Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Brain Research 1722 (2019) 146361

Contents lists available at ScienceDirect

Brain Research
journal homepage: www.elsevier.com/locate/brainres

Research report

Kaempferol attenuates neuroinflammation and blood brain barrier T


dysfunction to improve neurological deficits in cerebral ischemia/
reperfusion rats

Wei-Han Lia,b, Xiao Chenga,b, Ying-Lin Yanga,b, Man Liub, Shan-Shan Zhangb, Yue-Hua Wanga,b, ,

Guan-Hua Dua,b,
a
State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union
Medical College, Beijing 100050, China
b
Beijing Key Laboratory of Drug Target Identification and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical
College, Beijing 100050, China

H I GH L IG H T S

• Kaempferol attenuates inflammation after cerebral ischemia reperfusion.


• Kaempferol protects blood brain barrier integrity after cerebral ischemia reperfusion.
• The anti-inflammatory mechanism of kaempferol might be mediated by NF-κB pathway.

A R T I C LE I N FO A B S T R A C T

Keywords: Kaempferol has been reported to act as an anti-inflammatory agent in LPS-induced neuroinflammation in vitro
Cerebral ischemia/reperfusion (I/R) and in vivo, but its role in the inflammation after cerebral ischemia/reperfusion (I/R) is unclear. The present
Inflammation study was to investigate the effect of kaempferol on inflammation in ischemic brain tissue and explore its me-
Cytokines chanisms in cerebral I/R rats. Cerebral I/R rat model was established by middle cerebral artery occlusion for
Microglial activation
60 min and following reperfusion. Kaempferol at doses of 25, 50 and 100 mg/kg was administered for 7 days
NF-κB pathway
after cerebral I/R. Kaempferol treatment significantly reduced cerebral infarct volume, attenuated inflammation
and blood-brain barrier (BBB) disruption after cerebral I/R, thus improved neurological outcomes at the day 7
after cerebral I/R. Furthermore, the results also showed kaempferol treatment decreased the phosphorylation
and nuclear transposition of transcription factor NF-κB p65, thus inhibited expression of various pro-in-
flammatory proteins. In conclusion, kaempferol attenuates neuroinflammation and blood brain barrier dys-
function to improve neurological deficits in cerebral I/R rats, its mechanism is related to NF-κB pathway.

1. Introduction symptom (Manning et al., 2014; Thomalla et al., 2018). This poses a
challenge for the patients to obtain an effective therapy. Also, even
Ischemic stroke is a neurologic disorder characterized by blocking though the therapy is initiated by restoring blood flow, the damage can
cerebral artery, reducing oxygen and glucose supply to brain and still continue in neurons and glial cells due to reperfusion injury (Leech
causing damage to brain cells (Dawson and Dawson, 2017). Restoring et al., 2019). Therefore, controlling the pathological processes after
blood supply is critical in the treatment of ischemic stroke, one standard reperfusion is important to limit neurological damage (Wicha et al.,
therapeutic approach is to recanalize cerebral vascular and restore 2017; Zhang et al., 2018a,b).
blood flow using thrombolytic drug such as tissue plasminogen acti- Many evidences suggest the damage of reperfusion in neurons and
vator (Lekoubou et al., 2017). However, the thrombolytic drug needs to glial cells is mainly caused by inflammatory injury (Hoogland et al.,
be administered at a very limited time (e.g., 4.5 h) after the onset of 2015; Mu et al., 2016). Many cellular events have been proposed to


Corresponding authors at: Beijing Key Laboratory of Drug Target Identification and New Drug Screening, Institute of Materia Medica, Chinese Academy of
Medical Sciences & Peking Union Medical College, Beijing 100050, China.
E-mail addresses: wangyuehua@pku.org.cn (Y.-H. Wang), dugh@imm.ac.cn (G.-H. Du).

https://doi.org/10.1016/j.brainres.2019.146361
Received 19 June 2019; Received in revised form 29 July 2019; Accepted 31 July 2019
Available online 01 August 2019
0006-8993/ © 2019 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
W.-H. Li, et al. Brain Research 1722 (2019) 146361

contribute to inflammatory process after cerebral ischemia/reperfusion Meso-scale Multiplex Discovery plate to measure total nine in-
(I/R), such as activation of microglia. However, the roles of microglia in flammatory cytokines in brain tissues. As shown in Fig. 3, there was a
the inflammatory mechanisms following cerebral I/R are not fully un- significant increase in the production of KC/GRO (Fig. 3A), TNFα
derstood (Jiang et al., 2014; Jin et al., 2010; Lin et al., 2016; Zhan et al., (Fig. 3B), IL-1β (Fig. 3C), IL-5 (Fig. 3D) and IL-6 (Fig. 3E) in cerebral I/
2016). In addition, inflammation causes mitochondrial dysfunction, R rats as compared to sham group (All p < 0.01). However, there were
reducing superoxide dismutase (SOD) activities, disturbing balance no significant changes on IFN-γ (Fig. 3F), IL-4 (Fig. 3G), IL-13 (Fig. 3H),
between generating and detoxifying of reactive oxygen species (ROS) IL-10 (Fig. 3I) in I/R group as compared to sham group. Following
(Dawson and Dawson, 2017; Goyal et al., 2017; Li et al., 2017). Such kaempferol treatment, the elevated production of KC/GRO (Fig. 3A)
radicals attack cell membrane lipids, proteins, and glycosaminoglycans, and IL-5 (Fig. 3D) was significantly decreased at doses of 25, 50 and
causing further damage (Sies et al., 2017). Based on this information, 100 mg/kg. While TNF-α (Fig. 3B), IL-1β (Fig. 3C) and IL-6 (Fig. 3E)
understanding and interfering with the inflammatory process following was decreased significantly at higher dose of 100 mg/kg (Fig. 3B).
cerebral I/R are promising to find new therapeutic strategies for is-
chemic stroke. 2.4. Effects of kaempferol on the transcription and expression of iNOS and
Kaempferol (3, 4, 5, 7, -tetrahydroxyflavone, KAE), a natural fla- COX-2 in cerebral I/R rats
vonoid in variety of plants and some of traditional medicines, has been
increasingly investigated on its anti-inflammation and anti-oxidative To determine whether kaempferol inhibits other pro-inflammatory
effects (Rajendran et al., 2014; Yan et al., 2014). Its anti-inflammatory proteins in addition to inflammatory cytokines, the mRNA and protein
effect has been reported in cultured microglia and animal inflammatory expression of iNOS and COX-2 were examined by RT-PCR and Western
models induced by LPS (Cheng et al., 2018; Yang et al., 2019b; Park blotting. Compared with the Sham group, the mRNA and protein ex-
et al., 2011). Although the anti-inflammatory effect of kaempferol is pression of iNOS and COX-2 were both significantly increased in cere-
clear, its therapeutic potential and molecular mechanisms on in- bral I/R group (Fig. 4A and Fig. 4B). Following kaempferol treatment,
flammation caused by stroke remain unclear. In present study, we in- the expression of iNOS and COX-2 were dramatically attenuated
vestigated kaempferol therapeutic effect in cerebral I/R rats and ex- (Fig. 4C and Fig. 4D). These results indicated that the inhibition of iNOS
plored its potential mechanisms. and COX-2 transcription and protein expression might be one of me-
chanisms of kaempferol to attenuate the inflammatory response after
2. Results cerebral I/R.

2.1. Kaempferol reduced infarct volume and improved neurological deficits 2.5. Cytokine profile in ischemic cerebral tissue after in cerebral I/R rats
in cerebral I/R rats
To clarify the roles of cytokines playing in cerebral I/R, we set out a
The TTC staining and neurological deficits score were evaluated to cytokine profile using the Rat Cytokine Array Q67 (RayBiotech). The
investigate the protective effects of kaempferol on the ischemic brain above results have shown kaempferol at dose of 100 mg/kg exerts
tissues after cerebral I/R. The data showed that the infarct size of brain strong anti-inflammatory effect. Therefore, 100 mg/kg kaempferol
tissues was decreased in a dose-dependent manner by kaempferol treatment was selected for further study. We measured the 67 cytokines
treatment of 25-100 mg/kg. (Fig. 1A, 1B). Additionally, the brain tis- expression in ischemic cerebral tissue from Sham, cerebral I/R, cerebral
sues in kaempferol treatment groups showed a larger proportion of is- I/R + KAE-100 groups. In comparison with Sham group, 21 cytokines
chemic penumbra, especially at the high dose of 100 mg/kg. Neurolo- were found to be differently expressed in response to I/R (Fig. 5A). Out
gical deficits score was calculated to evaluate the protection of of the 21 cytokines, 13 cytokines marked in red (Fig. 5A) are upregu-
kaempferol on neurological function after I/R. We chose rats exhibiting lated and other 9 cytokines marked in blue (Fig. 5A) are down-
neurological deficits with the score of 1-3 at the 24 h after reperfusion regulated. From KEGG enrichment (Fig. 5B), we found the regulated
for the following experiments. On the day 1, neurological deficits genes mainly enriched in Th cell differentiation and TNF, JAK-STAT
showed no difference among ischemic groups. On the day 7, kaempferol signaling pathway. The results also showed kaempferol treatment suc-
treatment at the dose of 100 mg/kg significantly decreased neurological cessfully regulated proteins expression of 12 cytokines inculding TIMP-
deficits score (Fig. 1C). 1, MCP-1, ICAM-1, Notch-1, Notch-2, CNTF, P-Cadherin, Decorin,
Neuropilin-1, Galectin-1, Gas 1, and PDGF-AA after cerebral I/R
2.2. Kaempferol attenuated microglia activation in cerebral I/R rats (Fig. 5C).

Microglia is the major immune cell in the central nervous system, its 2.6. Effect of kaempferol on phosphorylation and nuclear translocation of
activation is one of the hallmarks of neuroinflammation. We performed NF-κB p65 in cerebral I/R rats
immunofluorescence staining to assess the effect of kaempferol on ac-
tivation of microglia. Iba-1 is a marker of activated microglia and often Microglia activation results in the activation of many signaling
used to characterize microglia cells, the activation of microglia cells pathways during the inflammatory response. The above results showed
includes the increasing in number, size and morphological change of that kaempferol attenuated inflammatory response in cerebral I/R rats.
microglia cells. However, the detailed signaling pathways and its regulation mechan-
As shown in the results (Fig. 2A), the number and size of microglia isms are still unclear. We performed Western blotting to measure the
were significantly increased in cerebral I/R group as compared to Sham phosphorylation of NF-κB p65 and IκB on day 7 in Sham, cerebral I/R
group. Also, the number and size of activated microglia was decreased and cerebral I/R + 100 mg/kg kaempferol treatment groups. The re-
in a dose-dependent manner by kaempferol treatment of 25–100 mg/kg sults showed that kaempferol treatment at dose of 100 mg/kg sig-
(Fig. 2B). These data suggested that kaempferol treatment attenuated nificantly decreased phosphorylation of NF-κB p65 (Fig. 6A). In com-
inflammation of ischemic brain tissues by inhibiting the microglia ac- parison, IκB was moderately phosphorylated in I/R group and
tivation after cerebral I/R. kaempferol had less effect on IκB phosphorylation (Fig. 6B). These re-
sults suggested kaempferol inhibited NF-κB activation by decreasing
2.3. Effect of kaempferol on inflammatory cytokines in cerebral I/R rats phosphorylation of NF-κB p65.
The phosphorylated NF-κB p65 can function as a transcriptional
To further evaluate the effect of kaempferol treatment on in- activator to increase expression of pro-inflammatory cytokines and
flammatory cytokines secretion, we conducted V-PLEX assays using exacerbate inflammation. To investigate translocation of NF-κB p65, we

2
W.-H. Li, et al. Brain Research 1722 (2019) 146361

Fig. 1. Effect of kaempferol on neurological function and cerebral infarction in cerebral I/R rats. (A) Representative images of TTC staining. (B) Statistical analysis of
infarct volume ratio (n = 4–5 in each group). (C) Statistical analysis of neurological scores at the day 7 (n = 6 in each group). Data were presented as mean ± SEM.
##
p < 0.01 vs. sham group; *p < 0.05 vs. I/R group.

extracted nuclear protein and cytoplasm protein using nuclear-cytosol permeability of BBB and exaggerated BBB disruption. In our study, we
extraction kit and performed Western blotting to detect the expression tested BBB permeability by measuring EB leakage. The amounts of EB
of NF-κB p65 in each sample. There were only small amounts of NF-κB leakage in ischemic hemisphere significantly increased in I/R group at
p65 detected in nuclear fraction in Sham group but significantly higher the day 7 after surgery. Kaempferol treatment markedly attenuated the
in I/R group (p < 0.01, Fig. 6C). The results indicated I/R activated leakage of EB in ischemic hemisphere (Fig. 7A). The finding demon-
NF-κB signal pathway and triggered NF-κB p65 nuclear translocation strated kaempferol treatment protected BBB integrity, decreasing the
from cytoplasm to nucleus. When treated with kaempferol at the dose of BBB permeability in cerebral I/R rats.
100 mg/kg, the NF-κB p65 nuclear transposition was significantly Matrix metalloproteinase 3 (MMP-3) is capable of breaking down
blocked (p < 0.05, Fig. 6C). In comparison, the amounts of NF-κB p65 many matrix compounds and activating other MMPs. In our study, we
had no significant difference in the cytoplasm fraction of Sham, cere- found the expression of this matrix metalloproteinase was upregulated
bral I/R cerebral I/R and cerebral I/R + kaempferol treatment groups significantly after I/R and kaempferol treatment decreased its expres-
(Fig. 6D). sion (Fig. 7B). Kaempferol attenuated MMP-3 expression in cerebral I/R
rats might be its one of the mechanisms to protect BBB integrity.

2.7. Kaempferol inhibited MMP-3 expression in ischemic cerebral tissue and


decreased BBB permeability in cerebral I/R rats 3. Discussion

Studies suggested inflammatory cytokines directly affected the The roles played by inflammation are changing in different stages of

3
W.-H. Li, et al. Brain Research 1722 (2019) 146361

firstly assessed effect of kaempferol on activation of microglia after


cerebral I/R. If microglia increased in number, size and changed in
morphology, they were judged as activated (Hoogland et al., 2015). In
the current study, we found that kaempferol inhibited the activation of
microglia after 7 days treatment in cerebral I/R rats.
Pro-inflammatory cytokines are biomarkers to evaluate extent of
inflammation (Doll et al., 2014). However, inflammatory cytokines are
complicated regulatory systems and strokes include a lot of subtypes in
clinical. Therefore, the prognostic value of inflammatory markers is
controversial in stroke patients. Active microglia secrete lots of in-
flammatory cytokines such as TNF-α, IL-6, IL-1β, which orchestrate
various molecular mechanisms involved in inflammation (Shabab et al.,
2017). Initial activation of microglia is beneficial to protect neurons,
but excessive inflammatory cytokines led to over-activation of micro-
glia which caused a cascade of inflammatory cytokines and un-
controlled inflammation. In our results, TNF-α, IL-6, IL-1β, IL-5, KC/
GRO were increased after cerebral I/R and attenuated by kaempferol
treatment. Interestingly, we found IL-2 and its receptor were down-
regulated in cerebral I/R group. This might be related to the decreasing
number of regulatory T cells (Tregs). Tregs are known to protect against
ischemic stroke, prevent autoimmunity and modulate inflammation
after cerebral I/R (Zhang et al., 2018a,b). However, kaempferol treat-
ment has no significant effect to the IL-2 level in cerebral I/R rats.
Besides, protein array results showed the increasing MCP-1 and
ICAM-1 could be significantly attenuated by kaempferol treatment.
MCP-1 is a chemokine that regulates migration and infiltration of
monocytes. ICAM-1 is an adhesion molecule promoting leukocyte trans-
endothelial migration (Jin et al., 2013). Both of two proteins promote
immune cell across the brain blood barrier into ischemic tissue, re-
leasing more pro-inflammation cytokines and exacerbating in-
flammatory response. Another important inflammatory biomakers,
iNOS and COX-2 were also decreased by kaempferol. They were vastly
secreted by microglia after being activated by reperfusion damage and
Fig. 2. Effect of kaempferol on microglia activation in ischemic cortices of caused series of inflammatory response (Balami et al., 2013;
cerebral I/R rats. (A) Representative images of microglia cells in cortices of Lambertsen et al., 2012). In conclusion, kaempferol inhibited over-ac-
ischemic hemispheres with positive immunochemical staining to Iba-1, scale
tivation of microglia and expression of pro-inflammatory protein in
bar = 50 μm. (B) Statistical analysis of integrated fluorescence intensity of Iba-
cerebral I/R rats.
1. Data were presented as mean ± SEM (n = 3 in each group). ##p < 0.01 vs.
sham group; *p < 0.05, **p < 0.01 vs. I/R group.
BBB integrity is important to function of nervous system (Zuo et al.,
2019). However, many neurologic diseases such as stroke caused neu-
roinflammation, oxidative stress and apoptosis, which break the BBB
stroke (Dawson and Dawson, 2017). During the acute phase, ischemic integrity (Li et al., 2019). In this study, we performed EB leakage ex-
tissue secretes pro-inflammatory cytokines rapidly, causing activation periment to test BBB integrity. The results showed EB leakage increased
of microglia and inflammatory response. Increasing MCP-1 and ICAM-1 significantly in cerebral I/R rats and kaempferol decreased EB leakage
promote leukocytes across the BBB into brain parenchyma, releasing into brain. From the result of the cytokine profile, we found the TIMP-1
more pro-inflammatory cytokines, which further amplify the in- expression increasing in I/R group and downregulated by kaempferol
flammation and cause death of neurons (Jin et al., 2013). However, in treatment. TIMP-1 is natural inhibitors of MMPs and the balance be-
chronic phase, inflammation is regarded as a beneficial factor to tissue tween TIMP-1 and MMPs plays an important role in inflammation
repair and functional recovery. Cytokines orchestrate the pathological (Eisner et al., 2017). It has been reported that kaempferol could sup-
processes in different stages after stroke (Lambertsen et al., 2012). press expression of MMPs (especially for MMP-3) in LPS-induced mi-
Accordingly, understanding mechanisms after stroke is important to croglia activation (Park et al., 2011). Therefore, we measured the MMP-
find new therapeutic strategies to stroke. 3 expression in cerebral I/R rats, data showed MMP-3 expression in-
In this study, we evaluated the protective effect of kaempferol to creased after I/R and was suppressed by kaempferol treatment. These
cerebral I/R injury and explored its mechanism. We found kaempferol results indicated kaempferol protected BBB integrity, which might be
treatment at dose of 100 mg/kg reduced infarct volume and neurolo- mediated by inhibiting MMP-3 expression.
gical deficits significantly after 7 days treatment. Then we set out a NF-κB p65 is an essential transcription factor involved in many
cytokine profile using protein array to explore the regulatory roles of types of cellular processes, phosphorylation of NF-κB p65 is required for
different cytokines. From KEGG enrichment, we found the significantly NF-κB activation and promoting transcription factor activity (Diel et al.,
regulated genes after ischemic stroke mainly enriched in immune cells 2011; Oeckinghaus et al., 2011; Yang et al., 2019a). NF-κB p65 was
differentiation and inflammatory signal pathways. This means in- activated in ischemic hemisphere, as determined by increased expres-
flammatory response still plays a key role at the day 7 after cerebral I/ sion of an NF-κB driven reporter transgene (Schneider et al., 1999).
R. Inhibiting NF-κB prior to MCAO protected ischemic cerebral tissue and
Kaempferol was reported as an anti-inflammatory agent eliciting reduced infarct size (Anttila et al., 2017). Based on the information,
protection to LPS-induced inflammation and microglia activation pharmacological inhibition of NF-κB is a rational therapeutic target to
(Cheng et al., 2018; Park et al., 2011). It also attenuated the expression diseases involved with NF-κB. Kaempferol was reported to inhibit NF-
of adhesion molecules, reduced oxidative stress. However, its protective κB activation in isoproterenol-induced heart failure in diabetic rats
effect on cerebral I/R rats have not been documented. In this study, we (Zhang et al., 2019). However, the effect of kaempferol on NF-κB

4
W.-H. Li, et al. Brain Research 1722 (2019) 146361

Fig. 3. Effect of kaempferol on inflammatory cytokines in ischemic cortices of cerebral I/R rats. The concentrations of each inflammatory cytokines were shown as
pg/mg protein. Data were presented as mean ± SEM (n = 6 in each group). ##p < 0.01 vs. sham group; *p < 0.05, **p < 0.01 vs. I/R group.

activation after ischemic stroke has not been reported. In this study, we and improving neurological outcome in experimental rat stroke model.
investigated the mechanisms to attenuate neuroinflammation following Inhibiting NF-κB pathway might be key factor among the mechanisms
kaempferol treatment and determined if NF-κB signal pathway was of kaempferol protecting from brain tissue damage after I/R injury.
involved in the pathological processes after cerebral I/R. Since phos- However, the pathological processes are complicated during cerebral I/
phorylation is a critical post-translational modification that regulates R, further studies are needed to explore the other signal pathways and
the activities of key components in NF-κB signaling pathway, we per- investigate their interaction with NF-κB. In addition, the inflammatory
formed Western blotting to verify the phosphorylation changes of NF- response is very complicated and lasts months after stroke. Further
κB p65 and its inhibitory protein IκB on day 7 after surgery. Phos- studies are needed to evaluate kaempferol in a long term. The results
phorylation of NF-κB p65 was significantly reduced at treatment of obtaining from the current study provided useful evidences in devel-
100 mg/kg kaempferol, while kaempferol treatment has less effect on oping kaempferol as a promising therapeutic drug to treat stroke pa-
the phosphorylation of IκB. This demonstrated that kaempferol in- tients, while the clinical effects have to be proved in the future.
hibited NF-κB pathway mainly from inhibiting phosphorylation of NF-
κB p65. In additon, the effect of kaempferol on nuclear translocation of
4. Material and methods
NF-κB p65 was also investigated in this study. The data showed
kaempferol treatment significantly inhibited NF-κB p65 nuclear trans-
4.1. Animals and experiment design
location after cerebral I/R. NF-κB p65 functions as a direct transcrip-
tional activator in cell nuclear, it could also affect promoter accessi-
110 male Sprague-Dawley rats (240–260 g) were purchased from
bility to transcription factors and thereby indirectly regulate their gene
Vital River Laboratory Animal Technology Co. (SCXK (Jing) 2016-
expression. In conclusion, the results suggested that the inhibition of
0006, Beijing, China). Animals were housed under standard tempera-
phosphorylation and nuclear translocation of NF-κB p65 is the one of
ture and humidity, diurnal lighting conditions and allowed food and tap
the mechanisms of kaempferol to attenuate inflammation after cerebral
water ad libitum. All animal care and experimental procedures were
I/R. This data is consistent with the studies that has shown transcription
approved by the ethic committees of Institute of Materia Medical,
factor NF-κB p65 is involved in the regulation of inflammation response
Chinese Academy of Medical Sciences & Peking Union Medical College.
after stroke (Schneider et al., 1999).
The focal cerebral I/R was established by transient MCAO for 60 min
In our study, kaempferol was proved to be a potential therapeutic
and following reperfusion. Briefly, after acclimatized for 3 days, rats
strategy to attenuate inflammation, reducing cerebral infarct volume
were anesthetized by the intraperitoneal injection of sodium

5
W.-H. Li, et al. Brain Research 1722 (2019) 146361

Fig. 4. Effect of kaempferol on transcription


and expression of iNOS and COX-2 in is-
chemic cortice of cerebral I/R rats. (A)
Protein expression of COX-2 (n = 4 in each
group). (B) mRNA expression of COX-2
(n = 6 in each group). (C) Protein expres-
sion of iNOS (n = 4 in each group). (D)
mRNA expression of iNOS (n = 6 in each
group). Data were presented as
mean ± SEM. #p < 0.05, ##p < 0.01 vs.
sham group; *p < 0.05 vs. I/R group.

pentobarbital (30 mg/kg). Heating pad was used to maintain a rectal 4.2. Neurological assessments and infarct volume evaluation
temperature of 37.0 ± 0.5 °C during ischemia period. The right
common carotid artery (CCA), external carotid artery (ECA) and in- To evaluate the degree of stroke in each animal following the sur-
ternal carotid artery (ICA) were isolated. Then, the standardized nylon gical procedure, neurological deficits assessments and brain infarct
suture (0.36 ± 0.02 mm, 2636A2, Beijing Cinontech, China) was in- volume were evaluated. Neurological deficits were assessed after
serted from the ECA into the ICA until the maker on suture shown on kaempferol treatment using neurobehavioral test, which is graded on a
the intersection of ECA and ICA, which indicated that the suture suc- five point scale as described in previous report (Yang, et al., 2003): 0,
cessfully blocked the MCA. Regional cerebral blood flow (rCBF) was no neurological deficits; 1, cannot extend left forelimb fully; 2, cannot
monitored using Laser Doppler Flowmetry (LDF) (PeriFlux 5000, walk straightly; 3, circle to the left; 4, inability to walk spontaneously or
PERIMED, Sweden), LDF showed that MCAO reduced blood flow to losing consciousness.
20% of the baseline during MCAO. After 60 min MCAO, nylon suture After neurological assessments, the rats were decapitated infarct
was removed to allow reperfusion, LDF showed blood flow returned volume analysis immediately. The brain tissues were dissected cor-
comparably in all animals. Rats were randomly divided into five onally into 2-mm slices using a scalpel. The sections were stained by 1%
groups: Sham group (Sham), cerebral I/R group (I/R), cerebral I/R with 2,3,5-tripenyltetrazolium chloride (TTC, Sigma, St. Louis, MO, USA) at
different doses of kaempferol groups: I/R + KAE 25 mg/kg, I/R + KAE 37 °C for 30 min, and then fixed in 4% paraformaldehyde at room
50 mg/kg and I/R + KAE 100 mg/kg. Rats in the sham operation group temperature overnight prior to imaging. In the stained sections, the red
underwent the same procedure without occlusion of the MCA. Rats that region represents the viable tissues and the white area indicates the
died or were not successfully established after MCAO surgery were infarct tissue. The infarct area in each section was evaluated using the
excluded from the experiments. Mortality in ischemic groups was Image J/Fiji processing system (Schindelin, et al., 2012). Infarct volume
13.8% (12/87) because of brain damage but not observed in Sham ratio = (The sum of infract area/ the sum of sections area) * 100%.
group. According to a five-point scale used in the study (Yang, et al.,
2003), the rats exhibiting no neurological deficits (score 0) after 24 h
4.3. Immunofluorescence detection of Iba-1
reperfusion were considered not successfully established stroke models
(2/75) and excluded from the experiments. The experiment design and
After kaempferol treatment, rats were anesthetized and perfused
using of each animal was shown in Fig. 8.
intracardially with PBS and then with 4% paraformaldehyde (PFA).
Kaempferol, purchased from Nanjing Zelang Biotechnology Co.,
Subsequently, the brain sections were fixed in 4% PFA overnight at 4 °C.
Ltd., was prepared in saline containing 0.5% sodium carbox-
The brain tissue sessions (20 μm) were obtained using a rotary micro-
ymethylcellulose (CMC-Na) and delivered by intragastric at doses of 25,
tome (RM2016, Leica, Germany) by paraffin section method, then im-
50 and 100 mg/kg body weight per day for 7 days, respectively. The
munofluorescence was performed on paraffin sections. Firstly, dewax
rats in the sham group and I/R group were treated with the vehicle
paraffin section to water and repair antigen in citric acid antigen repair
(0.5% CMC-Na in saline) in the same manner. Drug and vehicle were
buffer. Then blocked by 2% BSA at RT for 30 min. Spun the BSA slightly
administered in a blinded fashion.
and incubated the samples with ionized calcium binding adaptor mo-
lecule 1 (Iba-1) primary antibody (Wako: 019-19741, 1:200) at 4 °C
overnight. Primary antibodies were detected by using goat anti-rabbit
secondary antibodies (Wuhan Servicebio technology, China) for 50 min

6
W.-H. Li, et al. Brain Research 1722 (2019) 146361

Fig. 5. Cytokine profile assayed by Protein Array in ischemic cortices of cerebral I/R rats. (A) Interaction network of significantly regulated genes after cerebral I/R
(upregulation marked by red and downregulation marked by blue). (B) KEGG enrichment of significantly regulated genes after cerebral I/R. (C) Cytokine changes
regulated by kaempferol after cerebral I/R, the concentrations of regulated cytokines were shown as pg/mg protein in Fig. 6. Data were presented as mean ± SEM
(n = 4 in each group). #p < 0.05, ##p < 0.01 vs. sham group; *p < 0.05, **p < 0.01 vs. I/R group.

at room temperature. After washing with PBS for three times, spun PBS technology) for 10 min. All images were acquired using fluorescence
slightly and incubated with CY3 (Servicebio technology) for 10 min and microscope (Eclipse C1, Nikon, Japan). Fluorescence intensity was
wash it with TBST for 3 times. Spun slightly and added fluorescent analyzed using the image processing package Image J/Fiji (Schindelin,
quencher to the slice, incubated for 5 min. After washing with flowing et al., 2012). Cortices of ischemic hemispheres were defined to areas to
water for 10 min, cell nuclei was stained with DAPI (Servicebio be analyzed and representative images were presented (3 rats in each

7
W.-H. Li, et al. Brain Research 1722 (2019) 146361

Fig. 6. Effect of kaempferol on phosphorylation in NF-κB pathway in ischemic cortice of cerebral I/R rats. (A) Phosphorylation of NF-κB p65. (B) Phosphorylation of
IκB. (C) NF-κB p65 in nuclear fraction. (D) NF-κB p65 in cytoplasmic fraction. Data were presented as mean ± SEM (n = 4 in each group). ##p < 0.01 vs. sham
group; *p < 0.05 vs. I/R group.

group and 3 slides per rat). 4.5. Analysis of cytokine profile by protein array assay

The supernatant of ischemic cortices was handled with the same


4.4. Pro-inflammatory cytokine measurement by multiplex enzyme-linked procedure as Western blot, the amounts of protein were determined by
immunosorbent assay BCA protein assay kit (Thermo, Rockford, USA). Then we conduct
Protein Array assay (Cat#: QAR-CAA-67, RayBiotech, Norcross, GA)
At the necropsy, the brain tissues were quickly removed and the according to the manufacturer’s instructions. Firstly, block each chip
cortices of ischemic hemispheres were dissected for frozen in liquid with blocking buffer for 30 min. Decant the buffer and add 50 μg
nitrogen. When conducting the experiment, the tissue samples were samples to each well, incubating chips overnight at 4 °C. Decant the
homogenized in PBS buffer and then centrifuged at 13,000 rpm at 4 °C sample and wash 5 times with washing buffer. After that, incubate each
for 15 min. The supernatant was collected and the total amounts of chip with biotinylated antibody for 2 h, then incubate with Cy3
protein were determined by BCA protein assay kit (Thermo, Rockford, Equivalent Dye Streptavidin for 1 h, wash 5 times after each incubation.
USA) according to the manufacturer’s instructions. The levels of IL-1β, Finally, detect fluorescence at wave length of 532 nm with InnoScan
IL-6, TNF-α, IFN-γ, IL-2, IL-4, IL-5, IL-10, and KC/GRO were measured 300 Microarray Scanner (Innopsys, France). Gene interaction network
by immunoassay using V-PLex Pro-inflammmatory Panel 1 (rats) kit was generated by Cytoscape (Shannon et al., 2003) and KEGG enrich-
(Meso scale Discovery, Gaithersburg, USA) on the Sector Imager 2400 ment was performed by clusterProfiler database (R/Bioconductor).
according to the manufacturer’s instructions. The concentration of each
cytokine was normalized by the total amount of protein as measured
4.6. Evans blue (EB) leakage
from BCA protein assay.
At the day 7 after surgery, 4% Evans blue dye (Solarbio, Beijing,
China) dissolved in 0.9% saline was injected (0.25 ml/100 g) into the

8
W.-H. Li, et al. Brain Research 1722 (2019) 146361

Fig. 7. Effect of kaempferol on BBB permeability and


MMP-3 expression in ischemic cortice of cerebral I/R
rats. (A) Representative images of Evans blue
staining and quantitative analysis of Evans blue
content in ischemic hemisphere (n = 6 in each
group). (B) MMP-3 expression. (n = 4 in each group).
Data were presented as mean ± SEM. #p < 0.05 vs.
sham group; *p < 0.05 vs. I/R group.

tail vein of rats in each group. Two hours after injection, the rats were phospho-IκBα was normalized with total amount of NF-κB p65, IκBα,
anesthetized and transcardially perfused with saline until the outflow respectively.
fluid from the right atrium was clear. The brains were removed, dis-
sected coronally into 2-mm and imaged. The ischemic hemispheres
were collected immediately and homogenized in 50% trichloroacetic 4.8. Quantitative Real-Time PCR
acid (100 mg/300 μl) and centrifuged at 13,000 rpm at 4 °C for 5 min.
The supernatant was collected and determined their optical density Total RNA was extracted from the cortices of ischemic brain tissue
(OD) at 620 nm (Cheng et al., 2018). using Trizol reagent (Ambion, Carlsbad, CA, USA) according to the
manufacturer’s instructions. The concentration of RNA was measured
by ultraviolet spectrophotometry and determined using a formula A
4.7. Isolation of proteins and Western blot analysis
260 * dilution * 40 = μg RNA/mL. The cDNA was synthesized using the
PrimeScript™ RT reagent Kit (Takara Bio, Otsu, Japan). The primer
At the end of study, the cortices of ischemic hemisphere were col-
sequences used to measure mRNA expression were listed as followings:
lected to examine the expression of iNOS and COX-2 in each group,
5′-GCTGTACAAGCAGTGGCAAA-3′ (forward) and 5′- GTCTGGAGT
phosphorylation of NF-κB p65 and IκB in Sham, I/R and I/R + KAE-100
GGGAGGCACT-3′ (reverse) for COX-2;
treatment groups and also expression of MMP-3 in the three groups.
5′-TCATTGACCTCAACTACATGGT-3′ (forward) and 5′- CTAAGCA
Besides, the nuclear fraction and cytoplasmic fraction were extracted in
GTTGGTGGTGCAG-3′ (reverse) for GAPDH;
Sham, I/R and I/R + KAE-100 groups with nuclear-cytosol extraction
5′-GCATCCCAAGTACGAGTGGT-3′ (forward) and 5′- CCATGATGG
kit (Applygen Technologies Inc, Beijing, China) according to the man-
TCACATTCTGC-3′ (reverse) for iNOS.
ufacturer’s instructions. Following the extraction, nuclear translocation
The iNOS and COX-2 mRNA expression was measured using quan-
of NF-κB p65 was measured in three groups. The cortices of ischemic
titative Real-Time Polymerase Chain Reaction (RT-PCR) analysis. The
brain tissue were mixed with RIPA buffer including cocktail protease
samples were mixed in a final volume of 25 μL including 2 μL cDNA
inhibitor (100 mg/mL). Then the tissue was homogenized and cen-
synthesized before using SYBR® Premix Ex Taq II (TakaraBio, Otsu,
trifuged at 13000 rpm for 15 min. The supernatant was collected and
Japan). PCR was performed using the following protocol: 30 s at 95 °C,
mixed with loading buffer (Applygen Technologies Inc, Beijing, China)
followed by 40 cycles of 5 s at 95 °C, 30 s at 60 °C, and 15 s at 95 °C, 30 s
and heated at 100 ℃ for 10 min. Proteins were separated by sodium
for 60 °C and 15 s for 95 °C. With the relative quantification of 2−⊿⊿Ct
dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and
method, the transcription expression of target genes of iNOS, and COX-
transferred to PVDF membrane. The membrane was blocked with 5%
2 was determined using GAPDH as internal reference. GAPDH was
BSA in Tris-buffered saline with Tween-20 (TBS-T) for 1 h and then
wildly reported as a reference gene and validated no change on the day
incubated with anti-COX-2 antibody (ab15191, 1:1000), anti-iNOS
7 after I/R in our study.
antibody (ab204017, 1:500), anti-MMP-3 antibody (ab52915, 1:1000),
anti-phospho NF-κB p65 antibody (CST#3033, 1:1000), anti-NF-κB p65
antibody (CST#8242, 1:1000), anti-phospho IκBα antibody
4.9. Statistical analysis
(CST#2859, 1:1000), anti-IκBα antibody (CST#4814, 1:1000), anti-β-
actin antibody (C1313, 1:1000) and anti-histone H3 antibody
GraphPad Prism software version 7.04 (GraphPad Software, 7825
(orb136531, 1:1000) overnight at 4 °C, respectively. After washed with
Fay Avenue, Suite 230 La Jolla, CA 92037 USA) was performed to
TBS-T for 3 times, the membranes were incubated with secondary an-
compare the differences between each group. Data were presented as
tibody for 2 h at room temperature and then detected using enhanced
mean ± SEM. One-way ANOVA was used to calculate differences be-
ECL system. Optical density values were analyzed using the ImageJ/Fiji
tween the various groups. Tukey’s multiple comparison post hoc test
processing system. The signal density of iNOS, COX-2 and NF-κB p65 in
was performed to determine significance levels. Statistical significance
cytoplasm were normalized by total amount of internal β-actin protein.
was considered at p < 0.05.
The signal density of NF-κB p65 in nuclear was normalized by total
amount of histone H3. The signal density of phospho-NF-κB p65 and

9
W.-H. Li, et al. Brain Research 1722 (2019) 146361

Fig. 8. Experiment design. The rats in Experiment 1 were evaluated for infract volume. The rats in Experiment 2 were evaluated for Immunofluorescence detection of
Iba-1. In experiment 3 and 4, Western blot was performed to detect activation of NF-κB pathway and protein expression of MMP-3, iNOS and COX-2, PCR was
performed to detect mRNA expression, MSD was performed to measure inflammatory cytokine levels and Protein Array was also performed to make the Cytokine
Profile. The rats in Experiment 5 were evaluated for Evens blue leakage.

5. Authors’ contributions Declaration of Competing Interest

Yue-Hua Wang and Guan-Hua Du designed the experiments; Wei- All authors declare that they have no competing interests.
Han Li, Xiao Cheng, Ying-Lin Yang, Man Liu, and Shan-Shan Zhang
performed the animal experiments; Wei-Han Li and Yue-Hua Wang Appendix A. Supplementary data
analyzed the data and wrote the manuscript; and all authors read and
approved the final manuscript. Supplementary data to this article can be found online at https://
doi.org/10.1016/j.brainres.2019.146361.

Funding
References

This project was supported by the National Key Research &


Anttila, J.E., et al., 2017. Role of microglia in ischemic focal stroke and recovery: focus on
Development Plan of China (2018YFC0311005); the National Natural Toll-like receptors. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 79, 3–14.
Science Foundation of China (81473383), the Significant New-Drugs Balami, J.S., Chen, R.L., Buchan, A.M., 2013. Stroke syndromes and clinical management.
Creation of Science and Technology Major Projects of China QJM 106, 607–615.
Cheng, X., Yang, Y.L., Yang, H., Wang, Y.H., Du, G.H., 2018. Kaempferol alleviates LPS-
(2018ZX09711001-003-019), the Medical and Health Innovation induced neuroinflammation and BBB dysfunction in mice via inhibiting HMGB1 re-
Project of Chinese Academy of Medical Sciences in China (2016-I2M-3- lease and down-regulating TLR4/MyD88 pathway. Int. Immunopharmacol. 56,
007), and Innovation Fund for Doctoral Students of Beijing Union 29–35.
Dawson, T.M., Dawson, V.L., 2017. Mitochondrial mechanisms of neuronal cell death:
Medical College in China (2018-1007-04).

10
W.-H. Li, et al. Brain Research 1722 (2019) 146361

potential therapeutics. Annu. Rev. Pharmacol. Toxicol. 57, 437–454. cerebral ischemia. Nat. Med. 5, 554–559.
Diel, D.G., Luo, S., Delhon, G., Peng, Y., Flores, E.F., Rock, D.L., 2011. A nuclear inhibitor Shabab, T., Khanabdali, R., Moghadamtousi, S.Z., Kadir, H.A., Mohan, G., 2017.
of NF-kappaB encoded by a poxvirus. J. Virol. 85, 264–275. Neuroinflammation pathways: a general review. Int. J. Neurosci. 127, 624–633.
Doll, D.N., Barr, T.L., Simpkins, J.W., 2014. Cytokines: their role in stroke and potential Shannon, P., Markiel, A., Ozier, O., Baliga, N.S., Wang, J.T., Ramage, D., Amin, N.,
use as biomarkers and therapeutic targets. Aging Dis. 5, 294–306. Schwikowski, B., Ideker, T., 2003. Cytoscape: a software environment for integrated
Eisner, L., Vambutas, A., Pathak, S., 2017. The balance of tissue inhibitor of metallo- models of biomolecular interaction networks. Genome Res. 13, 2498–2504.
proteinase-1 and matrix metalloproteinase-9 in the autoimmune inner ear disease Sies, H., Berndt, C., Jones, D.P., 2017. Oxidative stress. Annu. Rev. Biochem. 86,
patients. J. Interferon Cytokine Res. 37, 354–361. 715–748.
Goyal, N., Kashyap, B., Singh, N.P., Kaur, I.R., 2017. Neopterin and oxidative stress Thomalla, G., Simonsen, C.Z., Boutitie, F., Andersen, G., Berthezene, Y., Cheng, B.,
markers in the diagnosis of extrapulmonary tuberculosis. Biomarkers 22, 648–653. Cheripelli, B., Cho, T.H., et al., 2018. MRI-guided thrombolysis for stroke with un-
Hoogland, I.C., Houbolt, C., van Westerloo, D.J., van Gool, W.A., van de Beek, D., 2015. known time of onset. N. Engl. J. Med. 379, 611–622.
Systemic inflammation and microglial activation: systematic review of animal ex- Wicha, P., Tocharus, J., Janyou, A., Jittiwat, J., Changtam, C., Suksamrarn, A., Tocharus,
periments. J. Neuroinflamm. 12, 114. C., 2017. Hexahydrocurcumin protects against cerebral ischemia/reperfusion injury,
Jiang, M., Li, J., Peng, Q., Liu, Y., Liu, W., Luo, C., Peng, J., Li, J., Yung, K.K., Mo, Z., attenuates inflammation, and improves antioxidant defenses in a rat stroke model.
2014. Neuroprotective effects of bilobalide on cerebral ischemia and reperfusion PLoS One 12, e0189211.
injury are associated with inhibition of pro-inflammatory mediator production and Yan, Y., Min, Y., Min, H., Chao, C., Ying, Q., Zhi, H., 2014. n-Butanol soluble fraction of
down-regulation of JNK1/2 and p38 MAPK activation. J. Neuroinflamm. 11, 167. the water extract of Chinese toon fruit ameliorated focal brain ischemic insult in rats
Jin, R., Liu, L., Zhang, S., Nanda, A., Li, G., 2013. Role of inflammation and its mediators via inhibition of oxidative stress and inflammation. J. Ethnopharmacol. 151,
in acute ischemic stroke. J. Cardiovasc. Transl. Res. 6, 834–851. 176–182.
Jin, R., Yang, G., Li, G., 2010. Inflammatory mechanisms in ischemic stroke: role of in- Yang, M.Y., Yu, Q.L., Huang, Y.S., Yang, G., 2019a. Neuroprotective effects of andro-
flammatory cells. J. Leukoc. Biol. 87, 779–789. grapholide derivative CX-10 in transient focal ischemia in rat: involvement of Nrf2/
Lambertsen, K.L., Biber, K., Finsen, B., 2012. Inflammatory cytokines in experimental and AE and TLR/NF-kappaB signaling. Pharmacol. Res. 144, 227–234.
human stroke. J. Cereb. Blood Flow Metab. 32, 1677–1698. Yang, Y.L., Cheng, X., Li, W.H., Liu, M., Wang, Y.H., Du, G.H., 2019b. Kaempferol at-
Leech, T., Chattipakorn, N., Chattipakorn, S.C., 2019. The beneficial roles of metformin tenuates LPS-induced striatum injury in mice involving anti-neuroinflammation,
on the brain with cerebral ischaemia/reperfusion injury. Pharmacol. Res. 104261. maintaining BBB integrity, and down-regulating the HMGB1/TLR4 pathway. Int. J.
Lekoubou, A., Awoumou, J.J., Kengne, A.P., 2017. Incidence of seizure in stroke patients Mol. Sci. 20, 491. https://doi.org/10.3390/ijms20030491.
treated with recombinant tissue plasminogen activator: A systematic review and Yang, Y.R., Wang, R.Y., Wang, P.S., 2003. Early and late treadmill training after focal
meta-analysis. Int. J. Stroke 12, 923–931. brain ischemia in rats. Neurosci. Lett. 339, 91–94.
Li, Q., Cui, J., Fang, C., Liu, M., Min, G., Li, L., 2017. S-adenosylmethionine attenuates Zhan, J., Qin, W., Zhang, Y., Jiang, J., Ma, H., Li, Q., Luo, Y., 2016. Upregulation of
oxidative stress and neuroinflammation induced by amyloid-β through modulation of neuronal zinc finger protein A20 expression is required for electroacupuncture to
glutathione metabolism. J. Alzheimers Dis. 58, 549–558. attenuate the cerebral inflammatory injury mediated by the nuclear factor-kB sig-
Li, S., Bian, L., Fu, X., Ai, Q., Sui, Y., Zhang, A., Gao, H., Zhong, L., Lu, D., 2019. Gastrodin naling pathway in cerebral ischemia/reperfusion rats. J. Neuroinflamm. 13, 258.
pretreatment alleviates rat brain injury caused by cerebral ischemic-reperfusion. Zhang, H., Xia, Y., Ye, Q., Yu, F., Zhu, W., Li, P., Wei, Z., Yang, Y., Shi, Y., Thomson, A.W.,
Brain Res. 1712, 207–216. Chen, J., Hu, X., 2018a. In vivo expansion of regulatory T cells with IL-2/IL-2 anti-
Lin, R., Cai, J., Kostuk, E.W., Rosenwasser, R., Iacovitti, L., 2016. Fumarate modulates the body complex protects against transient ischemic stroke. J. Neurosci. 38,
immune/inflammatory response and rescues nerve cells and neurological function 10168–10179.
after stroke in rats. J. Neuroinflamm. 13, 269. Zhang, L., et al., 2019. The protective effect of kaempferol on heart via the regulation of
Manning, N.W., Campbell, B.C., Oxley, T.J., Chapot, R., 2014. Acute ischemic stroke: Nrf2, NF-kappabeta, and PI3K/Akt/GSK-3beta signaling pathways in isoproterenol-
time, penumbra, and reperfusion. Stroke 45, 640–644. induced heart failure in diabetic rats. Drug Dev. Res. 80, 294–309.
Mu, S., Liu, B., Ouyang, L., Zhan, M., Chen, S., Wu, J., Chen, J., Wei, X., Wang, W., Zhang, Zhang, W.W., Xu, F., Wang, D., Ye, J., Cai, S.Q., 2018b. Buyang Huanwu Decoction
J., Lei, W., 2016. Characteristic changes of astrocyte and microglia in rat striatum ameliorates ischemic stroke by modulating multiple targets with multiple compo-
induced by 3-NP and MCAO. Neurochem. Res. 41, 707–714. nents: In vitro evidences. Chin. J. Nat. Med. 16, 194–202.
Oeckinghaus, A., Hayden, M.S., Ghosh, S., 2011. Crosstalk in NF-kappaB signaling Zuo, X., Lu, J., Manaenko, A., Qi, X., Tang, J., Mei, Q., Xia, Y., Hu, Q., 2019. MicroRNA-
pathways. Nat. Immunol. 12, 695–708. 132 attenuates cerebral injury by protecting blood-brain-barrier in MCAO mice. Exp.
Park, S.E., Sapkota, K., Kim, S., Kim, H., Kim, S.J., 2011. Kaempferol acts through mi- Neurol. 316, 12–19.
togen-activated protein kinases and protein kinase B/AKT to elicit protection in a
model of neuroinflammation in BV2 microglial cells. Br. J. Pharmacol. 164,
1008–1025. Further reading
Rajendran, P., Rengarajan, T., Nandakumar, N., Palaniswami, R., Nishigaki, Y., Nishigaki,
I., 2014. Kaempferol, a potential cytostatic and cure for inflammatory disorders. Eur. Wu, G., McBride, D.W., Zhang, J.H., 2018. Axl activation attenuates neuroinflammation
J. Med. Chem. 86, 103–112. by inhibiting the TLR/TRAF/NF-kappaB pathway after MCAO in rats. Neurobiol. Dis.
Schindelin, J., Arganda-Carreras, I., Frise, E., Kaynig, V., Longair, M., Pietzsch, T., 110, 59–67.
Preibisch, S., Rueden, C., Saalfeld, S., Schmid, B., Tinevez, J.Y., White, D.J., Zhong, Y.H., Dhawan, J., Kovoor, J.A., Sullivan, J., Zhang, W.X., Choi, D., Biegon, A.,
Hartenstein, V., Eliceiri, K., Tomancak, P., Cardona, A., 2012. Fiji: an open-source 2017. Aromatase and neuroinflammation in rat focal brain ischemia. J. Steroid
platform for biological-image analysis. Nat. Methods 9, 676–682. Biochem. Mol. Biol. 174, 225–233.
Schneider, A., et al., 1999. NF-kappaB is activated and promotes cell death in focal

11

You might also like