4658.2009.07176.x Chen, H., Yoshioka, H., Kim, G. S., Jung, J. E., Okami, N., Sakata, H.,
Arfian, N., Emoto, N., Vignon‐Zellweger, N., Nakayama, K., Yagi, K., … Chan, P. H. (2011). Oxidative stress in ischemic brain damage:
& Hirata, K. (2012). ET‐1 deletion from endothelial cells protects Mechanisms of cell death and potential molecular targets for neuro-
the kidney during the extension phase of ischemia/reperfusion in- protection. Antioxidants & Redox Signaling, 14, 1505–1517. https​://
jury. Biochemical and Biophysical Research Communications, 425, doi.org/10.1089/ars.2010.3576
443–449. https​://doi.org/10.1016/j.bbrc.2012.07.121 Cho, E. S., Jang, Y. J., Hwang, M. K., Kang, N. J., Lee, K. W., & Lee,
Auer, R. N., Jensen, M. L., & Whishaw, I. Q. (1989). Neurobehavioral H. J. (2009). Attenuation of oxidative neuronal cell death by coffee
deficit due to ischemic brain damage limited to half of the CA1 sec- phenolic phytochemicals. Mutation Research, 661, 18–24. https​://
tor of the hippocampus. The Journal of Neuroscience, 9, 1641–1647. doi.org/10.1016/j.mrfmmm.2008.10.021
https​://doi.org/10.1523/JNEUR​OSCI.09-05-01641.1989 Culmsee, C., Zhu, C., Landshamer, S., Becattini, B., Wagner, E.,
Bagdas, D., Cam Etoz, B., Inan Ozturkoglu, S., Cinkilic, N., Ozyigit, Pellecchia, M., … Plesnila, N. (2005). Apoptosis‐inducing factor
M. O., Gul, Z., … Gurun, M. S. (2014). Effects of systemic chlo- triggered by poly(ADP‐ribose) polymerase and Bid mediates neu-
rogenic acid on random‐pattern dorsal skin flap survival in diabetic ronal cell death after oxygen‐glucose deprivation and focal cerebral
rats. Biological & Pharmaceutical Bulletin, 37, 361–370. https​:// ischemia. The Journal of Neuroscience, 25, 10262–10272. https​://
doi.org/10.1248/bpb.b13-00635​ doi.org/10.1523/JNEUR​OSCI.2818-05.2005
Banasiak, K. J., Xia, Y., & Haddad, G. G. (2000). Mechanisms underly- Davenport, A. P., Hyndman, K. A., Dhaun, N., Southan, C., Kohan, D. E.,
ing hypoxia‐induced neuronal apoptosis. Progress in Neurobiology, Pollock, J. S., … Maguire, J. J. (2016). Endothelin. Pharmacological
62, 215–249. https​://doi.org/10.1016/S0301-0082(00)00011-3 Reviews, 68, 357–418. https​://doi.org/10.1124/pr.115.011833
Bear, M., Connors, B., & Paradiso, M. (2007). Neuroscience exploring Eltzschig, H. K., & Collard, C. D. (2004). Vascular ischaemia and
the brain. Philadelphia, PA: Lippincott Williams & Wilkins. reperfusion injury. British Medical Bulletin, 70, 71–86. https​://doi.
Bendel, O., Bueters, T., von Euler, M., Ove Ogren, S., Sandin, J., & org/10.1093/bmb/ldh025
von Euler, G. (2005). Reappearance of hippocampal CA1 neurons Eltzschig, H. K., & Eckle, T. (2011). Ischemia and reperfusion–from
after ischemia is associated with recovery of learning and memory. mechanism to translation. Nature Medicine, 17, 1391–1401. https​://
Journal of Cerebral Blood Flow and Metabolism, 25, 1586–1595. doi.org/10.1038/nm.2507
https​://doi.org/10.1038/sj.jcbfm.9600153 Esquivel, P., & Jiménez, V. M. (2012). Functional properties of coffee
Bernaudin, M., Nouvelot, A., MacKenzie, E. T., & Petit, E. (1998). and coffee by‐products. Food Research International, 46, 488–495.
Selective neuronal vulnerability and specific glial reactions in hip- https​://doi.org/10.1016/j.foodr​es.2011.05.028
pocampal and neocortical organotypic cultures submitted to isch- Fagan, S. C., Hess, D. C., Hohnadel, E. J., Pollock, D. M., & Ergul, A. (2004).
emia. Experimental Neurology, 150, 30–39. https​://doi.org/10.1006/ Targets for vascular protection after acute ischemic stroke. Stroke, 35,
exnr.1997.6728 2220–2225. https​://doi.org/10.1161/01.STR.00001​38023.60272.9e
Bonita, J. S., Mandarano, M., Shuta, D., & Vinson, J. (2007). Coffee and Fang, S. Q., Wang, Y. T., Wei, J. X., Shu, Y. H., Xiao, L., & Lu, X. M.
cardiovascular disease: In vitro, cellular, animal, and human studies. (2016). Beneficial effects of chlorogenic acid on alcohol‐induced
Pharmacological Research, 55, 187–198. https​://doi.org/10.1016/j. damage in PC12 cells. Biomedicine & Pharmacotherapy, 79, 254–
phrs.2007.01.006 262. https​://doi.org/10.1016/j.biopha.2016.02.018
Bouet, V., Freret, T., Ankri, S., Bezault, M., Renolleau, S., Boulouard, Gee, C. E., Benquet, P., Raineteau, O., Rietschin, L., Kirbach,
M., … Schumann‐Bard, P. (2010). Predicting sensorimotor and S. W., & Gerber, U. (2006). NMDA receptors and the differ-
memory deficits after neonatal ischemic stroke with reperfusion ential ischemic vulnerability of hippocampal neurons. The
in the rat. Behavioural Brain Research, 212, 56–63. https​://doi. European Journal of Neuroscience, 23, 2595–2603. https​ ://doi.
org/10.1016/j.bbr.2010.03.043 org/10.1111/j.1460-9568.2006.04786.x
Boyce, R. W., Dorph‐Petersen, K. A., Lyck, L., & Gundersen, H. J. Gibbons, G. H., & Dzau, V. J. (1994). The emerging concept of vas-
(2010). Design‐based stereology: Introduction to basic concepts cular remodeling. The New England Journal of Medicine, 330,
and practical approaches for estimation of cell number. Toxicologic 1431–1438.
Pathology, 38, 1011–1025. https​://doi.org/10.1177/01926​23310​ Gonthier, M. P., Verny, M. A., Besson, C., Remesy, C., & Scalbert, A.
385140 (2003). Chlorogenic acid bioavailability largely depends on its me-
Buch, P., Patel, V., Ranpariya, V., Sheth, N., & Parmar, S. (2012). tabolism by the gut microflora in rats. The Journal of Nutrition, 133,
Neuroprotective activity of Cymbopogon martinii against cere- 1853–1859. https​://doi.org/10.1093/jn/133.6.1853
bral ischemia/reperfusion‐induced oxidative stress in rats. Journal Greenberg, D. A., & Jin, K. (2013). Vascular endothelial growth fac-
of Ethnopharmacology, 142, 35–40. https​ ://doi.org/10.1016/j. tors (VEGFs) and stroke. Cellular and Molecular Life Sciences, 70,
jep.2012.04.007 1753–1761. https​://doi.org/10.1007/s00018-013-1282-8
16
|    HERMAWATI et al.

Gundersen, H. J. G. (1977). Notes on the estimation of the numerical endothelial cell adhesion molecule‐1 in the hippocampal CA1 region
density of arbitrary profiles: The edge effect. Journal of Microscopy, in gerbils. Brain Research, 1048, 251–257. https​://doi.org/10.1016/j.
111, 219–223. https​://doi.org/10.1111/j.1365-2818.1977.tb000​62.x brain​res.2005.04.049
Gundersen, H. J. (1986). Stereology of arbitrary particles. A review Inagaki, T., & Etgen, A. M. (2013). Neuroprotective action of acute
of unbiased number and size estimators and the presentation of estrogens: Animal models of brain ischemia and clinical impli-
some new ones, in memory of William R. Thompson. Journal of cations. Steroids, 78, 597–606. https​ ://doi.org/10.1016/j.stero​
Microscopy, 143, 3–45. https​://doi.org/10.1111/j.1365-2818.1986. ids.2012.12.015
tb027​64.x Ito, H., Sun, X. L., Watanabe, M., Okamoto, M., & Hatano, T. (2008).
Gundersen, H. J., & Jensen, E. B. (1987). The efficiency of systematic Chlorogenic acid and its metabolite m‐coumaric acid evoke neurite
sampling in stereology and its prediction. Journal of Microscopy, outgrowth in hippocampal neuronal cells. Bioscience, Biotechnology,
147, 229–263. https​://doi.org/10.1111/j.1365-2818.1987.tb028​37.x and Biochemistry, 72, 885–888. https​://doi.org/10.1271/bbb.70670​
Hartman, R. E., Lee, J. M., Zipfel, G. J., & Wozniak, D. F. (2005). Kandel, E., Schwartz, J., & Jessel, T. (2000). Principles of neural sci-
Characterizing learning deficits and hippocampal neuron loss fol- ence. New York, NY: McGraw‐Hill.
lowing transient global cerebral ischemia in rats. Brain Research, Karthikesan, K., Pari, L., & Menon, V. P. (2010). Protective effect
1043, 48–56. https​://doi.org/10.1016/j.neuro​scien​ce.2010.05.049 of tetrahydrocurcumin and chlorogenic acid against streptozo-
Han, J., Miyamae, Y., Shigemori, H., & Isoda, H. (2010). Neuroprotective tocin–nicotinamide generated oxidative stress induced diabetes.
effect of 3,5‐di‐O‐caffeoylquinic acid on SH‐SY5Y cells and senes- Journal of Functional Foods, 2, 134–142. https​://doi.org/10.1016/j.
cence‐accelerated‐prone mice 8 through the up‐regulation of phos- jff.2010.04.001
phoglycerate kinase‐1. Neuroscience, 169, 1039–1045. https​://doi. Kim, J., Lee, S., Shim, J., Kim, H. W., Kim, J., Jang, Y. J., … Lee,
org/10.1016/j.neuro​scien​ce.2010.05.049 H. J. (2012). Caffeinated coffee, decaffeinated coffee, and the phe-
Hatakeyama, T., Matsumoto, M., Brengman, J. M., & Yanagihara, nolic phytochemical chlorogenic acid up‐regulate NQO1 expres-
T. (1988). Immunohistochemical investigation of ischemic sion and prevent H(2)O(2)‐induced apoptosis in primary cortical
and postischemic damage after bilateral carotid occlusion in neurons. Neurochemistry International, 60, 466–474. https​://doi.
gerbils. Stroke, 19, 1526–1534. https​ ://doi.org/10.1161/01. org/10.1016/j.neuint.2012.02.004
STR.19.12.1526 Kwon, S. H., Lee, H. K., Kim, J. A., Hong, S. I., Kim, H. C., Jo, T. H., …
Hayashi, T., & Abe, K. (2004). Ischemic neuronal cell death and or- Jang, C. G. (2010). Neuroprotective effects of chlorogenic acid on
ganellae damage. Neurological Research, 26, 827–834. https​://doi. scopolamine‐induced amnesia via anti‐acetylcholinesterase and anti‐
org/10.1179/01616​4104X​3770 oxidative activities in mice. European Journal of Pharmacology,
Hermawati, E. (2018). Pengaruh Asam Klorogenat Terhadap Struktur 649, 210–217. https​://doi.org/10.1016/j.ejphar.2010.09.001
dan Fungsi Hippocampus pada Tikus Model Iskemia Global Lafay, S., Morand, C., Manach, C., Besson, C., & Scalbert, A. (2006).
Transien (Kajian terhadap Fungsi Memori Spasial, Jumlah Sel Absorption and metabolism of caffeic acid and chlorogenic acid
Piramidal dan Respon Vaskuler). Indonesia: Universitas Gadjah in the small intestine of rats. The British Journal of Nutrition, 96,
Mada. 39–46. https​://doi.org/10.1079/BJN20​061714
Hermawati, E., Arfian, N., Mustofa, & Partadiredja, G. (2018). Spatial Lakhan, S. E., Kirchgessner, A., & Hofer, M. (2009). Inflammatory
memory disturbance following transient brain ischemia is associated mechanisms in ischemic stroke: Therapeutic approaches. Journal of
with vascular remodeling in hippocampus. Kobe Journal of Medical Translational Medicine, 7, 1–11.
Sciences, 64(3), E93–E106. Lapchak, P. A. (2007). The phenylpropanoid micronutrient chlorogenic
Hermawati, E., Sari, D. C., & Partadiredja, G. (2015). The effects of acid improves clinical rating scores in rabbits following multiple in-
black garlic ethanol extract on the spatial memory and estimated farct ischemic strokes: synergism with tissue plasminogen activator.
total number of pyramidal cells of the hippocampus of monoso- Experimental Neurology, 205, 407–413.
dium glutamate‐exposed adolescent male Wistar rats. Anatomical Lee, K., Lee, J. S., Jang, H. J., Kim, S. M., Chang, M. S., Park, S. H.,
Science International, 90, 275–286. https​ ://doi.org/10.1007/ … Bu, Y. (2012). Chlorogenic acid ameliorates brain damage and
s12565-014-0262-x edema by inhibiting matrix metalloproteinase-2 and 9 in a rat model
Howard, C., & Reed, M. (2005). Unbiased stereology three‐dimen- of focal cerebral ischemia. European Journal of Pharmacology,
sional measurement in microscopy. New York, NY: BIOS Scientific 689, 89–95.
Publishers. Leopoldini, M., Russo, N., & Toscano, M. (2011). The molecular
Hua, F., Ma, J., Ha, T., Xia, Y., Kelley, J., Williams, D. L., … Li, C. basis of working mechanism of natural polyphenolic antioxidants.
(2007). Activation of Toll‐like receptor 4 signaling contributes to Food Chemistry, 125, 288–306. https​ ://doi.org/10.1016/j.foodc​
hippocampal neuronal death following global cerebral ischemia/ hem.2010.08.012
reperfusion. Journal of Neuroimmunology, 190, 101–111. https​:// Leung, J. W., Ho, M. C., Lo, A. C., Chung, S. S., & Chung, S. K. (2004).
doi.org/10.1016/j.jneur​oim.2007.08.014 Endothelial cell‐specific over‐expression of endothelin‐1 leads to
Hung, V. K., Yeung, P. K., Lai, A. K., Ho, M. C., Lo, A. C., Chan, K. more severe cerebral damage following transient middle cerebral ar-
C., … Chung, S. K. (2015). Selective astrocytic endothelin‐1 over- tery occlusion. Journal of Cardiovascular Pharmacology, 44(Suppl
expression contributes to dementia associated with ischemic stroke 1), S293–S300. https​://doi.org/10.1097/01.fjc.00001​66277.70538.b0
by exaggerating astrocyte‐derived amyloid secretion. Journal of Li, S., Bian, H., Liu, Z., Wang, Y., Dai, J., He, W., … Luo, J. (2012).
Cerebral Blood Flow and Metabolism, 35, 1687–1696. https​://doi. Chlorogenic acid protects MSCs against oxidative stress by alter-
org/10.1038/jcbfm.2015.109 ing FOXO family genes and activating intrinsic pathway. European
Hwang, I. K., Kim, D. W., Yoo, K. Y., Jung, B. K., Song, J. H., Jung, Journal of Pharmacology, 674, 65–72. https​ ://doi.org/10.1016/j.
J. Y., … Won, M. H. (2005). Ischemia‐induced changes of platelet ejphar.2011.06.033
HERMAWATI et al.   
| 17

Li, H., Colbourne, F., Sun, P., Zhao, Z., Buchan, A. M., & Iadecola, Nakajima, H., Unoda, K., Ito, T., Kitaoka, H., Kimura, F., & Hanafusa,
C. (2000). Caspase inhibitors reduce neuronal injury after focal but T. (2012). The Relation of Urinary 8‐OHdG, A Marker of Oxidative
not global cerebral ischemia in rats. Stroke, 31, 176–182. https​://doi. Stress to DNA, and Clinical Outcomes for Ischemic Stroke. The
org/10.1161/01.STR.31.1.176 Open Neurology Journal, 6, 51–57. https​://doi.org/10.2174/18742​
Li, Y., Shi, W., Li, Y., Zhou, Y., Hu, X., Song, C., … Li, Y. (2008). 05X01​20601​0051
Neuroprotective effects of chlorogenic acid against apopto- Nyengaard, J. R. (1999). Stereologic methods and their application in
sis of PC12 cells induced by methylmercury. Environmental kidney research. Journal of the American Society of Nephrology,
Toxicology and Pharmacology, 26, 13–21. https​://doi.org/10.1016/j. 10, 1100–1123.
etap.2007.12.008 Olthof, M. R., Hollman, P. C., & Katan, M. B. (2001). Chlorogenic acid
Lipton, P. (1999). Ischemic cell death in brain neurons. Physiological and caffeic acid are absorbed in humans. The Journal of Nutrition,
Reviews, 79, 1431–1568. https​
://doi.org/10.1152/physr​ 131, 66–71. https​://doi.org/10.1093/jn/131.1.66
ev.1999.79.4.1431 Ostwald, K., Hagberg, H., Andine, P., & Karlsson, J. O. (1993).
Marinova, E. M., Toneva, A., & Yanishlieva, N. (2009). Comparison Upregulation of calpain activity in neonatal rat brain after hy-
of the antioxidative properties of caffeic and chlorogenic acids. poxic‐ischemia. Brain Research, 630, 289–294. https​ ://doi.
Food Chemistry, 114, 1498–1502. https​://doi.org/10.1016/j.foodc​ org/10.1016/0006-8993(93)90668-D
hem.2008.11.045 Panickar, K. S., & Anderson, R. A. (2011). Effect of polyphenols on
Martinez, G., Musumeci, G., Loreto, C., & Carnazza, M. L. (2007). oxidative stress and mitochondrial dysfunction in neuronal death
Immunohistochemical changes in vulnerable rat brain regions after and brain edema in cerebral ischemia. International Journal of
reversible global brain ischaemia. Journal of Molecular Histology, Molecular Sciences, 12, 8181–8207. https​://doi.org/10.3390/ijms1​
38, 295–302. https​://doi.org/10.1007/s10735-007-9102-9 2118181
Mehta, S. L., Manhas, N., & Raghubir, R. (2007). Molecular targets Park, J. J., Hwang, S. J., Park, J. H., & Lee, H. J. (2015). Chlorogenic
in cerebral ischemia for developing novel therapeutics. Brain acid inhibits hypoxia‐induced angiogenesis via down‐regulation of
Research Reviews, 54, 34–66. https​ ://doi.org/10.1016/j.brain​ the HIF‐1alpha/AKT pathway. Cellular Oncology (Dordrecht), 38,
resrev.2006.11.003 111–118. https​://doi.org/10.1007/s13402-014-0216-2
Miao, M., Cao, L., Li, R., Fang, X., & Miao, Y. (2017). Protective effect Partadiredja, G., & Bedi, K. (2010). Undernutrition during gestation and
of chlorogenic acid on the focal cerebral ischemia reperfusion rat suckling periods does not cause any loss of pyramidal neurons in the
models. Saudi Pharmaceutical Journal, 25, 556–563. https​://doi. CA2‐CA3 region of the rat hippocampus. Nutritional Neuroscience,
org/10.1016/j.jsps.2017.04.023 13, 1–7.
Michel, R. P., & Cruz‐Orive, L. M. (1988). Application of the Cavalieri Partadiredja, G., Karima, N., Utami, K. P., Agustiningsih, D., & Sofro,
principle and vertical sections method to lung: Estimation of vol- Z. M. (2019). The Effects of Light and Moderate Intensity Exercise
ume and pleural surface area. Journal of Microscopy, 150, 117–136. on the Femoral Bone and Cerebellum of d‐Galactose‐Exposed
https​://doi.org/10.1111/j.1365-2818.1988.tb046​03.x Rats. Rejuvenation Research, 22, 20–30. https​://doi.org/10.1089/
Mikami, Y., Kakizawa, S., & Yamazawa, T. (2016). Essential roles of rej.2018.2050
natural products and gaseous mediators on neuronal cell death or Partadiredja, G., Miller, R., & Oorschot, D. E. (2003). The num-
survival. International Journal of Molecular Sciences, 17, E1652. ber, size, and type of axons in rat subcortical white matter on left
https​://doi.org/10.3390/ijms1​7101652 and right sides: A stereological, ultrastructural study. Journal
Mikami, Y., & Yamazawa, T. (2015). Chlorogenic acid, a polyphenol of Neurocytology, 32, 1165–1179. https​ ://doi.org/10.1023/
in coffee, protects neurons against glutamate neurotoxicity. Life B:NEUR.00000​21910.65920.41
Sciences, 139, 69–74. https​://doi.org/10.1016/j.lfs.2015.08.005 Pulungan, Z. S. A., Sofro, Z. M., & Partadiredja, G. (2018). Sodium
Miki, T., Harris, S. J., Wilce, P., Takeuchi, Y., & Bedi, K. S. (2000). fluoride does not affect the working memory and number of
Neurons in the hilus region of the rat hippocampus are de- pyramidal cells in rat medial prefrontal cortex. Anatomical
pleted in number by exposure to alcohol during early postna- Science International, 93, 128–138. https​ ://doi.org/10.1007/
tal life. Hippocampus, 10, 284–295. https​ ://doi.org/10.1002/ s12565-016-0384-4
(ISSN)1098-1063 Purves, D., Augustine, G., Fitzpatrick, D., Hall, W., LaMantia, A.,
Miki, T., Harris, S. J., Wilce, P. A., Takeuchi, Y., & Bedi, K. S. McNamara, J., & Williams, S. (Eds.) (2004). Neuroscience.
(2004). Effects of age and alcohol exposure during early life on Sunderland, MA: Sinauer Associates Inc.
pyramidal cell numbers in the CA1‐CA3 region of the rat hippo- Puyal, J., Ginet, V., & Clarke, P. G. (2013). Multiple interacting cell death
campus. Hippocampus, 14, 124–134. https​ ://doi.org/10.1002/ mechanisms in the mediation of excitotoxicity and ischemic brain
(ISSN)1098-1063 damage: A challenge for neuroprotection. Progress in Neurobiology,
Miki, T., Satriotomo, I., Li, H. P., Matsumoto, Y., Gu, H., Yokoyama, T., 105, 24–48. https​://doi.org/10.1016/j.pneur​obio.2013.03.002
… Takeuchi, Y. (2005). Application of the physical disector to the cen- Quintard, H., Borsotto, M., Veyssiere, J., Gandin, C., Labbal, F.,
tral nervous system: Estimation of the total number of neurons in sub- Widmann, C., … Heurteaux, C. (2011). MLC901, a traditional
divisions of the rat hippocampus. Anatomical Science International, Chinese medicine protects the brain against global ischemia.
80, 153–162. https​://doi.org/10.1111/j.1447-073x.2005.00121.x Neuropharmacology, 61, 622–631. https​://doi.org/10.1016/j.neuro​
Movassaghi, S., Nadia Sharifi, Z., Soleimani, M., Joghataii, M. T., pharm.2011.05.003
Hashemi, M., Shafaroodi, H., & Mehdizadeh, M. (2012). Effect Ruszkiewicz, J., & Albrecht, J. (2015). Changes in the mitochondrial
of Pentoxifylline on Ischemia‐ induced Brain Damage and Spatial antioxidant systems in neurodegenerative diseases and acute brain
Memory Impairment in Rat. Iranian Journal of Basic Medical disorders. Neurochemistry International, 88, 66–72. https​ ://doi.
Sciences, 15, 1083–1090. org/10.1016/j.neuint.2014.12.012
18
|    HERMAWATI et al.

Sanderson, T. H., Reynolds, C. A., Kumar, R., Przyklenk, K., & Wan, C.‐W., Lee, Y.‐K., Kwok, C.‐Y., Chan, R. Y.‐K., Yu, P. H.‐F., &
Huttemann, M. (2013). Molecular mechanisms of ischemia‐reper- Chan, S.‐W. (2010). A validated HPLC method with dual wave-
fusion injury in brain: Pivotal role of the mitochondrial mem- length detection for chlorogenic acid with an internal standard
brane potential in reactive oxygen species generation. Molecular in plasma and its application in pharmacokinetic studies in rats.
Neurobiology, 47, 9–23. https​://doi.org/10.1007/s12035-012-8344-z Journal of Liquid Chromatography & Related Technologies, 33,
Sasi, N., Hwang, M., Jaboin, J., Csiki, I., & Lu, B. (2009). Regulated cell 324–335. https​://doi.org/10.1080/10826​07090​3524084
death pathways: New twists in modulation of BCL2 family func- Wang, X., Fan, X., Yuan, S., Jiao, W., Liu, B., Cao, J., & Jiang, W.
tion. Molecular Cancer Therapeutics, 8, 1421–1429. https​://doi. (2017). Chlorogenic acid protects against aluminium‐induced cyto-
org/10.1158/1535-7163.MCT-08-0895 toxicity through chelation and antioxidant actions in primary hippo-
Sawada, N., & Liao, J. K. (2009). Targeting eNOS and beyond: Emerging campal neuronal cells. Food and Function, 8, 2924–2934. https​://
heterogeneity of the role of endothelial Rho proteins in stroke pro- doi.org/10.1039/C7FO0​0659D​
tection. Expert Review of Neurotherapeutics, 9, 1171–1186. https​:// Wang, Y., Kilic, E., Kilic, U., Weber, B., Bassetti, C. L., Marti, H. H.,
doi.org/10.1586/ern.09.70 & Hermann, D. M. (2005). VEGF overexpression induces post‐isch-
Schaller, B. J. (2006). The role of endothelin in stroke: Experimental aemic neuroprotection, but facilitates haemodynamic steal phenom-
data and underlying pathophysiology. Archives of Medical Science, ena. Brain: A Journal of Neurology, 128, 52–63.
2, 146–158. Wang, X., & Michaelis, E. K. (2010). Selective neuronal vulnerability to
Selakovic, V., Korenic, A., & Radenovic, L. (2011). Spatial and tem- oxidative stress in the brain. Frontiers in Aging Neuroscience, 2, 12.
poral patterns of oxidative stress in the brain of gerbils submitted West, M. J. (2012a). Estimating volume in biological structures. Cold
to different duration of global cerebral ischemia. International Spring Harbor Protocols, 2012, 1129–1139.
Journal of Developmental Neuroscience, 29, 645–654. https​://doi. West, M. J. (2012b). The precision of estimates in stereological analy-
org/10.1016/j.ijdev​neu.2011.02.009 ses. Cold Spring Harbor Protocols, 2012, 937–949.
Sharifi, Z. N., Abolhassani, F., Zarrindast, M. R., Movassaghi, S., West, M. J. (2013). Tissue shrinkage and stereological studies. Cold
Rahimian, N., & Hassanzadeh, G. (2012). Effects of FK506 on Spring Harbor Protocols, 2013, 208–209.
Hippocampal CA1 Cells Following Transient Global Ischemia/ Yeung, P. K., Shen, J., Chung, S. S., & Chung, S. K. (2013).
Reperfusion in Wistar Rat. Stroke Research and Treatment, 2012, Targeted over‐expression of endothelin‐1 in astrocytes leads
809417. to more severe brain damage and vasospasm after subarach-
Shen, J., Zhu, Y., Yu, H., Fan, Z. X., Xiao, F., Wu, P., … Zhan, R. Y. noid hemorrhage. BMC Neuroscience, 14, 131–145. https​ ://doi.
(2014). Buyang Huanwu decoction increases angiopoietin‐1 expres- org/10.1186/1471-2202-14-131
sion and promotes angiogenesis and functional outcome after focal Yokota, M., Saido, T. C., Kamitani, H., Tabuchi, S., Satokata, I., &
cerebral ischemia. Journal of Zhejiang University. Science. B, 15, Watanabe, T. (2003). Calpain induces proteolysis of neuronal cy-
272–280. https​://doi.org/10.1631/jzus.B1300166 toskeleton in ischemic gerbil forebrain. Brain Research, 984, 122–
Sheng, T., Zhang, X., Wang, S., Zhang, J., Lu, W., & Dai, Y. (2015). 132. https​://doi.org/10.1016/S0006-8993(03)03121-4
Endothelin‐1‐induced mini‐stroke in the dorsal hippocampus or lat- Yu, D. K., Yoo, K. Y., Shin, B. N., Kim, I. H., Park, J. H., Lee, C. H., …
eral amygdala results in deficits in learning and memory. Journal of Won, M. H. (2012). Neuronal damage in hippocampal subregions
Biomedical Research, 29, 362–369. induced by various durations of transient cerebral ischemia in ger-
Shi, H., Dong, L., Bai, Y., Zhao, J., Zhang, Y., & Zhang, L. (2009). bils using Fluoro‐Jade B histofluorescence. Brain Research, 1437,
Chlorogenic acid against carbon tetrachloride‐induced liver fibrosis 50–57. https​://doi.org/10.1016/j.brain​res.2011.12.029
in rats. European Journal of Pharmacology, 623, 119–124. https​:// Yuan, B., Qiao, M., Xu, H., Wang, L., & Li, F. (2006). Determination of
doi.org/10.1016/j.ejphar.2009.09.026 chlorogenic acid in rat plasma by high performance chromatography
Shichita, T., Sakaguchi, R., Suzuki, M., & Yoshimura, A. (2012). Post‐ after peritoneal administration of compound Daqingye injection.
ischemic inflammation in the brain. Frontiers in Immunology, 3, Yakugaku zasshi: Journal of the Pharmaceutical Society of Japan,
132. 126, 811–814. https​://doi.org/10.1248/yakus​hi.126.811
Soares, L. M., Schiavon, A. P., Milani, H., & de Oliveira, R. M. (2013). Zang, L. Y., Cosma, G., Gardner, H., Castranova, V., & Vallyathan,
Cognitive impairment and persistent anxiety‐related responses V. (2003). Effect of chlorogenic acid on hydroxyl radical.
following bilateral common carotid artery occlusion in mice. Molecular and Cellular Biochemistry, 247, 205–210. https​://doi.
Behavioural Brain Research, 249, 28–37. https​://doi.org/10.1016/j. org/10.1023/A:10241​03428348
bbr.2013.04.010 Zhao, X., Zhang, H., Guo, J., & Zhang, Y. (2013). The effects of
Stanika, R. I., Winters, C. A., Pivovarova, N. B., & Andrews, S. B. bilateral common carotid artery occlusion on expression of pe-
(2010). Differential NMDA receptor‐dependent calcium loading and ripherin and choline acetyltransferase activity in C57BL/6 mice.
mitochondrial dysfunction in CA1 vs. CA3 hippocampal neurons. Brain Research, 1491, 167–175. https​ ://doi.org/10.1016/j.brain​
Neurobiology of Disease, 37, 403–411. https​://doi.org/10.1016/j. res.2012.11.012
nbd.2009.10.020 Zheng, Y. Q., Liu, J. X., Wang, J. N., & Xu, L. (2007). Effects of cro-
Sterio, D. C. (1984). The unbiased estimation of number and sizes of cin on reperfusion‐induced oxidative/nitrative injury to cerebral
arbitrary particles using the disector. Journal of Microscopy, 134, microvessels after global cerebral ischemia. Brain Research, 1138,
127–136. https​://doi.org/10.1111/j.1365-2818.1984.tb025​01.x 86–94. https​://doi.org/10.1016/j.brain​res.2006.12.064
Vauzour, D. (2012). Dietary polyphenols as modulators of brain func- Zhou, L., Zhang, T., Lu, B., Yu, Z., Mei, X., Abulizi, P., & Ji, L. (2016).
tions: Biological actions and molecular mechanisms underpinning Lonicerae Japonicae Flos attenuates diabetic retinopathy by inhib-
their beneficial effects. Oxidative Medicine and Cellular Longevity, iting retinal angiogenesis. Journal of Ethnopharmacology, 189,
2012, 1–16. https​://doi.org/10.1155/2012/914273 117–125. https​://doi.org/10.1016/j.jep.2016.05.039
HERMAWATI et al.   
| 19

Zhu, C., Qiu, L., Wang, X., Hallin, U., Cande, C., Kroemer, G., …
Blomgren, K. (2003). Involvement of apoptosis‐inducing factor in How to cite this article: Hermawati E, Arfian N,
neuronal death after hypoxia‐ischemia in the neonatal rat brain. Mustofa, M, Partadiredja G. Chlorogenic acid
Journal of Neurochemistry, 86, 306–317. ameliorates memory loss and hippocampal cell death
Zhu, C., Xu, F., Wang, X., Shibata, M., Uchiyama, Y., Blomgren, K., &
after transient global ischemia. Eur J Neurosci.
Hagberg, H. (2006). Different apoptotic mechanisms are activated
2019;00:1–19. https​://doi.org/10.1111/ejn.14556​
in male and female brains after neonatal hypoxia‐ischaemia. Journal
of Neurochemistry, 96, 1016–1027. https​://doi.org/10.1111/j.1471-
4159.2005.03639.x

You might also like