ArticlePDF Available

The Effect of Oleoylethanolamide (OEA) Add-On Treatment on Inflammatory, Oxidative Stress, Lipid, and Biochemical Parameters in the Acute Ischemic Stroke Patients: Randomized Double-Blind Placebo-Controlled Study

Wiley
Oxidative Medicine and Cellular Longevity
Authors:

Abstract and Figures

Methods: Sixty patients with a mean age of 68.60 ± 2.10 comprising 29 females (48.33%), who were admitted to an academic tertiary care facility within the first 12 hours poststroke symptoms onset or last known well (LKW), in case symptom onset time is not clear, were included in this study. AIS was confirmed based on a noncontrast head CT scan and also neurological symptoms. Patients were randomly and blindly assigned to OEA of 300 mg/day (n = 20) or 600 mg/day (n = 20) or placebo (n = 20) in addition to the standard AIS treatment for three days. A blood sample was drawn at 12 hours from symptoms onset or LKW as the baseline followed by the second blood sample at 72 hours post symptoms onset or LKW. Blood samples were assessed for inflammatory and biochemical parameters, oxidative stress (OS) biomarkers, and lipid profile. Results: Compared to the baseline, there is a significant reduction in the urea, creatinine, triglyceride, high-density lipoprotein, cholesterol, alanine transaminase, total antioxidant capacity, malondialdehyde (MDA), total thiol groups (TTG), interleukin-6 (IL-6), and C-reactive protein levels on the follow-up blood testing in the OEA (300 mg/day) group. In patients receiving OEA (600 mg/day) treatment, there was only a significant reduction in the MDA level comparing baseline with follow-up blood testing. Also, the between-group analysis revealed a statistically significant difference between patients receiving OEA (300 mg/day) and placebo in terms of IL-6 and TTG level reduction when comparing them between baseline and follow-up blood testing. Conclusion: OEA in moderate dosage, 300 mg/day, add-on to the standard stroke treatment improves short-term inflammatory, OS, lipid, and biochemical parameters in patients with AIS. This effect might lead to a better long-term neurological prognosis.
Content may be subject to copyright.
Research Article
The Effect of Oleoylethanolamide (OEA) Add-On Treatment on
Inflammatory, Oxidative Stress, Lipid, and Biochemical
Parameters in the Acute Ischemic Stroke Patients: Randomized
Double-Blind Placebo-Controlled Study
Mohammadmahdi Sabahi ,
1,2
Sara Ami Ahmadi,
1
Azin Kazemi,
1
Maryam Mehrpooya ,
3
Mojtaba Khazaei ,
4
Akram Ranjbar ,
5,6
and Ashkan Mowla
7
1
Neurosurgery Research Group (NRG), Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
2
Behavioral Disorders and Substances Abuse Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
3
Department of Clinical Pharmacy, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
4
Department of Neurology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
5
Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
6
Nutrition Health Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
7
Department of Neurological Surgery, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA, USA
Correspondence should be addressed to Mojtaba Khazaei; khazaeimojtaba@yahoo.com
and Akram Ranjbar; akranjbar2015@gmail.com
Mohammadmahdi Sabahi, Sara Ami Ahmadi, and Azin Kazemi contributed equally to this work.
Received 25 January 2022; Revised 1 April 2022; Accepted 5 June 2022; Published 8 September 2022
Academic Editor: Anna M. Giudetti
Copyright © 2022 Mohammadmahdi Sabahi et al. This is an open access article distributed under the Creative Commons
Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work
is properly cited.
Background and Objective. There is a growing body of evidence for the ecacy of oleoylethanolamide (OEA) in patients with
inammatory disorders. The present randomized double-blind placebo-controlled study is aimed at evaluating the ecacy of
OEA add-on treatment in patients with acute ischemic stroke (AIS). Methods. Sixty patients with a mean age of 68:60 ± 2:10
comprising 29 females (48.33%), who were admitted to an academic tertiary care facility within the rst 12 hours poststroke
symptoms onset or last known well (LKW), in case symptom onset time is not clear, were included in this study. AIS was
conrmed based on a noncontrast head CT scan and also neurological symptoms. Patients were randomly and blindly
assigned to OEA of 300 mg/day (n=20) or 600 mg/day (n=20) or placebo (n=20) in addition to the standard AIS treatment
for three days. A blood sample was drawn at 12 hours from symptoms onset or LKW as the baseline followed by the second
blood sample at 72 hours post symptoms onset or LKW. Blood samples were assessed for inammatory and biochemical
parameters, oxidative stress (OS) biomarkers, and lipid prole. Results. Compared to the baseline, there is a signicant
reduction in the urea, creatinine, triglyceride, high-density lipoprotein, cholesterol, alanine transaminase, total antioxidant
capacity, malondialdehyde (MDA), total thiol groups (TTG), interleukin-6 (IL-6), and C-reactive protein levels on the follow-
up blood testing in the OEA (300 mg/day) group. In patients receiving OEA (600 mg/day) treatment, there was only a
signicant reduction in the MDA level comparing baseline with follow-up blood testing. Also, the between-group analysis
revealed a statistically signicant dierence between patients receiving OEA (300 mg/day) and placebo in terms of IL-6 and
TTG level reduction when comparing them between baseline and follow-up blood testing. Conclusion. OEA in moderate
dosage, 300 mg/day, add-on to the standard stroke treatment improves short-term inammatory, OS, lipid, and biochemical
parameters in patients with AIS. This eect might lead to a better long-term neurological prognosis.
Hindawi
Oxidative Medicine and Cellular Longevity
Volume 2022, Article ID 5721167, 11 pages
https://doi.org/10.1155/2022/5721167
1. Introduction
The prevalence and mortality of noncommunicable diseases
have surpassed infectious diseases globally. Neurological dis-
orders such as stroke, Alzheimers, and Parkinsons are asso-
ciated with high mortality and morbidity, and thus far, there
is no cure for them. These disorders have revealed common
pathological features such as inammation, oxidative stress
(OS) production, abnormal protein accumulation, disrup-
tion of normal calcium homeostasis, and apoptosis. Among
these, stroke is one of the leading causes of death worldwide
and a major cause of disability in adults [13].
Stroke is a neurological disorder that is divided into
ischemic and hemorrhagic types [4, 5]. In the ischemic type,
the hypoxia of the brain tissue occurs with the cessation of
blood ow to the brain tissue and leads to the destruction
of neurons and glial cells [2, 3, 6, 7]. The sequence of the
events responding to ischemia is known as the ischemic cas-
cade, including glutamate release, calcium inux, OS,
inammation, and ultimately, apoptosis, which leads to irre-
versible neuronal death [8, 9].
Oleoylethanolamide (OEA) is a member of the N-
acylethanolamine (NAE) family, which are endogenously bio-
active lipids and are formed from amidation of membrane
phospholipids. This fatty acid is mostly found in sources such
as olives and sesame [10, 11]. Pure olive oil counteracts cell
death by reducing lipid peroxidation (LPO), brain prostaglan-
din E2, and nitric oxide production, while increasing glutathi-
one concentrations. The family of NAEs is involved in a wide
rangeofbodyprocessessuchasinammation, nerve protec-
tion,acutestress,painrelief,anxiety,hypotension,sleep,and
energy balance [11]. Based on previous ndings, using olive
oil fatty acid supplements reduces damage and protects the
brain tissue during an ischemic stroke [3]. Peroxisome
proliferator-activated receptor alpha (PPAR-α) and cannabi-
noid receptors (CBR) have critical roles in regulating inam-
mations [12]. The activation of the CBR1 can protect neural
tissues against ischemic injury or tissue reperfusion injury by
reducing OS, releasing lactate dehydrogenase, and activating
caspase-3 in vitro [13]. The members of the NAE family have
been identied as the natural ligands of the CBR. In addition
to the CBR, OEA can bind to PPAR-α, and consequently, exert
its anti-inammatory eects [12], since PPAR-αacts as a neg-
ative regulator of the inammatory response through direct
binding to the p65nuclearfactor(NF)-κB, which subsequently
antagonizes NF-κB transcription factor pathways [14, 15].
This study sought to determine the eect of OEA supple-
mentation on the biomarkers of OS, inammatory parame-
ters, lipid prole, and renal and hepatic parameters in
patients with acute ischemic stroke.
2. Materials and Methods
2.1. Trial Setting and Design. This single-center three-day
randomized, placebo-controlled, double-blind, parallel-
group trial was conducted in Farshchian (Sina) Hospital,
an aliate of the Hamadan University of Medical Sciences,
Hamadan, Iran. All patients were screened and enrolled
between April 21
st
, 2020 and July 22
nd
, 2020.
2.2. Standard Protocol Approvals, Registrations, and Patient
Consents. The trial was registered in the Iranian Registry of
Clinical Trials (Identier: IRCT20130501013194N4) and
approved by the Ethics Committee of the Hamadan Univer-
sity of Medical Sciences with the ethical code of IR.UM-
SHA.REC.1398.720-722. Before participation in the study,
all participants and/or their next of kin signed a written
informed consent form in compliance with the Declaration
of Helsinki.
2.3. Patients. In general, 82 patients were assessed for their
eligibility; among whom, 12 and 10 patients did not meet
the inclusion criteria or declined to participate, respectively.
As such, 60 patients aged 30-93 years who met the inclusion
and exclusion criteria were consented to participate in the
study (Figure 1). No patients lost to follow-up since they were
all admitted in the hospital for at least three days after the
onset of their neurological symptoms or last known well
(LKW), in case stroke onset time was unclear. The National
Institute of the Health Stroke Scale (NIHSS) score and the
modied Rankin Scale (mRS) were used to measure the neuro-
logical decits and functional outcome poststroke, respec-
tively. The NIHSS is a scoring system which oers a
quantitative assessment neurological decit caused by stroke.
This 15-item scale measures the decits caused by acute stroke
on several domains including awareness, language, eye move-
ments, motor force, ataxia, and sensorium. Each item is
graded on a three- to ve-point scale leading to scores between
0 and 42, where 0 indicates no decit and 42 translates to
death [16]. Furthermore, the mRS is a functional outcome
measure, which ranges from 0 to6, where 0 denes as no neu-
rological symptoms and 6 indicates death [17].
The patients were included if they were reaching the
emergency department within 12 hours from the neurologi-
cal symptom onset or LKW in case the onset time was
unclear, had no evidence of intracranial hemorrhage on their
initial noncontrast head CT, and had NIHSS score 2 and
<20 [18]. Exclusion criteria is as follows: patients aged <18,
had a temperate 38.0
°
C and/or a white blood cell count
16,000 cells/mm
3
, given we did this study during the
COVID pandemic, history or suspicion of active malig-
nancy, inammatory vasculopathy, systemic inammatory
disease, connective tissue disease, hypercoagulable state,
and chronic renal failure, as well as patients who had a glo-
merular ltration rate <30, hepatic failure and/or cirrhosis,
gastrointestinal bleeding, uncontrolled diabetes mellitus,
chronic respiratory diseases, hematologic illnesses, and sei-
zure at presentation. Also, use of medications/supplements,
having either anti-inammatory or anti-OS eect, prior sen-
sitivity to compounds, alcohol and drug abuse, and a vegeta-
tive state were the other exclusion criteria.
These 60 patients underwent quick neurological assess-
ment upon admission, as per standard practice [19, 20].
Patients meeting both inclusion and exclusion criteria were
divided into two categories. Patients who were eligible for
acute reperfusion therapy including intravenous and/or
intra-arterial thrombolytic therapy and those who were not
eligible for acute treatment. In both categories, in addition
to OEA or placebo, patientsunderlying diseases such as
2 Oxidative Medicine and Cellular Longevity
diabetes mellitus, hypertension, and dyslipidemia were
addressed as per standard of care. They also receive anti-
platelets or anticoagulants when appropriate as per standard
of care. Patients were not allowed to receive any additional
treatment in the course of the trial.
The baseline blood sample was drawn at 12 hours after
LKW, testing OS, and lipid, along with renal and hepatic
parameters during the acute phase. The follow-up sample
was drawn at 72 hours after LKW.
Patients were randomly divided into three groups using
Minitab software, patients who received OEA 300 mg/day,
600 mg/day, or placebo capsules with respect to concealment,
respectively. All three groups received the treatment for three
consecutive days. Each individual received a computer code,
and the code was disclosed during the treatment period. The
allocation was hidden behind a series of sequentially num-
bered, opaque, and sealed envelopes. A study coordinator
opened the envelope after the recruitment of each patient.
The patients were randomly assigned and assessed by two
physicians. The statistician, the patients, the referring physi-
cian, and the physician evaluating the patients and providing
the trial drugs, were all blinded to the allocation. The treat-
ment codes were revealed at the end of the study and after
closing the database. All three groups were adjusted for age
and gender, extent and severity of ischemic stroke using the
NIHSS score, baseline mRS, acute infarct volumes on noncon-
trast head CT, door to needle time in case intravenous throm-
bolytic therapy was administered, whether acute reperfusion
therapy was performed, body mass index, history of smoking,
and history of hypertension. Furthermore, groups were
adjusted for history of hyperlipidemia, coronary artery disease,
diabetes mellitus, deep vein thrombosis, prior stroke, systolic
and diastolic blood pressure, and home medication prior to
admission (Table 1). Frequency matching methods were used
for this purpose.
2.4. Chemicals. The applied chemicals in this investigation
included 5,5-Dithiobis-(2-nitrobenzoic acid) (DTNB),
2,4,6-tripyridyl-S-triazine (TPTZ), Thiobarbituric acid
(TBA), Nitro blue tetrazolium (NBT), and sodium phos-
phate buer that were obtained from the Sigma-Aldrich
(St. Louis, MO, USA). OEA supplements were provided by
Karen Pharma and Food Supplement Co.
2.5. Biochemical Analysis. Phlebotomy was employed to col-
lect ten milliliters of venous blood into chilled ethylenedi-
aminetetraacetic acid-containing tubes, which were then
centrifuged for plasma separation, aliquoted in 1.5 ml vials,
Allocation
Randomized (n=60)
Assessed for eligibility (n=82)
Excluded (n=22)
󳴴 Not meeting inclusion criteria (n=12)
󳴴 Declined to participate (n=10)
Enrollment
Intervention groups, OEA 300 mg/day
(n=20) and OEA 600mg/day (n=20)
Placebo group
(n=20)
Lost to follow-up (n=0)
Non-adherence to treatment (n=0)
Adverse eects (n=0)
Lost to follow-up (n=0)
Non-adherence to treatment (n=0)
Adverse eects (n=0)
Analysed (n=40)
󳴴 Excluded from analysis (n=0)
Analysed (n=20)
󳴴 Excluded from analysis (n=0)
Analysis
Figure 1: The ow diagram of the study.
3Oxidative Medicine and Cellular Longevity
snap-frozen, and kept at 80
°
C until further analyses. After the
treatment period, interleukin- (IL-) 6 levels were measured in
the blood samples of the subjects by enzyme-linked immuno-
sorbent assay, and C-reactive protein (CRP) levels were mea-
sured by turbidometry. Portion of the blood sample was sent
to the laboratory to measure the lipid prole including triglyc-
erides (TG), total cholesterol, and high-density lipoprotein
(HDL) in addition to serum hematological parameters com-
prising urea, creatinine, aspartate transaminase (AST), and ala-
nine transaminase (ALT). On the other portion of the blood
sample, the following parameters were measured: LPO level
by assessing malondialdehyde (MDA), capacity of the serum
antioxidants which is called total antioxidant capacity (TAC),
the level of total thiol groups (TTG), and the activity of the
superoxide dismutase (SOD) enzyme by a spectrophotometer
using the TBA reagent at 532 nm, the TPTZ reagent at
593 nm, the DTNB reagent at 412 nm, and the NBT reagent
at 560 nm wavelength, respectively.
Table 1: Demographic and clinical characteristics of the study participants.
Number (%) or mean ± SD
All (n=60)Placebo
(n=20)
OEA 300 mg/day
(n=20)
OEA 600 mg/day
(n=20)Pvalue
Male/female 31/29 11/9 10/10 10/10 0.935
Age (years) 68:60 ± 2:10 65:4±3:21 67:2±2:65 69:7±3:09 0.648
BMI (kg/m
2
)26:9±4:12 27:2±3:71 26:4±4:32 27:1±3:45 0.762
Risk factors
Smoking 34 (56.6%) 11 (55%) 9 (45%) 14 (70%) 0.275
Hypertension 51 (85%) 16 (80%) 17 (85%) 18 (90%) 0.676
Hyperlipidemia 47 (78.3%) 14 (70%) 16 (80%) 15 (75%) 0.766
CAD 35 (58.3%) 17 (85%) 12 (60%) 16 (80%) 0.155
Diabetes mellitus 16 (26.6%) 5 (25%) 4 (20%) 7 (35%) 0.551
Prior stroke 17 (28.3%) 7 (35%) 4 (20%) 6 (30%) 0.563
DVT 6 (10%) 1 (5%) 2 (10%) 2 (10%) 0.804
SBP, mmHg 149 ± 8:29 152 ± 11:01 149 ± 10:73 148 ± 12:14 0.742
DBP, mmHg 87 ± 10:02 87 ± 8:73 89 ± 10:20 88 ± 12:65 0.933
Acute ischemic
stroke event
characteristics
NIHSS 5:2±1:26 5:12 ± 1:72 5:18 ± 1:50 5:26 ± 1:37 0.790
Infarct volume (cm
3
)1:2±0:87 1:1±1:02 1:2±0:98 1:4±1:25 0.720
mRS 2:1±0:34 2:03 ± 0:53 2:12 ± 0:36 1:96 ± 0:27 0.502
DTN, hours 8:3±4:57 7:2±5:19 7:9±4:21 8:3±4:71 0.191
Thrombolytic therapy 27 (45%) 9 (45%) 8 (40%) 10 (50%) 0.817
Nonthrombolytic therapy 33 (55%) 11 (55%) 12 (60%) 10 (50%) 0.817
Medications prior
to admission
Antiplatelet therapy 18 (30%) 8 (40%) 4 (20%) 6 (30%) 0.386
Anticoagulation therapy 18 (30%) 8 (40%) 4 (20%) 6 (30%) 0.386
Statin therapy 47 (78.3%) 14 (70%) 16 (80%) 15 (75%) 0.766
ACEI therapy 50 (83.3) 16 (80%) 17 (85%) 17 (85%) 0.877
ARB therapy 4 (6.6%) 1 (5%) 2 (10%) 1 (5%) 0.765
BB therapy 29 (48.3%) 11 (55%) 8 (40%) 10 (50%) 0.627
CCB therapy 36 (60%) 12 (60%) 14 (70%) 10 (50%) 0.435
Diuretics therapy 9 (15%) 3 (15%) 3 (15%) 3 (15%) 1.00
Insulin with oral
antidiabetic therapy 10 (16.6%) 3 (15%) 2 (10%) 5 (25%) 0.432
Oral anti diabetic therapy 6 (10%) 2 (10%) 2 (10%) 2 (10%) 1.00
Underlying mechanism
for stroke
Atherothrombotic 33 (55%) 12 (60%) 10 (50%) 11 (55%) 0.817
Cardioembolic 9 (15%) 3 (15%) 2 (10%) 4 (20%) 0.676
Lacunar infract 9 (15%) 3 (15%) 3 (15%) 3 (15%) 1.00
Other causes 6 (10%) 2 (10%) 2 (10%) 2 (10%) 1.00
Undetermined 3 (5%) 1 (5%) 1 (5%) 1 (5%) 1.00
Abbreviations: BMI, body mass index; CAD, coronary artery disease; DVT, deep vein thrombosis; SBP, systolic blood pressure; DBP, diastolic blood pressure;
NIHSS, National Institutes of Health Stroke Scale; mRS, Modied Rankin scale; DTN, door-to-needle time; ACEI, angiotensin-converting-enzyme inhibitors;
ARB, Angiotensin II receptor blockers; BB, beta blocker; CCB, calcium channel blocker.
4 Oxidative Medicine and Cellular Longevity
2.6. Statistical Analysis. Descriptive analysis and comparison
of dierences between demographics in each group were
performed by SPSS 16 software. The mean, standard devia-
tion, frequency, and percentage were used to represent the
data. The Kolmogorov-Smirnov test was applied to deter-
mine if the distribution was normal. The Chi-squared test
and Fishers exact test were employed for comparing qualita-
tive variables between the groups. In addition, the indepen-
dent t-test and MannWhitney Utest were used to
compare quantitative variables. Furthermore, intragroup dif-
ferences between baseline and follow-up parameters were
examined using the Wilcoxon signed-rank test. Finally, the
between-group dierence analysis was conducted through
the Kruskal-Wallis test, followed by a post hoc test, and P
values less than 0.05 were considered statistically signicant.
2.7. Data Availability Policy. Any competent investigator can
request anonymized data from the corresponding author for
replicating techniques and ndings.
3. Results
Thirty-one men (51.66%) and 29 women (48.33%) partici-
pated in this study, and their mean age was 68:60 ± 2:10
(range 30-93). Demographic and clinical characteristics of
the study participants including the cardiovascular risk fac-
tors, acute ischemic stroke event characteristics, medications
prior to admission, and the underlying mechanism for
stroke are summarized in Table 1. No signicant dierences
were observed between the baseline characteristics of these
three groups. Our primary outcome was to evaluate the
eect of OEA on inammatory and OS parameters. The sec-
ondary outcome was to investigate the eect of OEA on the
lipid prole, as well as renal and hepatic parameters.
3.1. Renal Parameters. As shown in Table 2, although the
between-group analysis revealed no statistically signicant
dierence in the blood urea and creatinine levels, there was
a statistically signicant decline in the level of urea in
patients receiving 300 mg/day OEA. Overall, OEA 300 mg/
day reduced both urea and creatinine levels and might act
as a renoprotective factor in acute ischemic stroke patients.
3.2. Lipid Prole. As demonstrated in Table 2, a statistically
signicant reduction in total cholesterol and TG and a rise
in HDL levels were seen in patients receiving OEA 300 mg/
day, while changes in other groups were not statistically sig-
nicant in either between-group or within-group analysis.
3.3. Hepatic Parameters. As shown in Table 2, although
between-group analysis revealed no statistically signicant
dierence regarding ALT and AST levels, there was a statis-
tically signicant decline in the level of ALT in patients
receiving OEA 300 mg/day.
3.4. OS Parameters. The levels of OS parameters are shown
in Table 2. Although SOD levels have no signicant dier-
ence between the groups, within-group analysis indicated
that consumption of OEA could signicantly alter TAC,
MDA only in the 300 mg/day group, and TTG in both
300 mg/day and 600 mg/day groups. Also, between-group
analysis indicated statistically signicant dierences in
TTG, and the results of post hoc test revealed that this dier-
ence was noticeable between patients receiving OEA 300 mg/
day and a placebo (P=0:047).
3.5. Inammatory Prole. The levels of inammatory param-
eters are displayed in Table 2. In within-group analysis,
patients who received moderate dose of OEA, 300mg/day,
showed statistically signicant decrease in IL-6 and CRP
levels. Between-group analysis demonstrated statistically sig-
nicant dierences in IL-6, and based on the results of post
hoc test, this dierence was observed between patients receiv-
ing OEA, 300mg/day, and placebo (P=0:037).
3.6. Complications. Table 3 presents the occurrence of
adverse eects. Nausea, vomiting, dyspepsia, and headache
were the most frequently reported adverse eects among
the participants regardless of their treatment group. There
were no statistically signicant dierences among dierent
groups regarding the reported side eects.
4. Discussion
OS and inammation are considered as an important path-
ophysiological mechanism in acute ischemic stroke. OS is a
consequence of the failure in the equilibrium between the
endogenous reactive oxygen species (ROS) production and
their cleansing by endogenous antioxidant defense systems
[21]. The rapid increase in ROS production immediately
after acute ischemic stroke quickly overwhelms the antioxi-
dant capacity of the brain tissue and causes further tissue
damage. ROS can damage cell macromolecules, resulting in
autophagy, apoptosis, and necrosis. Furthermore, the rapid
restoration of the blood ow after reperfusion increases the
level of oxygen delivery to the tissue, leading to a new wave
of ROS production and thus further tissue damage [22].
Inammation is also one of the main pathological mecha-
nisms of acute ischemic stroke and may cause OS [23]. Vari-
ous cytokines such as tumor necrosis factor (TNF), IL-1, and
IL-6 are known to regulate the tissue damage in stroke models,
and therefore, play a crucial role in poststroke therapies. The
eect of these cytokines on the development and progression
of infarction in human and animal modelsdepends on various
factors, including their availability in the penumbra of the
stroke area at the onset of early symptoms [24].
Diet and lifestyle are known to play important roles in
preventing noncommunicable diseases. High-fat diets may
cause metabolic changes, and obesity can induce chronic
inammation. Diet-induced obesity and related metabolic
disorders such as hyperlipidemia are also considered risk
factors for cardiovascular diseases and stroke [25].
A Mediterranean diet, which has widely known to be
benecial on health, is a diet that is characterized by the
presence of useful bioactive compounds such as monounsat-
urated fatty acids (MUFAs) and polyunsaturated fatty acids
or polyphenols [26].
OEA is derived from the unsaturated fatty acid oleic
acid, which is part of MUFA. This fatty acid is mostly found
5Oxidative Medicine and Cellular Longevity
Table 2: Laboratory characteristics of the study participants.
Lab parameter Before or after
receiving the capsule
Mean ±SD Pvalue
Placebo (n=20) OEA 300 mg/day (n=20) OEA 600 mg/day (n=20)
Ur
Before 43:16 ± 10:98 52:25 ± 12:65 41:8±12:63
0.796
After 38:5±12:34 42:25 ± 8:22 36:6±14:04
Pvalue 0.56 0.046 0.168
Cr
Before 1:12 ± 0:32 1:34 ± 0:15 1:06 ± 0:17
0.466
After 1:05 ± 0:23 1:17 ± 0:24 0:98 ± 0:13
Pvalue 0.353 0.030 0.210
TG
Before 109:5±37:22 116:12 ± 44:87 108 ± 60:81
0.964
After 77:75 ± 7:22 88:37 ± 27:72 74:5±20:5
Pvalue 0.126 0.044 0.449
Chl
Before 149:5±41:58 163:4±37:62 156:5±30:55
0.659
After 135:25 ± 47:61 136:8±28:60 140:9±30:96
Pvalue 0.217 0.027 0.138
HDL
Before 47:85 ± 28:32 33:25 ± 6:38 35 ± 3:69
0.317
After 47:42 ± 30:70 37:83 ± 5:89 39 ± 7:89
Pvalue 0.834 0.035 0.288
ALT
Before 16:71 ± 6:87 19:33 ± 4:50 15 ± 3:31
0.170
After 18:57 ± 7:67 18:22 ±4:65 14:2±1:92
Pvalue 0.374 0.007 0.374
AST
Before 13:5±2:51 19:66 ± 2:65 19:57 ± 7:16
0.424
After 16 ± 8:28 19 ± 2:75 19:14 ± 6:98
Pvalue 0.591 0.235 0.078
IL-6
Before 2:42 ± 1:32 3:24 ± 1:81 3:20 ± 1:57
0.035
After 3:18 ± 1:54 2:13 ± 0:59 3:26 ± 1:19
Pvalue 0.216 0.040 0.888
CRP
Before 4:25 ± 3:77 4:98 ± 3:16 4:17 ± 2:20
0.235
After 4:59 ± 4:12 3:04 ± 1:54 4:44 ± 1:28
Pvalue 0.831 0.049 0.743
SOD
Before 0:24 ± 0:011 0:251 ± 0:013 0:25 ± 0:01
0.369
After 0:25 ± 0:018 0:253 ± 0:023 0:24 ± 0:02
Pvalue 0.187 0.792 0.435
TAC
Before 0:59 ± 0:08 0:63 ± 0:082 0:70 ± 0:08
0.464
After 0:61 ± 0:12 0:71 ± 0:099 0:73 ± 0:14
Pvalue 0.637 0.020 0.478
MDA
Before 0:089 ± 0:011 0:106 ± 0:021 0:097 ± 0:026
0.054
After 0:090 ± 0:017 0:091 ± 0:018 0:080 ± 0:016
Pvalue 0.820 0.026 0.007
TTG
Before 0:140 ± 0:017 0:151 ± 0:016 0:155 ± 0:017
0.022
After 0:134 ± 0:025 0:167 ± 0:016 0:151 ± 0:017
Pvalue 0.494 0.016 0.266
Abbreviations: Cr, Creatinine; Ur, Urea; Chl, Cholesterol; TG, Triglyceride; HDL, high-density lipoprotein; AST, aspartate transaminase; ALT, alanine
transaminase; CRP, C-reactive protein; IL-6, interlukin-6; TAC, total antioxidant capacity; TTG, total thiol groups; MDA, malondialdehyde; SOD,
superoxide dismutase.
6 Oxidative Medicine and Cellular Longevity
in olives and sesame [10, 11]. In an experimental study on
the eect of high MUFA diet on cerebral ischemia, an
improvement was observed in the neurological and motor
function in acute ischemic stroke mice models receiving
olive oil fatty acid supplements compared to placebo [3].
Targeting CBR2 has several eects on ROS-induced neu-
roinammation, as it can reduce ROS/reactive nitrogen spe-
cies (RNS) production in active glial cells, reduce vascular
inammation, improve blood-brain barrier (BBB) function,
and inhibit leukocyte cell uptake and thus reduce nerve cell
death [13].
The results of the present study conrmed the eect of
OEA supplementations on the biomarkers of OS, inamma-
tory parameters (IL-6, CRP, and lipid prole), and renal and
hepatic parameters in patients with acute ischemic stroke.
Our investigation indicated that OEA has remarkable
eects on OS parameters, which can reduce OS and increase
anti-OS marker in the patients with acute ischemic stroke. In
a neuron-like SH-SY5Y cell line, Giusti et al. observed that
10μM oleocanthal, as a phenolic component of extra virgin
olive oil (EVOO), neutralizes OS which was induced by
H
2
O
2
, and leads to increased cell viability, decreased produc-
tion of ROS, and an increased intracellular glutathione
(GSH) level [27]. Furthermore, Tasset et al. demonstrated that
EVOO, which represents 10% of calorie intake in the total
standard daily diet of rats, reduces oxidative damage in Hun-
tingtons disease-like rat model induced by 3-nitropropionic
acid (3NP) [28]. They further found that in all studied sam-
ples, 3NP increased lipid peroxides but decreased GSH levels
[28]. However, their results revealed that EVOO reduced the
level of LPO and blocked the GSH deciency caused by 3NP
in the striatum and other parts of the brain of Wistar rats
[28]. In our study,TAC, TTG, and MDA levels had signicant
changes, conrming the antioxidant eects of OEA.
Previous studies have shown that mitochondrial CBR1
expression plays a unique role in cannabinoid-driven neuro-
protection and might directly regulate mitochondrial ROS
formation under this pathological condition [13]. Moreover,
targeting CBR2 has several eects on ROS-induced neuroin-
ammation, as it can reduce ROS/RNS production in active
glial cells, reduce vascular inammation, improve blood-
brain barrier function, inhibit leukocyte cell uptake, and
consequently, decrease nerve cell death [13]. Therefore, the
antioxidative eect of OEA in our study might be due to
the activation of both CBR1 and CBR2.
Although the neuroprotective eects of OEA after acute
cerebral ischemic injury in the experimental model have
been reported, to the best of our knowledge, there are no
clinical trials on humans to prove this concept [29]. It has
been indicated that OEA (40 mg/kg, intraperitoneally (ip))
attenuates apoptosis by inhibiting the Toll-like receptor
(TLR4)/NF-κB and ERK1/2 signaling pathways in mice
model of acute ischemic stroke [29]. N15, an analogue of
OEA, has the ability to protect the brain against ischemic
injury. Li et al. assessed both neuropreventive (50, 100, or
200 mg/kg, ip) and neurotherapeutic eects of N15
(200 mg/kg, ip) in mice model of acute ischemic stroke and
also lipopolysaccharide- (LPS-) stimulated BV-2 microglial
cells [30]. Furthermore, the anti-inammatory properties
of N15 may contribute to its neuroprotective eects on cere-
bral ischemia, at least in part, by boosting PPAR/dual signal-
ing and suppressing the activation of the NF-κB, STAT3,
and ERK1/2 signaling pathways [30]. These data imply that
OEA might be a promising therapy option for ischemic
stroke prevention and treatment.
The evaluation of inammatory parameters revealed that
in the OEA 300 mg/day group, the IL-6 and CRP levels were
signicantly dierent before and after OEA administration.
Contrarily, these levels were not signicantly dierent in
other groups before and after OEA administration.
Between-group analysis showed that IL-6 in patients receiv-
ing OEA 300 mg/day was statistically lower than its level in
patients who received placebo. A controlled clinical trial
evaluated the anti-inammatory and antioxidative eects
of OEA (250 mg/day) on obesity, as well as assessing LPO,
TAC, CRP, IL-6, and TNF-αlevels. Based on the results,
IL-6 and TNF-αconcentrations were signicantly reduced
in the intervention group, but other changes were not signif-
icantly dierent [31]. Sayd et al. demonstrated that systemic
administration of OEA (10 mg/kg, ip) could decrease levels
of IL-1βand IL-6 as proinammatory cytokines and also
markers of nitrosative/OS nitrites and MDA which was
induced by LPS (0.5 mg/kg, ip) in rats [32]. In another study,
Xu et al. investigated the anti-inammatory eect of dier-
ent doses of OEA (10 μM, 20 μM, 50 μM, and 100 μM) and
concluded that OEA reduces inammatory cytokines (IL-6
and IL-8) and adhesion molecules on TNF-α(20 ng/ml)-
induced inammation in human umbilical vein endothelial
cells through the activation of the CBR2 and PPAR-α[12].
Zhou et al. assessed both neuropreventive (10, 20,or 40 mg/
kg, intragavage (ig)) and neurotherapeutic eects of OEA
(40 mg/kg, ig) in a mice model of ischemic stroke and con-
cluded that orally administered OEA protects mice from
focal cerebral ischemic injury particularly BBB disruption
by activating PPAR-α[33]. This impact is tremendously
important since poststroke BBB disruption can exacerbate
ischemic damage by raising edema and inducing bleeding
[34]. During an acute ischemic stroke, cerebral edema is
the most prevalent cause of neurological impairment and
death [35]. The BBBs integrity can help avoid brain swelling
and subsequent tissue damage. Since OEA is rapidly
depleted in vivo due to hydrolysis, its therapeutic potential
is limited. As a result, encapsulating OEA in a nanoparticu-
late structure like cubosomes, which may be utilized to target
Table 3: Frequency of drug-related adverse eects among patients
in each groups.
Number (%)
All
(n=60)
Placebo
(n=20)
OEA
300 mg/day
(n=20)
OEA
600 mg/day
(n=20)
P
value
Nausea 16 (26.6%) 4 (20%) 6 (30%) 6 (30%) 0.711
Vomiting 11 (18.3%) 3 (15%) 4 (20%) 4 (20%) 0.895
Dyspepsia 24 (40%) 6 (30%) 8 (40%) 10 (50%) 0.435
Headache 26 (43.3%) 7 (35%) 10 (50%) 9 (45%) 0.622
Dizziness 15 (25%) 5 (25%) 5 (25%) 5 (25%) 1.000
7Oxidative Medicine and Cellular Longevity
the BBB, protects it from hydrolysis and allows therapeutic
amounts to reach the brain [36]. Another study by Wu
et al. showed that the survival rate, behavioral score, cerebral
infarct volume, edema degree, spatial learning, and memory
capacity of stroke-model rats could all be improved greatly
using endogenous OEA crystals loaded lipid nanoparticles
[37]. Further studies should be done in order to nd the
most eective format for OEA administration.
Likewise, Luo et al. in both in vitro and in vivo experi-
ments found that OEA (10-50 μM) inhibits glial activation
via modulating PPAR-αand promotes motor function
recovery after brain ischemia [38]. Also, SUL (3 and
10 mg/kg) treatment, a stable OEA-modeled compound, in
addition to PPAR-αantagonist, GW6471 (1 mg/kg),
improves the brain damage and accompanying motor and
cognitive impairments caused by hypoxia-ischemia in mice,
most likely through regulating alterations in neuroinam-
mation/immune system mediators [39].
A meta-analysis demonstrated that acute kidney injury is
a common complication following acute ischemic stroke and
is associated with increased mortality following acute ische-
mic stroke [40]. Additionally, it was shown that impaired
kidney function is associated with the presence of cerebral
microbleeds in acute ischemic stroke [41], and renal dys-
function increases the risk of recurrent stroke in those
patients [42]. Thus, kidney function preservation is crucially
important in acute ischemic stroke patients. In our study,
within-group analysis revealed that blood urea and creati-
nine, as the markers of renal function, had signicantly
changed in patients receiving OEA 300 mg/day, indicating
that OEA might also be used as a renal protective factor in
acute ischemic patients. Thus, OEA not only has no renal
toxicity character, but also can prevent deleterious eects
on kidneys.
In terms of the lipid prole, within-group analysis indi-
cated a statistically signicant decrease in the TG and cho-
lesterol levels, while a rise in the HDL level in patients
receiving OEA 300 mg/day. In terms of hepatic function, a
statistically signicant reduction was found in ALT in
patients receiving OEA 300 mg/day. It is concluded that
OEA has no hepatotoxic character, but can improve liver
function at moderate doses (300 mg/day). In an experimen-
tal study on KDS-5104, a nonhydrolyzable lipid OEA ana-
log, Thabuis et al. showed that the most signicant
bioindicator of OEA activity is adipose tissue fatty-acid
translocase (FAT)/CD36 expression, which appears to be a
determining player in the OEA fat-lowering response [43].
In addition, Fu et al. demonstrated PPAR-αand other
PPAR-target genes, such as FAT/CD36, liver fatty-acid bind-
ing protein (L-FABP), and uncoupling protein-2 (UCP-2),
are activated by subchronic OEA administration (5 mg/kg/
day, ip, for two weeks) in Zucker rats [44]. Furthermore,
OEA lowers hepatocyte neutral lipid content as well as blood
cholesterol and TG levels. The ndings imply that OEA
might modulate lipid metabolism [44].
Similarly, Li et al. evaluated the eect of 17 weeks of
OEA administration (5 mg/kg/day, ip) on nonalcoholic fatty
liver disease (NAFLD) in Sprague Dawley rats and found
that the treatment with OEA delayed the progression of
NAFLD by regulating plasma TG and cholesterol levels
and reducing ALT, AST, and inammatory liver cytokines
compared with controls [45]. On the other hand, the study
of the liver and plasma tissue gene expression in these ani-
mal models showed that OEA increases lipid oxidation
through PPAR-αactivation [45]. The results of this study
also represented that treatment with OEA inhibits the
expression of genes involved in fat synthesis [45].
Higher levels of TG than HDL-C are associated with pre-
mature neuronal degradation, while lower ratios are linked
with early clinical improvements. Several studies reported
that TG/HDL-C can predict mortality and worsen clinical
outcomes after acute ischemic stroke, and thus it is a simple
and inexpensive indicator for predicting disease prognosis.
The TG/HDL-C ratio is independently associated with mor-
tality and poor prognosis in acute ischemic stroke patients
[46, 47]. As such and considering the importance of lower-
ing lipid prole, OEA might be considered in patients with
acute ischemic stroke.
In terms of preclinical use of OEA in acute ischemic
stroke treatment, an experimental study on Sprague Dawley
rats demonstrated that chronic OEA therapy (30 mg/kg/day
for 28 days) can promote neurogenesis in the hippocampus
through increasing the expression of brain-derived neuro-
trophic factor (BDNF) and PPAR-αresulting in functional
recovery of cognitive decits and neuroprotective benets
against cerebral ischemic insult, indicating that OEA might
be used therapeutically for cerebral ischemia [48].
In addition, by increasing collagen content and decreas-
ing necrotic core size in plaques and also by modulating
macrophage polarization both via the AMPK- PPAR-α
pathway, OEA increased atherosclerotic plaque stability in
both in vivo and in vitro experiments [49]. These data imply
preventive role of OEA in ischemic events.
In general, a wide range of studies have pointed out the
role of OS, inammatory parameters, and lipid prole in
the pathogenesis and prognosis of acute ischemic stroke.
Accordingly, it is recommended that OEA, as a member of
the NAE family, might be used to reduce the complications
of acute ischemic stroke.
4.1. Limitations and Strengths. Despite the uniqueness of our
ndings, several limitations warn against extrapolating the
ndings too far including a relatively small number of
patients enrolled in this study. More studies with larger sam-
ple sizes are warranted. The second drawback of this study is
that each patient only had two samples obtained at baseline
and 72 hours posttreatment. As a result, long-term under-
standing of the biomarker changes postacute ischemic stroke
is not feasible. Third, the patients in our trial were given
OEA supplements orally. As such, no conclusion can be
withdrawn on intravenous (IV) OEA treatment eects.
Fourth, testing the eects of OEA supplementation in a
short length of time and lack of long-term follow-up is a dis-
advantage. Fifth, the eects of OEA on clinical outcome of
these patients were not assessed. Sixth, the window for treat-
ment with OEA in our trial was within 12 hours following
the onset of stroke symptoms; earlier treatment may have
dierent ecacy. Finally, IV thrombolytics are the only
8 Oxidative Medicine and Cellular Longevity
FDA-approved pharmacological treatment for acute ische-
mic stroke [5060] and roughly half of our patients received
this treatment given they were eligible. Thus, this treatment
might have inuenced our results.
Our study has the following advantages. It beneted
from randomization, double-blinding, and the presence of
a control group as a pilot clinical trial. Furthermore, the
changes in inammatory markers after treatment with
OEA might be an indication of reduction in brain tissue
inammation happening after acute ischemic stroke.
5. Conclusion
Our results indicate that OEA add-on to standard acute
ischemic stroke treatment improves the short-term inam-
matory, OS status, and lipid and biochemical parameters in
those patients, particularly in moderate dosage, 300 mg/
day, which might lead to the better functional outcome.
Our ndings need to be conrmed in larger-scale studies
with larger sample sizes and longer intervention duration
and follow-up.
Data Availability
The authors conrm that the data supporting the ndings of
this study are available within the article.
Ethical Approval
All procedures performed in this study involving human
participants were in accordance with the ethical standards
and approved by the Hamadan University of Medical Sci-
ences Ethical Committee (IR.UMSHA.REC.1398.720-722)
and the 1964 Helsinki Declaration and its later amendments
or comparable ethical standards. The trial was registered in
the Iranian Registry of Clinical Trials (Identier: IRCT2013
0501013194N4).
Consent
Informed consent was obtained from all individual partici-
pants and/or their proxies included in the study. Informed
consent to participate in this study was obtained from par-
ticipants included in the study. All authors give their consent
to the submission of this article to the Oxidative Medicine
and Cellular Longevity.
Conflicts of Interest
All authors certify that they have no aliations with or
involvement in any organization or entity with any nancial
interest (such as honoraria; educational grants; participation
in speakersbureaus; membership, employment, consultan-
cies, stock ownership, or other equity interest; and expert
testimony or patent-licensing arrangements) or nonnancial
interest (such as personal or professional relationships, al-
iations, knowledge or beliefs) in the subject matter or mate-
rials discussed in this manuscript.
AuthorsContributions
Mohammadmahdi Sabahi, Sara Ami Ahmadi, and Azin
Kazemi contributed equally to this work.
References
[1] P. Reboredo-Rodríguez, A. Varela-López, T. Y. Forbes-Her-
nández et al., Phenolic compounds isolated from olive oil as
nutraceutical tools for the prevention and management of can-
cer and cardiovascular diseases,International Journal of
Molecular Sciences, vol. 19, no. 8, p. 2305, 2018.
[2] C. Angeloni, M. Malaguti, M. Barbalace, and S. Hrelia, Bioac-
tivity of olive oil phenols in neuroprotection,International
Journal of Molecular Sciences, vol. 18, no. 11, p. 2230, 2017.
[3] R. Gonzalo-Gobernado, M. I. Ayuso, L. Sansone et al., Neuro-
protective eects of diets containing olive oil and DHA/EPA in
a mouse model of cerebral ischemia,Nutrients, vol. 11, no. 5,
p. 1109, 2019.
[4] M. Dolatshahi, M. Sabahi, S. Shahjouei, E. Koza, V. Abedi, and
R. Zand, Intravenous thrombolysis in ischemic stroke
patients with a prior intracranial hemorrhage: a meta-analy-
sis,Therapeutic Advances in Neurological Disorders, vol. 15,
p. 175628642210741, 2022.
[5] M. A. AbdelRazek, A. Mowla, D. Hojnacki et al., Prior asymp-
tomatic parenchymal hemorrhage does not increase the risk
for intracranial hemorrhage after intravenous thrombolysis,
Cerebrovascular Diseases, vol. 40, no. 5-6, pp. 201204, 2015.
[6] R. Rikhtegar, M. Youse, S. Dolati et al., Stem cell-based cell
therapy for neuroprotection in stroke: a review,Journal of
Cellular Biochemistry, vol. 120, no. 6, pp. 88498862, 2019.
[7] K. Singh, A. Mowla, S. Mehla et al., Safety of intravenous
thrombolysis for acute ischemic stroke in patients with preex-
isting intracranial neoplasms: a case series,International
Journal of Stroke, vol. 10, no. 3, pp. E29E30, 2015.
[8] M. I. Ayuso, E. Martínez-Alonso, C. Cid, M. A. De Leciñana,
and A. Alcázar, The translational repressor eIF4E-binding
protein 2 (4E-BP2) correlates with selective delayed neuronal
death after ischemia,Journal of Cerebral Blood Flow &
Metabolism, vol. 33, no. 8, pp. 11731181, 2013.
[9] B. C. White, J. M. Sullivan, D. J. DeGracia et al., Brain ische-
mia and reperfusion: molecular mechanisms of neuronal
injury,Journal of the Neurological Sciences, vol. 179, no. 1-2,
pp. 133, 2000.
[10] K. J. Bowen, P. M. Kris-Etherton, G. C. Shearer, S. G. West,
L. Reddivari, and P. J. Jones, Oleic acid-derived oleoylethano-
lamide: a nutritional science perspective,Progress in Lipid
Research, vol. 67, pp. 115, 2017.
[11] F. Fanelli, M. Mezzullo, A. Repaci et al., Proling plasma N-
acylethanolamine levels and their ratios as a biomarker of obe-
sity and dysmetabolism,Molecular Metabolism, vol. 14,
pp. 8294, 2018.
[12] X. Xu, H. Guo, Z. Jing et al., N-oleoylethanolamine reduces
inammatory cytokines and adhesion molecules in TNF-α-
induced human umbilical vein endothelial cells by activating
CB2 and PPAR-α,Journal of Cardiovascular Pharmacology,
vol. 68, no. 4, pp. 280291, 2016.
[13] J. Paloczi, Z. V. Varga, G. Hasko, and P. Pacher, Neuroprotec-
tion in oxidative stress-related neurodegenerative diseases:
role of endocannabinoid system modulation,Antioxidants
& Redox Signaling, vol. 29, no. 1, pp. 75108, 2018.
9Oxidative Medicine and Cellular Longevity
[14] A. Zambon, P. Gervois, P. Pauletto, J.-C. Fruchart, and
B. Staels, Modulation of hepatic inammatory risk markers
of cardiovascular diseases by PPARαactivators,Arterioscle-
rosis, Thrombosis, and Vascular Biology, vol. 26, no. 5, pp. 977
986, 2006.
[15] M. Dolatshahi, M. H. Ranjbar Hameghavandi, M. Sabahi, and
S. Rostamkhani, Nuclear factor-kappa B (NF-κB) in patho-
physiology of Parkinson disease: diverse patterns and mecha-
nisms contributing to neurodegeneration,European Journal
of Neuroscience, vol. 54, no. 1, pp. 41014123, 2021.
[16] D. C. Tong, M. A. Yenari, G. W. Albers, M. O'brien, M. P.
Marks, and M. E. Moseley, Correlation of perfusion- and
diusion-weighted MRI with NIHSS score in acute (<6.5
hour) ischemic stroke,Neurology, vol. 50, no. 4, pp. 864
869, 1998.
[17] G. Sulter, C. Steen, and J. De Keyser, Use of the Barthel index
and modied Rankin scale in acute stroke trials,Stroke,
vol. 30, no. 8, pp. 15381541, 1999.
[18] P. Lyden, T. Brott, B. Tilley et al., Improved reliability of the
NIH stroke scale using video training. NINDS TPA stroke
study group,Stroke, vol. 25, no. 11, pp. 22202226, 1994.
[19] W. J. Powers, A. A. Rabinstein, T. Ackerson et al., 2018 guide-
lines for the early management of patients with acute ischemic
stroke: a guideline for healthcare professionals from the Amer-
ican Heart Association/American Stroke Association,Stroke,
vol. 49, no. 3, pp. e46e99, 2018.
[20] W. J. Powers, A. A. Rabinstein, T. Ackerson et al., Guidelines
for the early management of patients withacute ischemic stroke:
2019 update tothe 2018 guidelines for the earlymanagement of
acute ischemic stroke: a guideline for healthcare professionals
from the American Heart Association/American Stroke Associ-
ation,Stroke,vol.50,no.12,pp.e344e418, 2019.
[21] R. N. Mitra, S. M. Conley, and M. I. Naash, Therapeutic
Approach of Nanotechnology for Oxidative Stress Induced
Ocular Neurodegenerative Diseases,in Retinal Degenerative
Diseases, pp. 463469, Springer, Cham, 2016.
[22] R. Rodrigo, R. Fernandez-Gajardo, R. Gutierrez et al., Oxida-
tive stress and pathophysiology of ischemic stroke: novel ther-
apeutic opportunities,CNS & Neurological Disorders-Drug
Targets (Formerly Current Drug Targets-CNS & Neurological
Disorders), vol. 12, no. 5, pp. 698714, 2013.
[23] E. Miller, A. Walczak, J. Saluk, M. B. Ponczek, and I. Majsterek,
Oxidative modication of patient's plasma proteins and its
role in pathogenesis of multiple sclerosis,Clinical Biochemis-
try, vol. 45, no. 1-2, pp. 2630, 2012.
[24] K. L. Lambertsen, K. Biber, and B. Finsen, Inammatory cyto-
kines in experimental and human stroke,Journal of Cerebral
Blood Flow & Metabolism, vol. 32, no. 9, pp. 16771698, 2012.
[25] S. Yusuf, S. Hawken, S. Ôunpuu et al., Eect of potentially
modiable risk factors associated with myocardial infarction
in 52 countries (the INTERHEART study): case-control
study,The Lancet, vol. 364, no. 9438, pp. 937952, 2004.
[26] M. I. Ayuso, R. Gonzalo-Gobernado, and J. Montaner, Neu-
roprotective diets for stroke,Neurochemistry International,
vol. 107, pp. 410, 2017.
[27] L. Giusti, C. Angeloni, M. C. Barbalace et al., A proteomic
approach to uncover neuroprotective mechanisms of oleo-
canthal against oxidative stress,International Journal of
Molecular Sciences, vol. 19, no. 8, p. 2329, 2018.
[28] I. Tasset, A. Pontes, A. Hinojosa, R. De La Torre, and I. Túnez,
Olive oil reduces oxidative damage in a 3-nitropropionic
acid-induced Huntington's disease-like rat model,Nutritional
Neuroscience, vol. 14, no. 3, pp. 106111, 2011.
[29] H. Zhou, W.-s. Yang, Y. Li et al., Oleoylethanolamide attenu-
ates apoptosis by inhibiting the TLR4/NF-κB and ERK1/2 sig-
naling pathways in mice with acute ischemic stroke,Naunyn-
Schmiedebergs Archives of Pharmacology, vol. 390, no. 1,
pp. 7784, 2017.
[30] Y. Li, L. Xu, K. Zeng et al., Propane-2-sulfonic acid octadec-9-
enyl-amide, a novel PPARα/γdual agonist, protects against
ischemia-induced brain damage in mice by inhibiting inam-
matory responses,Brain, Behavior, and Immunity, vol. 66,
pp. 289301, 2017.
[31] L. Payahoo, Y. Khajebishak, M. A. Jafarabadi, and
A. Ostadrahimi, Oleoylethanolamide supplementation
reduces inammation and oxidative stress in obese people: a
clinical trial,Advanced Pharmaceutical Bulletin, vol. 8,
no. 3, pp. 479487, 2018.
[32] A. Sayd, M. Antón, F. Alén et al., Systemic administration of
oleoylethanolamide protects from neuroinammation and
anhedonia induced by LPS in rats,International Journal of
Neuropsychopharmacology, vol. 18, no. 6, p. pyu111, 2015.
[33] Y. Zhou, L. Yang, A. Ma et al., Orally administered oleoy-
lethanolamide protects mice from focal cerebral ischemic
injury by activating peroxisome proliferator-activated receptor
α,Neuropharmacology, vol. 63, no. 2, pp. 242249, 2012.
[34] M. A. Yenari, L. Xu, X. N. Tang, Y. Qiao, and R. G. Giard,
Microglia potentiate damage to bloodbrain barrier constitu-
ents: improvement by minocycline in vivo and in vitro,
Stroke, vol. 37, no. 4, pp. 10871093, 2006.
[35] A. Vakili, S. Mojarrad, M. M. Akhavan, and A. Rashidy-Pour,
Pentoxifylline attenuates TNF-αprotein levels and brain
edema following temporary focal cerebral ischemia in rats,
Brain Research, vol. 1377, pp. 119125, 2011.
[36] M. Younus, R. N. Prentice, A. N. Clarkson, B. J. Boyd, and S. B.
Rizwan, Incorporation of an endogenous neuromodulatory
lipid, oleoylethanolamide, into cubosomes: nanostructural
characterization,Langmuir, vol. 32, no. 35, pp. 89428950,
2016.
[37] S. Wu, D. Liao, X. Li et al., Endogenous oleoylethanolamide
crystals loaded lipid nanoparticles with enhanced hydropho-
bic drug loading capacity for ecient stroke therapy,Inter-
national Journal of Nanomedicine, vol. 16, pp. 81038115,
2021.
[38] D. Luo, Y. Zhang, X. Yuan et al., Oleoylethanolamide inhibits
glial activation via moudulating PPAR αand promotes motor
function recovery after brain ischemia,Pharmacological
Research,vol. 141, pp. 530540, 2019.
[39] E. Kossatz, D. Silva-Peña, J. Suárez, F. R. De Fonseca,
R. Maldonado, and P. Robledo, Octadecylpropyl sulfamide
reduces neurodegeneration and restores the memory decits
induced by hypoxia-ischemia in mice,Frontiers in Pharma-
cology, vol. 9, p. 376, 2018.
[40] A. Zorrilla-Vaca, W. Ziai, E. S. Connolly Jr., R. Geocadin,
R. Thompson, and L. Rivera-Lara, Acute kidney injury fol-
lowing acute ischemic stroke and intracerebral hemorrhage: a
meta-analysis of prevalence rate and mortality risk,Cerebro-
vascular Diseases, vol. 45, no. 1-2, pp. 19, 2018.
[41] A.-H. Cho, S. Lee, S. Han, Y.-M. Shon, D.-W. Yang, and
B. Kim, Impaired kidney function and cerebral microbleeds
in patients with acute ischemic stroke,Neurology, vol. 73,
no. 20, pp. 16451648, 2009.
10 Oxidative Medicine and Cellular Longevity
[42] I.-K. Wang, L.-M. Lien, J.-T. Lee et al., Renal dysfunction
increases the risk of recurrent stroke in patients with acute
ischemic stroke,Atherosclerosis, vol. 277, pp. 1520, 2018.
[43] C. Thabuis, F. Destaillats, D. M. Lambert et al., Lipid trans-
port function is the main target of oral oleoylethanolamide to
reduce adiposity in high-fat-fed mice S,Journal of Lipid
Research, vol. 52, no. 7, pp. 13731382, 2011.
[44] J. Fu, F. Oveisi, S. Gaetani, E. Lin, and D. Piomelli, Oleoy-
lethanolamide, an endogenous PPAR-αagonist, lowers body
weight and hyperlipidemia in obese rats,Neuropharmacology,
vol. 48, no. 8, pp. 11471153, 2005.
[45] L. Li, L. Li, L. Chen et al., Eect of oleoylethanolamide on
diet-induced nonalcoholic fatty liver in rats,Journal of Phar-
macological Sciences, vol. 127, no. 3, pp. 244250, 2015.
[46] Q.-W. Deng, S. Li, H. Wang et al., The short-term prognostic
value of the triglyceride-to-high-density lipoprotein choles-
terol ratio in acute ischemic stroke,Aging and Disease,
vol. 9, no. 3, pp. 498506, 2018.
[47] Q. W. Deng, H. Wang, C. Z. Sun et al., Triglyceride to high-
density lipoprotein cholesterol ratio predicts worse outcomes
after acute ischaemic stroke,European Journal of Neurology,
vol. 24, no. 2, pp. 283291, 2017.
[48] L.-c. Yang, H. Guo, H. Zhou et al., Chronic oleoylethanola-
mide treatment improves spatial cognitive decits through
enhancing hippocampal neurogenesis after transient focal
cerebral ischemia,Biochemical Pharmacology, vol. 94, no. 4,
pp. 270281, 2015.
[49] Z. Chen, R. Zhuo, Y. Zhao et al., Oleoylethanolamide stabi-
lizes atherosclerotic plaque through regulating macrophage
polarization via AMPK-PPARαpathway,Biochemical and
Biophysical Research Communications., vol. 524, no. 2,
pp. 308316, 2020.
[50] A. Mowla, B. Shakibajahromi, A. Arora, A. Sei, R. N. Sawyer,
and P. Shirani, Thrombolysis for stroke in elderly in the late
window period,Acta Neurologica Scandinavica, vol. 144,
no. 6, pp. 663668, 2021.
[51] A. Mowla, M. Sharian-Dorche, S. Mehla et al., Safety and
ecacy of antiplatelet use before intravenous thrombolysis
for acute ischemic stroke,Journal of the Neurological Sciences,
vol. 425, p. 117451, 2021.
[52] A. Mowla, A. Memon, S. M. Razavi et al., Safety of intrave-
nous thrombolysis for acute ischemic stroke in patients taking
warfarin with subtherapeutic INR,Journal of Stroke and Cere-
brovascular Diseases, vol. 30, no. 5, p. 105678, 2021.
[53] H. Kamal, B. K. Mehta, M. K. Ahmed et al., Laboratory factors
associated with symptomatic hemorrhagic conversion of acute
stroke after systemic thrombolysis,Journal of the Neurological
Sciences, vol. 420, p. 117265, 2021.
[54] A. Mowla, H. Shah, N. S. Lail, C. B. Vaughn, P. Shirani, and
R. N. Sawyer, Statins use and outcome of acute ischemic
stroke patients after systemic thrombolysis,Cerebrovascular
Diseases (Basel, Switzerland), vol. 49, no. 5, pp. 503508, 2020.
[55] S. Shahjouei, G. Tsivgoulis, N. Goyal et al., Safety of intrave-
nous thrombolysis among patients taking direct oral anticoag-
ulants: a systematic review and meta-analysis,Stroke, vol. 51,
no. 2, pp. 533541, 2020.
[56] A. Mowla, H. Kamal, N. S. Lail et al., Intravenous thromboly-
sis for acute ischemic stroke in patients with thrombocytope-
nia,Journal of Stroke and Cerebrovascular Diseases, vol. 26,
no. 7, pp. 14141418, 2017.
[57] A. Mowla, K. Singh, S. Mehla et al., Is acute reperfusion ther-
apy safe in acute ischemic stroke patients who harbor unrup-
tured intracranial aneurysm,International Journal of Stroke,
vol. 10, no. SA100, pp. 113118, 2015.
[58] H. Kamal, A. Mowla, S. Farooq, and P. Shirani, Recurrent
ischemic stroke can happen in stroke patients very early after
intravenous thrombolysis,Journal of the Neurological Sci-
ences, vol. 358, no. 1-2, pp. 496-497, 2015.
[59] A. Mowla, H. Kamal, S. Mehla, P. Shirani, and R. N. Sawyer,
Rate, clinical features, safety prole and outcome of intrave-
nous thrombolysis for acute ischemic stroke in patients with
negative brain imaging,Journal of Neurology Research,
vol. 10, no. 4, pp. 144-145, 2020.
[60] A. Mowla, H. Shah, N. S. Lail, and P. Shirani, Successful intra-
venous thrombolysis for acute stroke caused by polycythemia
vera,Archives of Neuroscience,vol. 4, no. 4, 2017.
11Oxidative Medicine and Cellular Longevity
... OEA also modulates cognitive deficits induced by MDMA (3,4-methylenedioxymethamphetamine) in mice [120], and induces recovery of cognitive deficits due to a cerebral ischemic insult in rats [121]. In an RCT of patients with acute ischemic stroke, OEA supplementation improved inflammation, oxidative stress, and lipid and biochemical parameters [122]. ...
Article
Full-text available
Background The Mediterranean diet (MedDiet) has demonstrated efficacy in preventing age-related cognitive decline and modulating plasma concentrations of endocannabinoids (eCBs) and N-acylethanolamines (NAEs, or eCB-like compounds), which are lipid mediators involved in multiple neurological disorders and metabolic processes. Hypothesizing that eCBs and NAEs will be biomarkers of a MedDiet intervention and will be related to the cognitive response, we investigated this relationship according to sex and apolipoprotein E (APOE) genotype, which may affect eCBs and cognitive performance. Methods This was a prospective cohort study of 102 participants (53.9% women, 18.8% APOE-ɛ4 carriers, aged 65.6 ± 4.5 years) from the PREDIMED-Plus-Cognition substudy, who were recruited at the Hospital del Mar Research Institute (Barcelona). All of them presented metabolic syndrome plus overweight/obesity (inclusion criteria of the PREDIMED-Plus) and normal cognitive performance at baseline (inclusion criteria of this substudy). A comprehensive battery of neuropsychological tests was administered at baseline and after 1 and 3 years. Plasma concentrations of eCBs and NAEs, including 2-arachidonoylglycerol (2-AG), anandamide (AEA), oleoylethanolamide (OEA), palmitoylethanolamide (PEA), and N-docosahexaenoylethanolamine (DHEA), were also monitored. Baseline cognition, cognitive changes, and the association between eCBs/NAEs and cognition were evaluated according to gender (crude models), sex (adjusted models), and APOE genotype. Results At baseline, men had better executive function and global cognition than women (the effect size of gender differences was − 0.49, p = 0.015; and − 0.42, p = 0.036); however, these differences became nonsignificant in models of sex differences. After 3 years of MedDiet intervention, participants exhibited modest improvements in memory and global cognition. However, greater memory changes were observed in men than in women (Cohen’s d of 0.40 vs. 0.25; p = 0.017). In men and APOE-ε4 carriers, 2-AG concentrations were inversely associated with baseline cognition and cognitive changes, while in women, cognitive changes were positively linked to changes in DHEA and the DHEA/AEA ratio. In men, changes in the OEA/AEA and OEA/PEA ratios were positively associated with cognitive changes. Conclusions The MedDiet improved participants’ cognitive performance but the effect size was small and negatively influenced by female sex. Changes in 2-AG, DHEA, the OEA/AEA, the OEA/PEA and the DHEA/AEA ratios were associated with cognitive changes in a sex- and APOE-dependent fashion. These results support the modulation of the endocannabinoid system as a potential therapeutic approach to prevent cognitive decline in at-risk populations. Trial registration ISRCTN89898870.
... Oleoylethanolamide (OEA) is another compound with multiple antioxidant effects. This fatty acid has shown to reduce lipid peroxidation, prostaglandins, NO formation, and enhancing the GSH levels [101]. ...
Article
Full-text available
Stroke is a major contributor to global mortality and disability. While reperfusion is essential for preventing neuronal death in the penumbra, it also triggers cerebral ischemia-reperfusion injury, a paradoxical injury primarily caused by oxidative stress, inflammation, and blood–brain barrier disruption. An oxidative burst inflicts marked cellular damage, ranging from alterations in mitochondrial function to lipid peroxidation and the activation of intricate signalling pathways that can even lead to cell death. Thus, given the pivotal role of oxidative stress in the mechanisms of cerebral ischemia-reperfusion injury, the reinforcement of the antioxidant defence system has been proposed as a protective approach. Although this strategy has proven to be successful in experimental models, its translation into clinical practice has yielded inconsistent results. However, it should be considered that the availability of numerous antioxidant molecules with a wide range of chemical properties can affect the extent of injury; several groups of antioxidant molecules, including polyphenols, carotenoids, and vitamins, among other antioxidant compounds, can mitigate this damage by intervening in multiple signalling pathways at various stages. Multiple clinical trials have previously been conducted to evaluate these properties using melatonin, acetyl-L-carnitine, chrysanthemum extract, edaravone dexborneol, saffron, coenzyme Q10, and oleoylethanolamide, among other treatments. Therefore, multi-antioxidant therapy emerges as a promising novel therapeutic option due to the potential synergistic effect provided by the simultaneous roles of the individual compounds.
... Moreover, a significant reduction in the expression levels of nuclear factor-kappa B (NF-kB) and IL-6 was observed in obese patients with NAFLD after OEA supplementation (Tutunchi et al., 2021). On the other hand, in the recent investigation by Sabahi et al. (2022), OEA treatment (300 mg/ day) could improve the short-term inflammatory and oxidative stress status in patients with acute ischemic stroke. A significant reduction in serum levels of IL-1β, monocyte chemoattractant protein-1 (MCP-1), and TNF-α were also found by OEA supplementation (5 mg/kg) in alcohol intoxicated rats (Antón et al., 2017). ...
Article
Full-text available
Background: Oxidative stress is considered a major factor in the pathophysiology of non-alcoholic liver disease (NAFLD). A growing body of evidence indicates that oleoylethanolamide (OEA), a bioactive lipid mediator, has anti-inflammatory and antioxidant properties. This trial investigated the effects of OEA administration on inflammatory markers, oxidative stress and antioxidant parameters of patients with NAFLD. Methods: The present randomized controlled trial was conducted on 60 obese patients with NAFLD. The patients were treated with OEA (250 mg/day) or placebo along with a low-calorie diet for 12 weeks. Inflammatory markers and oxidative stress and antioxidant parameters were evaluated pre-and post-intervention. Results: At the end of the study, neither the between-group changes, nor the within-group differences were significant for serum levels of high-sensitivity C-reactive protein (hs-CRP), interleukin-1 beta (IL-1β), IL-6, IL-10, and tumor necrosis-factor α (TNF-α). Serum levels of total antioxidant capacity (TAC) and superoxide dismutase (SOD) significantly increased and serum concentrations of malondialdehyde (MDA) and oxidized-low density lipoprotein (ox-LDL) significantly decreased in the OEA group compared to placebo at study endpoint (p = 0.039, 0.018, 0.003 and 0.001, respectively). Although, no significant between-group alterations were found in glutathione peroxidase and catalase. There were significant correlations between percent of changes in serum oxidative stress and antioxidant parameters with percent of changes in some anthropometric indices in the intervention group. Conclusion: OEA supplementation could improve some oxidative stress/antioxidant biomarkers without any significant effect on inflammation in NAFLD patients. Further clinical trials with longer follow-up periods are demanded to verify profitable effects of OEA in these patients. Clinical Trial Registration: www.irct.ir, Iranian Registry of Clinical Trials IRCT20090609002017N32.
... Similar to that described above for the liver, OEA anti-inflammatory events could be associated with the inhibition of NFκB PPARα mediation [95] (Figure 1). OEA plays analogous neuroprotective roles in numerous models of neurological disorders and brain injuries [101]. ...
Article
Full-text available
In 2010, the Mediterranean diet was recognized by UNESCO as an Intangible Cultural Heritage of Humanity. Olive oil is the most characteristic food of this diet due to its high nutraceutical value. The positive effects of olive oil have often been attributed to its minor components ; however, its oleic acid (OA) content (70-80%) is responsible for its many health properties. OA is an effective biomolecule, although the mechanism by which OA mediates beneficial physiological effects is not fully understood. OA influences cell membrane fluidity, receptors, in-tracellular signaling pathways, and gene expression. OA may directly regulate both the synthesis and activities of antioxidant enzymes. The anti-inflammatory effect may be related to the inhibition of proinflammatory cytokines and the activation of anti-inflammatory ones. The best-characterized mechanism highlights OA as a natural activator of sirtuin 1 (SIRT1). Oleoylethanolamide (OEA), derived from OA, is an endogenous ligand of the peroxisome proliferator-activated receptor alpha (PPARα) nuclear receptor. OEA regulates dietary fat intake and energy homeostasis and has therefore been suggested to be a potential therapeutic agent for the treatment of obesity. OEA has anti-inflammatory and antioxidant effects. The beneficial effects of olive oil may be related to the actions of OEA. New evidence suggests that oleic acid may influence epigenetic mechanisms, opening a new avenue in the exploration of therapies based on these mechanisms. OA can exert beneficial anti-inflammatory effects by regulating microRNA expression. In this review, we examine the cellular reactions and intracellular processes triggered by OA in T cells, macrophages, and neutrophils in order to better understand the immune modulation exerted by OA.
... 57 Similarly, oleoylethanolamide shows reduced inflammation, oxidative stress, and dyslipidemia after ischemic stroke, but no functional recovery is reported. 58 Ongoing clinical trials are measuring IL-6 in relation to ischemic stroke prognosis (NCT05004389; NCT03297827), ischemic stroke treatment efficacy (NCT04705779), hemorrhagic stroke recovery using in-bed cycle ergometry (NCT04027049), and even IS and HS recovery following autologous mesenchymal stem cell therapy (NCT04063215). ...
Article
Full-text available
Interleukin (IL)‐6 is a unique cytokine due to its dual signaling, with one pathway being pro‐inflammatory (trans) and the other homeostatic (classical). Both of these pathways have been implicated in neuroinflammation following stroke, with initial inflammatory mechanisms being protective and later anti‐inflammatory signaling promoting ischemic tissue recovery. IL‐6 plays a major role in stroke pathology. However, given these distinctive IL‐6 signaling consequences, IL‐6 is a difficult cytokine to target for stroke therapies. Recent research suggests that the ratio between the pro‐inflammatory binary IL6:sIL6R complex and the inactive ternary IL6:sIL6R:sgp130 complex may be a novel way to measure IL‐6 signaling at different time points following ischemic injury. This ratio may approximate functional consequences on individualized stroke therapies, allowing clinicians to determine whether IL‐6 agonists or antagonists should be used at specific time points.
Article
Ischemic stroke is the most common, fatal and disabling disease of the central nervous system. Despite considerable progress in primary prevention, diagnostic testing, and treatment, the increased incidence of stroke has created a serious public health problem worldwide, with a significant impact on the families and societies of those affected. The study of ketogenic (KD) and calorie-restricted (CR) diets has become a hot topic of research and is gradually penetrating into the daily diet. Although there are many studies on the neuroprotective mechanisms of KD and CR, the duration of the interventions is relatively short and the mechanisms are not well understood. Therefore, whether long-term ketogenic diet and calorie-restricted diet treatment can improve the resistance to ischemic-hypoxic injury in healthy aged C57 mice, and the mechanism still deserves further investigation. Young 6-month-old mice were randomly divided into KD, CR and control groups, and an ischemic stroke model of endothelin-1 was established on the basis of dietary intervention for 15 months, and the mechanism was explored based on bioinformatics method. Our results confirm that long-term dietary treatment of KD and CR reduces neuronal death in infarct foci and improves neuronal resistance to ischemia and hypoxia by upregulating UBR4 to inhibit the CamkⅡ/TAK1/JNK signaling pathway, then suppress oxidative stress, thus acting as a neuroprotective agent. To be conclusive, ketogenic and calorie-restricted diets have a significant effect on ischemic stroke, but well-designed future clinical studies are necessary to provide solid evidence on this topic.
Article
Full-text available
Background The history of intracranial hemorrhage (ICrH) is considered a contraindication for intravenous thrombolysis (IVT) among patients with acute ischemic stroke (AIS). Objective: This study aimed at comparing the safety of IVT among patients with and without a history of ICrH. Methods We performed a systematic review of the literature. Data regarding all AIS patients with prior ICrH who received IVT were retrieved. Meta-analysis was performed to compare the rate of symptomatic hemorrhagic transformation (sHT), death within 90 days, and favorable and unfavorable 90-day functional outcomes based on modified Rankin Scale (mRS) among stroke patients with and without prior ICrH. Results Out of 13,032 reviewed records, 7 studies were included in the systematic review and meta-analysis. Quantitative synthesis of data regarding the rate of sHT (5068 patients) revealed no significant difference between the two groups [odds ratio, OR: 1.55 (0.77, 3.12); p = 0.22]. However, a significantly higher risk of death within 90 days [OR: 3.91 (2.16, 7.08); p < 0.00001] and a significantly higher 90-day poor functional outcomes (mRS, 4–6) [OR: 1.57 (1.07, 2.30); p = 0.02] were observed among patients with prior ICrH. Likewise, the percentage of 90-day good functional outcomes (mRS, 0–1) was lower in the prior ICrH group [OR: 0.54 (0.35, 0.84); p = 0.06]. Subgroup analyses in patients with a history of ICrH (based on both patients’ medical history and imaging confirmation) revealed no significant between-group differences in rates of sHT. Also, sensitivity analysis consisting of only studies using standard-dose IVT showed no difference in sHT rates and 90-day outcomes between the two groups. There was no evidence of heterogeneity ( I ² >50%) among included studies. Conclusion The results of this study indicated that prior history of ICrH does not increase the risk of sHT post-IVT, but it is associated with a higher risk of death and poor functional outcomes in 90 days.
Article
Full-text available
Introduction Although the preparation of lipid nanoparticles (LNPs) achieves great success, their retention of highly hydrophobic drugs is still problematic. Methods Herein, we report a novel strategy for efficiently loading hydrophobic drugs to LNPs for stroke therapy. Oleoylethanolamide (OEA), an endogenous highly hydrophobic molecule with outstanding neuroprotective effect, was successfully loaded to OEA-SPC&DSPE-PEG lipid nanoparticles (OSDP LNPs) with a drug loading of 15.9 ± 1.2 wt%. Efficient retention in OSDP LNPs greatly improved the pharmaceutical property and enhanced the neuroprotective effect of OEA. Results Through the data of positron emission tomography (PET) and TTC-stained brain slices, it could be clearly visualized that the acute ischemic brain tissues were preserved as penumbral tissues and bounced back with reperfusion. The in vivo experiments stated that OSDP LNPs could significantly improve the survival rate, the behavioral score, the cerebral infarct volume, the edema degree, the spatial learning and memory ability of the MCAO (middle cerebral artery occlusion) rats. Discussion These results suggest that the OSDP LNPs have a great chance to develop hydrophobic OEA into a potential anti-stroke formulation.
Article
Objectives Safety of intravenous thrombolysis (IVT) within 3–4.5 hours of stroke onset in patients ≥80 years is still disputable. We evaluated the association of symptom onset-to-treatment time (SOTT) with the symptomatic intracranial hemorrhage (sICH), poor outcome, and mortality in patients≥80 years. Materials and Methods In a retrospective study, patients treated with IVT following stroke were registered. Outcomes were poor outcome (mRS>2), sICH/ECASS-2, and in-hospital mortality. We compared the patients≥80 years who received IVT within 3 hours with those receiving IVT within 3–4.5 hours. We further compared the patients who were <80 years with those ≥80 years and SOTT of 3–4.5 hours. Results Of 834 patients, 265 aged over 80. In those above 80 and in multivariable analysis, the associations of SOTT with poor outcome (aOR: 1.401, CI: 0.503–3.903, p=0.519), sICH (aOR=2.50, CI=0.76–8.26, p= 0.132) and mortality (aOR=1.12, CI=0.39–3.25, p= 0.833) were not significant. 106 patients received IVT within 3–4.5 hours. In multivariable analysis, the associations of age (≥80 versus <80) with poor outcome (aOR=1.87, CI=0.65–5.37, p=0.246), sICH (aOR=0.65, CI=0.14–3.11, p=0.590), and mortality (aOR=0.87, 95% CI=0.16–4.57, p=0.867) were not significant in patients with SOTT of 3–4.5 hours. Conclusion IVT within 3–4.5 hours in patients ≥80 years is not associated with increased sICH, poor outcome, and mortality compared to the early time window, and also compared to the younger patients in 3–4.5 hours window period. The decision of IVT administration in this age group should not be made solely on the basis of stroke onset timing.
Article
Parkinson's disease (PD), the most common movement disorder, comprises several pathophysiologic mechanisms including misfolded alpha‐synuclein aggregation, inflammation, mitochondrial dysfunction and synaptic loss. Nuclear Factor‐Kappa B (NF‐κB), as a key regulator of a myriad of cellular reactions, is shown to be involved in such mechanisms associated with PD, and the changes in NF‐κB expression is implicated in PD. Alpha‐synuclein accumulation, the characteristic feature of PD pathology, is known to trigger NF‐κB activation in neurons, thereby propagating apoptosis through several mechanisms. Furthermore, misfolded alpha‐synuclein released from degenerated neurons, activates several signaling pathways in glial cells which culminate in activation of NF‐κB and production of pro‐inflammatory cytokines, thereby aggravating neurodegenerative processes. On the other hand, NF‐κB activation, acting as a double‐edged sword, can be necessary for survival of neurons. For instance, NF‐κB activation is necessary for competent mitochondrial function and deficiency in c‐Rel, one of the NF‐κB proteins, is known to propagate DA neuron loss via several mechanisms. Despite the dual role of NF‐κB in PD, several agents by selectively modifying the mechanisms and pathways associated with NF‐κB, can be effective in attenuating DA neuron loss and PD, as reviewed in this paper.
Article
Aim To study the effects of pretreatment with Antiplatelet (AP) before IV thrombolysis (IVT) on the rate of symptomatic intracranial hemorrhage (sICH) and functional outcome in patients with Acute Ischemic stroke (AIS). Method In this retrospective study, the medical records and cerebrovascular images of all the patients who received IVT for AIS in our center in a 9.6-year period were reviewed. Patients who took at least one dose of any APs in the last 24 h prior to IVT were identified. They were categorized according to the type of AP, single versus dual AP therapy (DAPT), and dose of AP. Rate of sICH and functional outcome at discharge were compared between the AP users and non-users. Results A total of 834 patients received IVT for AIS in our center during a 9.6- year period. Multivariate models were adjusted for age, NIHSS on admission, history of atrial fibrillation, history of hypertension, INR on admission, history of stroke and diabetes mellitus. In multivariate regression analyses and after adjusting for the variables mentioned above, the use of any AP was not associated with an increased rate of sICH (OR = 1.28 [0.70–2.34], p = 0.425). Furthermore, the use of DAPT did not significantly increase the rate of sICH in multivariate regression analyses. (OR = 0.663 [0.15–2.84], p = 0.580). The patients on any AP had a lower chance of having good functional outcome in univariate analysis (OR = 0.735 [0.552–0.979], p = 0.035). However, when adjusted for age, baseline NIHSS, history of diabetes, hypertension and prior stroke, AP use was not associated with a decreased chance of having a good functional outcome at discharge. (OR = 0.967 [0.690–1.357], p = 0.848). In addition, no significant difference was noted in the rate of good functional outcome between patients on DAPT and no AP users in multivariate regression analyses. (OR = 1.174 [0.612–2.253], p = 0.629). Conclusion Our study did not show any significant association between the risk of sICH and good functional outcome after IVT for AIS patients on AP therapy (dual or single) in comparison with AP naïve patients.
Article
Introduction Current guidelines allow the administration of intravenous recombinant tissue plasminogen activator (IV r-tPA) to warfarin-treated patients with acute ischemic stroke (AIS) who have an international normalized ratio (INR) of ≤1.7. However, concerns remain about the safety of using IV r-tPA in this situation due to a conceivable risk of symptomatic intracranial hemorrhage (sICH), lack of dedicated randomized controlled trials and the conflicts in the available data. We aimed to determine the risk of sICH in warfarin-treated patients with subtherapeutic INR who received IV r-tPA for AIS in our large volume comprehensive center. Methods Patients who had received IV r-tPA for AIS in a 9.6-year period were retrospectively investigated (n = 834). Patients taking warfarin prior to presentation were identified (n = 55). One patient was excluded due to elevated INR beyond the acceptable range for IV r-tPA treatment. Because of the significant difference in the sample size (54 vs 779), warfarin group was matched with 54 non-warfarin patients adjusted for independent risk factors for sICH (age, admission NIHSS, history of diabetes). Good outcome was defined as mRS of 0-2 on discharge and sICH was defined as an ICH causing increase in NIHSS ≥4 or death. Warfarin-treated group was further dichotomized based on INR (1-1.3 vs 1.3-1.7) and safety and outcome measures were compared between resultant groups. Results No significant difference was found between warfarin-treated and the non-warfarin groups in terms of chance of good outcome on discharge (27.8% in warfarin group vs 26.4% in non-warfarin group; p-value >0.05), or the rate of occurrence of sICH (3.7% in warfarin group vs 11.1% in non-warfarin group; p-value >0.05). Furthermore, rate of sICH (5.1% in patients with INR <1.3 versus 0.0% in patients with INR 1.3-1.7; p-value >0.05) or chance of good outcome on discharge (28.2% of patients with INR <1.3 versus 26.7% in patients with INR 1.3-1.7; p-value >0.05) were not found to be different after the warfarin-treated group was dichotomized. Conclusion Administration of IV r-tPA for AIS in warfarin-treated patients with subtherapeutic INR <1.7 does not increase the risk of sICH.
Article
Background Laboratory factors associated with hemorrhagic conversion (HC) after Intravenous thrombolysis with rtPA (IVT) for Acute Ischemic Stroke (AIS) remain nebulous despite advances in our knowledge of AIS. This study aimed to investigate the laboratory factors predisposing to HC in AIS patients receiving IVT. Methods We retrospectively reviewed the medical records of patients who received IV tPA for AIS at our comprehensive stroke center over a 9.6-year period. Besides age, gender, NIHSS, history of diabetes mellitus (DM), history of atrial fibrillation (Afib), we gathered their laboratory data including International Normalized Ratio (INR), lipid panel, serum albumin, serum creatinine, hemoglobin A1c (HbA1c), and admission blood glucose. Post-thrombolysis brain imagings were reviewed to evaluate for symptomatic ICH (sICH). The mean values of above mentioned laboratory data were compared between the group with sICH and patients with no sICH. Univariate and multivariate logistic regression were performed to evaluate the association of the laboratory findings with presence of sICH. sICH was defined as ICH causing an increase in NIHSS ≥4. Results Of the 794 subjects in this study 51 (6.4%) had sICH. In the univariate analysis, patients who developed sICH had significantly higher NIHSS on admission (14.2 ± 5.4 vs 11.2 ± 6.5, p < .001), LDL-cholesterol (113.3 mg/dl ±36.9 vs. 101.8 mg/dl ± 38.2, p = .032), HbA1c (6.9% ± 2.3 vs. 6.1 ± 1.3, p = .003) and lower levels of Albumin (3.5 g/dl ±0.4 vs. 3.9 g/dl ± 0.5, p < .001). Furthermore, a higher prevalence of history of DM (45% vs. 21.6%, p = .020) and Afib (25.4% vs. 10.4%, p = .028) was found in subjects who developed sICH. There were no significant group differences regarding age, sex, total cholesterol, blood glucose on admission, serum creatinine or INR levels (p > .05). After adjusting for multiple covariates, lower Albumin level and and higher HbA1c were significantly associated with an increased risk for sICH development (p < .05). Chances of sICH increased by 33% for every 1 g/dl below a normal albumin level of 4.0 g/dl (p < .05). Conclusion Lower endogenous albumin level and higher HbA1c have shown to predispose to a higher risk of sICH after IVT for AIS and might be good predictors of sICH post IVT.
Article
Aim: The aim of this was to study the effects of statins and their intensity on symptomatic intracranial hemorrhage (sICH) and outcome after IV thrombolysis (IVT) for acute ischemic stroke (AIS). Methods: We retrospectively reviewed the medical records and cerebrovascular images of all the patients treated with IVT for AIS in our center in a 10-year period. Patients were further characterized as any statin users versus non-users on admission to the emergency department. Statins were categorized in high intensity or low intensity statin based on its propensity to reduce lower low-density cholesterol by ≥45% or <45%, respectively. Safety and discharge modified Rankin Score were compared between statin users versus non-users and also between high-intensity versus low-intensity groups. Results: A total of 834 patients received IVT for AIS in our center during a 10-year period. Multivariate models were adjusted for age, NIH Stroke Scale at admission, INR, and history of DM and atrial fibrillation. There was no association between odds of sICH and any statin use (OR = 0.52 [0.26-1.03], p = 0.06). In multivariate model, any statin use was not associated with odds of poor outcome (Table 4: OR = 1.01 [0.79-1.55], p = 0.57). There was no significant association between odds of sICH among patients on high-intensity statin compared to low intensity statin (multivariate model OR = 0.39 [0.11-1.40], p = 0.15). There was 47% reduced odds of poor outcome among patients on high-intensity statin as compared to low-intensity statin (OR = 0.53[0.32-0.88] p = 0.01). However, this significant association was lost in the multivariate model (OR = 0.60 [0.35-1.05], p = 0.07). Conclusion: Our study does not show any significant association between risk of sICH and poor outcome after IVT for patients on prior statin therapy. We also did not find significant association between the risk of sICH and poor outcome after IVT and the intensity of the stain used.
Article
Background: Atherosclerotic plaque rupture is the major trigger of acute cardiovascular risk events, and manipulation of M1/M2 macrophage homeostasis is an effective strategy for regulating atherosclerotic plaque stability. This study was aimed to illuminate the effects of oleoylethanolamide (OEA) on macrophage polarization and plaque stability. Methods: Macrophages derived from THP-1 were treated with OEA followed by LPS/IFN-γ, and the markers of M1, M2 macrophages were monitored by western blot, real-time PCR and immunofluorescence staining. The effect of OEA on macrophage polarization in the arch of aortic arteries was tested by immunofluorescence staining and western blot, and the plaque stability was completed by Masson's trichrome and hematoxylin and eosin (HE) in apolipoprotein E (ApoE)-/- mice. Results: OEA treatment enhanced the expression of two classic M2 macrophage markers, macrophage mannose receptor (CD206) and transforming growth factor (TGF-β), while the expression of iNOS (M1 macrophages) was decreased in THP-1-derived macrophages. Blocking of PPARα using siRNA and inhibition of AMP-activated protein kinase (AMPK) by its inhibitor compound C attenuated the OEA-induced expression of M2 macrophage markers. In addition, OEA significantly suppressed M1, promoted M2 macrophage polarization, increased collagen content and decreased necrotic core size in atherosclerotic plaques in ApoE-/- mice, which were linked with the expression of PPARα. Conclusions: OEA improved atherosclerotic plaque stability through regulating macrophage polarization via AMPK-PPARα pathway.