ArticlePDF Available

The Prognostic Value of Serum Cytokines in Patients with Acute Ischemic Stroke

Authors:

Abstract and Figures

The inflammatory response is an unavoidable process and contributes to the destruction of cerebral tissue during the acute ischemic stroke (AIS) phase and has not been addressed fully to date. Insightful understanding of correlation of inflammatory mediators and stroke outcome may provide new biomarkers or therapeutic approaches for ischemic stroke. Here, we prospectively recruited 180 first-ever AIS patients within 72 hrs after stroke onset. We used the National Institutes of Health Stroke Scale (NIHSS) to quantify stroke severity and modified Rankin scale (mRS) to assess the 3-month outcome for AIS patients. Initially, we screened 35 cytokines, chemokines, and growth factors in sera from 75 AIS patients and control subjects. Cytokines that were of interest were further investigated in the 180 AIS patients and 14 heathy controls. We found that IL-1RA, IL-1β, IL-4, IL-5, IL-6, IL-7, IL-9, IL-10, IL-13, IL-15, EGF, G-CSF, Flt-3L, GM-CSF and Fractalkine levels were significantly decreased in severe stroke patients. In particular, IL-1β, IL-4, IL-5, IL-7, IL-9, IL-10, IL-15, G-CSF and GM-CSF were significantly reduced in AIS patients with poor outcome, compared to those with good prognosis. IL-6 was notably higher in the poor outcome group. Only IL-9 level decreased in the large infarct volume group. After adjusting for confounders, we found that IL-5 was an independent protective factor for prognosis in AIS patients with an adjusted OR of 0.042 (P = 0.007), whereas IL-6 was an independent risk predictor for AIS patients with an adjusted OR of 1.293 (P = 0.003). Our study suggests the levels of serum cytokines are related to stroke severity, short-term prognosis and cerebral infarct volume in AIS patients.
Content may be subject to copyright.
http://dx.doi.org/10.14336/AD.2018.0820
*Correspondence should be addressed to: Dr. Kunlin Jin (kunlin.jin@unthsc.edu) and Bei Shao (shaobei56@126.com), Department of
Neurology, The First Affiliated Hospital of Wenzhou Medical University, China. #These authors contributed equally to this study.
Copyright: © 2018 Lin S et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
ISSN: 2152-5250 1
Original Article
The Prognostic Value of Serum Cytokines in Patients with
Acute Ischemic Stroke
Xianmei Li1,#, Siyang Lin1,#, Xiaoli Chen2, Wensi Huang3, Qian Li4, Hongxia Zhang5, Xudong
Chen1, Shaohua Yang5, Kunlin Jin5, Bei Shao1,*
1Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
2Department of Rehabilitation, Wenzhou People's Hospital, Wenzhou, China
3Department of Neurology, The People's Hospital of Pingyang, Wenzhou, China
4Department of Neurology, Jinhua Municipal Central Hospital, Wenzhou, China
5Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth,
Texas, USA
[Received March 1, 2018; Revised August 18, 2018; Accepted August 20, 2018]
ABSTRACT: The inflammatory response is an unavoidable process and contributes to the destruction of cerebral
tissue during the acute ischemic stroke (AIS) phase and has not been addressed fully to date. Insightful
understanding of correlation of inflammatory mediators and stroke outcome may provide new biomarkers or
therapeutic approaches for ischemic stroke. Here, we prospectively recruited 180 first-ever AIS patients within
72 hrs after stroke onset. We used the National Institutes of Health Stroke Scale (NIHSS) to quantify stroke
severity and modified Rankin scale (mRS) to assess the 3-month outcome for AIS patients. Initially, we screened
35 cytokines, chemokines, and growth factors in sera from 75 AIS patients and control subjects. Cytokines that
were of interest were further investigated in the 180 AIS patients and 14 heathy controls. We found that IL-1RA,
IL-1β, IL-4, IL-5, IL-6, IL-7, IL-9, IL-10, IL-13, IL-15, EGF, G-CSF, Flt-3L, GM-CSF and Fractalkine levels
were significantly decreased in severe stroke patients. In particular, IL-1β, IL-4, IL-5, IL-7, IL-9, IL-10, IL-15,
G-CSF and GM-CSF were significantly reduced in AIS patients with poor outcome, compared to those with good
prognosis. IL-6 was notably higher in the poor outcome group. Only IL-9 level decreased in the large infarct
volume group. After adjusting for confounders, we found that IL-5 was an independent protective factor for
prognosis in AIS patients with an adjusted OR of 0.042 (P = 0.007), whereas IL-6 was an independent risk
predictor for AIS patients with an adjusted OR of 1.293 (P = 0.003). Our study suggests the levels of serum
cytokines are related to stroke severity, short-term prognosis and cerebral infarct volume in AIS patients.
Key words: acute ischemic stroke, cytokines, inflammation, prognosis, stroke severity
Cerebrovascular disease, a global health problem, has
become one of the major cause of adult mortality and
disability worldwide, and ranks second only to ischemic
heart disease [1-3]. Acute ischemic stroke (AIS), caused
by cerebral embolism or arterial thrombosis, accounts for
80-85% of cerebrovascular disease and is the major
subtype of all strokes [4]. Inflammation following AIS is
considered an inevitable pathological process involved in
post-ischemic injury in the brain [5, 6]. After the initial
injury, a series of detrimental secondary events occur and
the blood-brain barrier (BBB) becomes damaged.
Activated peripheral immune cells including neutrophils
and T-cells can then cross the BBB and accumulate at the
site of injury [5]. These cascade reactions would indeed
Volume 10, Number 2; xxx-xx, April 2019
Lin S., et al Serum cytokines and ischemic outcome
Aging and Disease Volume 10, Number 2, April 2019 2
aggravate cerebral infarction, which potentially
contributes to the destruction of cerebral tissue during the
AIS phase [7, 8]. On the other hand, an increasing number
of reports have showed that the inflammatory response
after stroke plays a critical role for functional recovery in
the later stages [9, 10]. A probable reason for this
phenomenon can be attributed to dynamic alterations in
the release of several pro- and anti-inflammatory
cytokines in the brain that could affect the progression of
cerebral infarction [11]. These chemokines are mainly
produced by resident microglial cells and infiltrating
immune cells, which could attract and activate leukocytes
[12, 13]. Some chemokines are even capable of recruiting
nonimmune cells like neural stem cells (NSCs), neural
progenitor cells (NPCs), endothelial cells and bone
marrow stromal cells (BMSCs) to the lesion site which
may exert beneficial impact on protection and recovery
[14]. Th2 type cytokines including interleukin-4 (IL-4)
and interleukin-5 (IL-5) were found to play beneficial
roles in the repair of brain damage, suppress post-stroke
inflammation, and have the capability to induce
neurotrophic factors in astrocytes [15-17]. Interleukin-6
(IL-6), well-known for its pro-inflammatory function also
possess neurotrophic and regenerative capabilities after
cerebral ischemia [18, 19]. Interleukin-7 (IL-7) is a
pleiotropic cytokine with multiple effects. Arya et al.
reported that IL-7 could enhance the expression of
monocyte chemoattractant protein-1 (MCP-1) in patients
with unstable angina, and was associated with
hyperlipidemia and atherosclerosis [20]. Interleukin-9
(IL-9) is a pro-inflammatory cytokine, secreted by Th9
cells. Interleukin-10 (IL-10) is generally viewed as an
anti-inflammatory cytokine that helps to restrain pro-
inflammatory cytokines and depress cytokine receptor
expression and receptor activation [21]. Interleukin-13
(IL-13) is a mediator of allergic inflammation and
different diseases including asthma [22]. Interleukin-15
(IL-15) mainly regulates the activation and proliferation
of T and natural killer (NK) cells. Fractalkine (CX3CL1),
macrophage-derived chemokine (MDC, CCL22) and
macrophage inflammatory protein-1 alpha (MIP-1α,
CCL3) are members of chemokine family, which serve
different functions in inflammation, such as guiding the
migration of immune cells or regulating activation and
maturation of cells. Fms-like tyrosine kinase-3 ligand
(Flt-3L), epidermal growth factor (EGF), granulocyte-
colony stimulating factor (G-CSF), granulocyte-
macrophage colony-stimulating factor (GM-CSF) are
growth factors. Therefore, these inflammatory mediators,
cytokines in particular, have been considered as
biomarkers for stroke pathogenesis and prognosis.
Previous reports characterized an acute immune response
to ischemic stroke by profiling certain cytokines and
chemokines (e.g., IL-1α and β, IL-6, IL-8, IL-9, IL-10, IL-
12, IL-18, TNFα and soluble TNF-receptors p55, p75 and
GRO-α) in the sera or cerebrospinal fluid of stroke
patients [23-25]. However, various known cytokines have
been studied in limited number thereby restricting the
significance of predictive value of these biomarkers in
patient outcome after AIS.
To determine the significance of serum
inflammatory cytokines in patient outcome after acute
ischemic stroke, we simultaneously measured 35
cytokines, chemokines, and growth factors in a single
patient group using a multiplex immunoassay and found
that the levels of inflammatory mediators in the serum can
be used to deduce stroke severity, short-term prognosis
and cerebral infarct volume.
MATERIALS AND METHODS
Study population
One hundred and eighty patients with first-ever AIS were
enrolled at the Department of Neurology, the First
Affiliated Hospital of Wenzhou Medical University, from
April 2014 to September 2016. All AIS patients within 72
hrs after stroke onset were selected based on the criteria
set out by the World Health Organization [26]. Etiology
subtypes of ischemic stroke were classified according to
the criteria of Trial of Org 10172 in Acute Stroke
Treatment (TOAST) [27]. The exclusion criteria for first-
ever AIS patients are: (i) history of any serious central
nervous system disease, such as Parkinson’s disease,
craniocerebral trauma, dementia, hematencephalon,
cerebral infarction or subarachnoid hemorrhage; (ii)
failure of important organs, such as heart failure, severe
liver or renal insufficiency; (iii) autoimmune disease
(AID) or the use of steroids or immunosuppressants; (iv)
a history of cancer; (v) a serious history of infection,
trauma or surgery within 4 weeks prior to onset.
The study followed ethical guidelines and obtained
the approval of the ethics committee at the First Affiliated
Hospital of Wenzhou Medical University. Patient consent
forms were signed by each patient or their relatives before
inclusion.
Data collection
The following basic clinical information of all subjects
during the first 24 hrs after onset of stroke were collected
by well-trained neurologists: gender, age, smoking habit,
alcohol abuse, hypertension, diabetes mellitus (DM),
hyperlipemia, cardiovascular diseases, systolic blood
pressure (SBP) and diastolic blood pressure (DBP) on
admission and biochemical indices. The National
Institutes of Health Stroke Scale (NIHSS) score was used
Lin S., et al Serum cytokines and ischemic outcome
Aging and Disease Volume 10, Number 2, April 2019 3
to measure the severity of AIS patients by the neurologist
within 24 hrs of admission and discharge [28, 29]. The
minor stroke was defined as NIHSS score < 5 according
to previous studies [30, 31]. The short-term functional
outcome was assessed by telephone interviews or
outpatient service at 3 months after stroke onset using the
modified Rankin scale (mRS, scores range from 0 to 6)
[32]. A poor functional outcome was defined as mRS
score 3-6, while favorable outcome was defined as mRS
score 0-2 [33, 34].
Table 1. Baseline characteristics of AIS patients with favorable or poor outcomes.
Characteristics
Total (N= 167)
Prognosis at 3 months follow-up
P value
Favorable Outcome
(N= 113)
Poor Outcome
(N=54)
Age (years)
63.02 ± 9.84
62.08 ± 10.35
65.00 ± 8.43
0.073
Males (%)
100 (60.0)
73 (64.6)
27 (50.0)
0.073
SBP (mmHg)
162.34 ± 24.41
160.18 ± 22.76
166.87 ± 27.20
0.097
DBP (mmHg)
85.43 ± 13.68
85.48 ± 13.13
85.31 ± 14.90
0.943
Hypertension (%)
141 (84.4)
93 (82.3)
48 (88.9)
0.273
Hyperlipidemia (%)
32 (19.2)
21 (18.6)
11 (20.4)
0.784
Diabetes (%)
55 (32.9)
33 (29.2)
22 (40.7)
0.139
Cardiac disease (%)
19 (11.4)
10 (8.8)
9 (16.7)
0.138
Smoking (%)
57 (34.1)
31 (27.4)
24 (44.4)
0.053
Alcohol drinking (%)
43 (25.7)
33 (29.2)
9 (16.7)
0.064
Stroke etiologic subtypes (%)
0.982
Large-artery atherosclerosis
110 (65.7)
76 (67.3)
34 (62.3)
-
Cardioembolic
13 (7.8)
8 (7.1)
5 (9.3)
-
Small-vessel disease
38 (22.8)
28 (24.8)
10 (18.5)
-
Other or unknown cause
6 (3.6)
2 (1.8)
4 (7.4)
-
BMI (kg/m2)
23.76 ± 3.14
23.72 ± 3.11
23.84 ± 3.23
0.808
Laboratory tests
WBC (109/L)
6.35 (5.57 7.79)
6.29 (5.44 7.58)
6.51 (5.82 8.60)
0.081
Neutrophils (109/L)
3.88 (3.03 4.92)
3.70 (2.89 4.82)
4.22 (3.38 5.74)
0.009
Hs-CRP (mmol/L)
1.86 (0.87 3.91)
1.67 (0.60 3.30)
2.64 (1.37 4.90)
0.027
IL-1RA (pg/mL)
2.21 (0.68 8.66)
2.30 (0.97 7.710)
1.4 (0.27 5.56)
0.165
IL- (pg/mL)
2.96 (0.32 17.69)
2.96 (0.32 17.69)
1.21 (0.08 27.21)
0.365
IL- (pg/mL)
1.06 (0.73 1.60)
1.11 (0.82 1.60)
0.87 (0.60 1.56)
0.007
IL-4 (pg/mL)
1.82 (0.54 6.47)
3.10 (1.13 8.15)
0.53 (0.28 1.32)
< 0.001
IL-5 (pg/mL)
0.58 (0.35 0.82)
0.63 (0.48 0.86)
0.32 (0.24 0.66)
< 0.001
IL-6 (pg/mL)
1.17 (0.65 1.95)
1.17 (0.68 1.90)
2.06 (0.49 2.13)
0.021
IL-7 (pg/mL)
1.62 (0.86 2.5)
1.78 (1.31 2.67)
0.77 (0.45 1.95)
< 0.001
IL-8 (pg/mL)
4.59 (3.03 9.17)
4.59 (3.11 7.16)
5.34 (2.86 14.43)
0.262
IL-9 (pg/mL)
0.8 (0.36 1.15)
0.95 (0.70 1.19)
0.31 (0.24 0.73)
< 0.001
IL-10 (pg/mL)
0.87 (0.60 1.80)
0.96 (0.65 2.13)
0.74 (0.49 1.22)
0.027
IL-13 (pg/mL)
0.05 (0.02 0.17)
0.06 (0.02 0.16)
0.03 (0.01 0.15)
0.071
IL-15 (pg/mL)
1.3 (0.77 1.77)
1.37 (0.94 1.98)
0.94 (0.54 1.65)
0.002
EGF (pg/mL)
3.92 (0.84 20.06)
5.96 (1.04 20.59)
1.81 (0.26 17.69)
0.084
G-CSF (pg/mL)
8.64 (4.27 17.69)
10.77 (5.68 16.06)
5.20 (0.20 18.61)
0.017
Flt-3 (pg/mL)
0.4 (0.07 2.26)
0.47 (0.08 2.53)
0.25 (0.03 1.60)
0.44
GM-CSF (pg/mL)
4.27 (1.19 20.61)
7.71 (2.13 26.46)
1.51 (0.83 6.47)
< 0.001
Fractalkine (pg/mL)
19.57 (3.62 59.16)
17.99 (7.06 56.99)
21.75 (0.07 60.03)
0.225
IFN-γ (pg/mL)
4.7 (2.06 9.17)
4.54 (2.19 7.75)
3.76 (1.43 13.67)
0.897
MDC (pg/mL)
335.44 (248.06 471.03)
337.56 (246.52 469.62)
352.74 (251.20 540.90)
0.365
MIP- (pg/mL)
2.03 (1.43 3.8)
2.03 (1.45 3.08)
2.39 (1.13 7.64)
0.485
Infarct volume (cm3)
1.26 (0.40 3.42)
0.86 (0.29 2.06)
3.07 (1.02 5.93)
< 0.001
NIHSS score on admission,
median (IQR)
3.00 (2.00 5.00)
3.00 (1.00 4.00)
7.00 (4.00 10.00)
< 0.001
NIHSS score on discharge,
median (IQR)
3.00 (1.00 5.00)
2.00 (1.00 3.00)
6.00 (4.75 9.00)
< 0.001
Medication, (%)
Statin
135 (80.8)
99 (87.6)
36 (66.7)
0.002
Lin S., et al Serum cytokines and ischemic outcome
Aging and Disease Volume 10, Number 2, April 2019 4
All participants were examined using Cranial Magnetic
Resonance Imaging (MRI) scans. The infarction size in
diffusion-weighted imaging (DWI) was measured using
G3PACS software by two neuroradiologists
independently. Firstly, we selected the slice with the
largest lesion by eye and the longest lesion axis (A axis)
on this slice was measured. A second line (B axis) was
drawn perpendicular to the first at the widest dimension.
A third axis, the z (C) axis, was computed by multiplying
the number of slices by slice thickness (7 mm). The
formula was 0.5 x A x B x C [35]. According to a previous
study, a small infarct volume was defined as less than 5
cm³, while a large infarct volume is larger than 5 cm³ [36].
Anticoagulation agents
8 (4.8)
6 (5.3)
2 (3.7)
0.448
Antiplatelet agents
132 (79.0)
97 (85.8)
35 (64.8)
0.001
Abbreviations: SBP, systolic blood pressure; DBP, diastolic blood pressure; BMI, body mass index; WBC, leukocyte; Hs-CRP, High-sensitivity C-
reactive protein; IL-1RA, interleukin-1 receptor antagonist; IL-1α, interleukin 1 alpha; IL-1β, interleukin 1 beta; IL-4, interleukin-4; IL-5, interleukin-
5; IL-6, interleukin-6; IL-7, interleukin-7; IL-8, interleukin-8; IL-9, interleukin-9; IL-10, interleukin-10; IL-13, interleukin-13; IL-15, interleukin-15;
Flt-3, Fms-like tyrosine kinase 3; EGF, epidermal growth factor ; G-CSF, granulocyte-colony stimulating factor; GM-CSF, granulocyte-macrophage
colony-stimulating factor; MDC, macrophage-derived chemokine; MIP-1α, macrophage inflammatory protein-1 alpha; IQR, interquartile range;
NIHSS, National Institutes of Health Stroke Scale.
Lin S., et al Serum cytokines and ischemic outcome
Aging and Disease Volume 10, Number 2, April 2019 5
Table 2. Levels of serum cytokines in different groups of stroke severity.
Median (IQR)
Cytokines
Minor Stroke Group
(N=53)
Severe Stroke Group
(N=94)
P value
Control (N=14)
Hs-CRP (mmol/L)
1.55 (0.59 3.50)
2.56 (1.39 4.81)
0.019
IL-1RA (pg/mL)
2.56 (1.35 9.45)
1.94 (0.48 8.15)
0.028
0.40(0.07-6.38)
IL- (pg/mL)
3.2 (0.35 14.43)
1.93 (0.19 24.28)
0.234
0.045 (0.02 28.49)
IL- (pg/mL)
1.11 (0.92 1.68)
0.93 (0.61 1.51)
< 0.001
0.73 (0.53 2.23)
IL-4 (pg/mL)
3.62 (1.48 9.31)
0.96 (0.32 3.10)
< 0.001
1.03 (0.44 5.45)
IL-5 (pg/mL)
0.70 (0.51 0.90)
0.40 (0.24 0.70)
< 0.001
0.23 (0.18 0.42)
IL-6 (pg/mL)
1.34 (0.80 2.32)
1.00 (0.49 1.76)
< 0.001
0.70 (0.33 1.36)
IL-7 (pg/mL)
2.10 (1.45 2.84)
1.09 (0.47 2.06)
< 0.001
0.82 (0.44 2.72)
IL-8 (pg/mL)
4.76 (3.39 8.45)
4.09 (2.38 11.73)
0.238
21.75 (2.05 82.14)
IL-9 (pg/mL)
1.01 (0.77 1.29)
0.37 (0.25 0.90)
< 0.001
0.33 (0.26 0.76)
IL-10 (pg/mL)
1.12 (0.69 2.14)
0.70 (0.49 1.27)
< 0.001
0.41 (0.23 1.16)
IL-13 (pg/mL)
0.075 (0.03 0.22)
0.03 (0.01 0.11)
< 0.001
0.02 (0.01 0.31)
IL-15 (pg/mL)
1.42 (1.07 2.13)
0.93 (0.59 1.65)
< 0.001
0.83 (0.55 1.38)
EGF (pg/mL)
6.69 (1.35 22.78)
2.14 (0.36 17.05)
0.016
17.83 (1.17 53.23)
G-CSF (pg/mL)
12.24 (7.00 19.82)
5.73 (0.22 14.05)
< 0.001
1.15 (0.27 9.91)
Flt-3 (pg/mL)
0.52 (0.10 2.83)
0.25 (0.04 1.60)
0.007
0.56 (0.10 23.93)
GM-CSF (pg/mL)
11.56 (2.42 41.66)
1.85 (0.91 9.17)
< 0.001
1.17 (0.86 5.27)
Fractalkine (pg/mL)
23.93 (7.69 73.27)
15.60 (0.20 50.88)
< 0.001
6.47 (0.20 23.93)
IFN-γ (pg/mL)
5.14 (2.53 7.98)
3.58 (1.53 11.00)
0.068
2.71 (1.39 11.44)
MDC (pg/mL)
333.35 (246.52 462.26)
352.74 (249.64 540.90)
0.123
731.67 (511.10 923.05)
MIP- (pg/mL)
2.18 (1.54 3.36)
1.75 (1.26 4.95)
0.754
5.23 (0.85 28.96)
Abbreviations: SBP, systolic blood pressure; DBP, diastolic blood pressure; BMI, body mass index; WBC, leukocyte; Hs-CRP, High-sensitivity C-
reactive protein; IL-1RA, interleukin-1 receptor antagonist; IL-1α, interleukin 1 alpha; IL-1β, interleukin 1 beta; IL-4, interleukin-4; IL-5,
interleukin-5; IL-6, interleukin-6; IL-7, interleukin-7; IL-8, interleukin-8; IL-9, interleukin-9; IL-10, interleukin-10; IL-13, interleukin-13; IL-15,
interleukin-15; Flt-3, Fms-like tyrosine kinase 3; EGF, epidermal growth factor ; G-CSF, granulocyte-colony stimulating factor; GM-CSF,
granulocyte-macrophage colony-stimulating factor; MDC, macrophage-derived chemokine; MIP-1α, macrophage inflammatory protein-1 alpha;
IQR, interquartile range; NIHSS, National Institutes of Health Stroke Scale.
Cytokine measurement
Blood samples of all participants were collected within 24
hrs after admission. The serum levels of cytokines,
chemokines, and growth factors were measured using
human cytokine/chemokine magnetic bead panel kit
(HCYTMAG-60K-PX38, EMD Millipore, Germany). All
procedures were performed according to the
manufacturer's instructions and data were analyzed with
the xPONENT software. Other laboratory parameters
included white blood cell (WBC) count, neutrophil and
high-sensitivity C-reactive protein (Hs-CRP) were tested
in the hospital’s central biochemistry laboratory.
Statistical analysis
All statistical analyses were performed with SPSS version
23.0 (Chicago, IL) and statistical significance was set at P
< 0.05. The Kolmogorov-Smirnov (K-S) test was applied
to assess the normality of continuous variables.
Continuous variables of a normal distribution were
expressed as the mean value ± standard deviation (SD)
and analyzed using the unpaired t-test. Non-normally
distributed variables were expressed as medians with
inter-quartile ranges (IQR) and analyzed using the Mann-
Whitney U test. Meanwhile, the Chi-Squared test was
used to compare frequency and percentage in categorical
variables. Spearman’s rank correlation or Pearson
correlation was performed for bivariate correlation
between NIHSS scores, infarct volume and serum
markers. The relationship between serum markers and 3-
months functional outcome of AIS patients was analyzed
by multivariate logistic regression analysis after adjusting
for confounders. The receiver operating characteristic
(ROC) curve was used to determine the predictive values
of the serum levels of these inflammatory markers to
predict short-term prognosis in AIS patients. In addition,
area under the curve (AUC) was computed as a
measurement of the accuracy of the data.
Lin S., et al Serum cytokines and ischemic outcome
Aging and Disease Volume 10, Number 2, April 2019 6
Figure 2. The relationship between various inflammatory cytokines and stroke outcomes. The levels of IL-4 and
IL-5 were significantly increased in the favorable outcome group compared with the poor outcome group. However, the
concentration of IL-6 was significantly decreased in the favorable outcome group. Data are presented as mean ± SD, *P
< 0.05, ***P < 0.001.
RESULTS
Baseline patient characteristics
A total of 180 patients that met the inclusion criteria were
recruited and 167 of them completed follow-up (13
patients could not be reached). The average age of AIS
patients was 63.02 ± 9.84 years and 60% of AIS patients
enrolled were male. The median (quartiles) NIHSS score
on admission was 3 (IQR, 2 5) and on discharge was 3
(IQR, 1 5). Poor outcome at 3 months was found in 54
patients (32.3%) with the average age being 65.00 ± 8.43
years. The characteristics of AIS patients with good or
poor 3-months outcomes are shown in Table 1.
Levels of serum cytokines reveal the severity of stroke
We initially screened 35 inflammatory mediators in 75
patients, and found that IL-4, IL-7, IL-9, GM-CSF and
MIP-1α were significantly decreased in the severe stroke
group, compared with control group (P < 0.05; data not
shown). AIS patients were then divided into two groups
according to the NIHSS score on admission: 53 (31.7%)
patients in the minor stroke group (NIHSS < 5) and 94
(68.3%) patients in the severe stroke group (NIHSS ≥ 5).
The serum levels of IL-1RA, IL-1β, IL-4, IL-5, IL-6, IL-
7, IL-9, IL-10, IL-13, IL-15, EGF, G-CSF, Flt-3L, GM-
CSF, and Fractalkine levels were significantly lower in
the severe versus (vs.) the minor stroke group (Fig. 1).
Circulating levels of IL-1α, MDC, MIP-1α, and IFN-γ not
altered (Table 2). Of note, the level of Hs-CRP was
significantly increased in the severe-stroke group vs. the
minor-stroke group.
Lin S., et al Serum cytokines and ischemic outcome
Aging and Disease Volume 10, Number 2, April 2019 7
Table 3. Logistic regression model with predictors of poor outcome (N=167).
Characteristics
Unadjusted OR (95% CI)
P value
Adjusted OR (95% CI)
P value
Age (years)
1.032 (0.997 – 1.069)
0.074
Male (%)
0.548 (0.284 – 1.058)
0.073
SBP (mmHg)
1.012 (0.998 – 1.026)
0.099
DBP (mmHg)
0.999 (0.976 – 1.023)
0.942
Hypertension (%)
1.720 (0.648 – 4.568)
0.276
Hyperlipidemia (%)
1.121 (0.496 – 2.530)
0.784
Diabetes (%)
1.667 (0.846 – 3.282)
0.139
Cardiac disease (%)
2.060 (0.784 – 5.414)
0.143
Smoking (%)
1.939 (0.990 – 3.800)
0.054
Alcohol drinking (%)
0.465 (0.204 – 1.056)
0.067
Stroke etiologic subtypes (%)
1.010 (0.699 – 1.460)
0.958
Large-artery atherosclerosis
Cardioembolic
Small-vessel disease
Other or unknown cause
BMI (kg/m2)
1.013 (0.913 – 1.123)
0.807
Laboratory tests
WBC (109 /L)
1.192 (0.998 – 1.423)
0.052
Neutrophils (109 /L)
1.333 (1.075 – 1.654)
0.009
Hs-CRP (mmol/L)
1.008 (0.985 – 1.033)
0.484
IL-1RA (pg/ml)
1.004 (0.996 – 1.011)
0.342
IL-1α (pg/ml)
1.001 (0.998 – 1.004)
0.593
IL-1β (pg/ml)
0.992 (0.813 – 1.210)
0.937
IL-4 (pg/ml)
0.869 (0.788 – 0.960)
0.005
IL-5 (pg/ml)
0.244 (0.087 – 0.682)
0.007
0.039 (0.003 – 0.475)
0.011
IL-6 (pg/ml)
1.051(1.002 – 1.103)
0.041
1.329 (1.095 – 1.612)
0.004
IL-7 (pg/ml)
0.981 (0.909 – 1.059)
0.630
IL-9 (pg/ml)
0.768 (0.508 – 1.161)
0.211
IL-10 (pg/ml)
0.962 (0.845 – 1.094)
0.554
IL-13 (pg/ml)
1.052 (0.853 – 1.299)
0.634
IL-15 (pg/ml)
0.726 (0.514 – 1.025)
0.069
EGF (pg/ml)
1.003 (0.996 – 1.009)
0.421
G-CSF (pg/ml)
1.004 (0.998 – 1.009)
0.165
GM-CSF (pg/ml)
1.000 (0.994 – 1.007)
0.916
Flt-3 (pg/ml)
1.015 (0.992 – 1.038)
0.200
Fractalkine (pg/ml)
1.002 (0.999 – 1.005)
0.244
IFN-γ (pg/ml)
1.004 (0.996 – 1.013)
0.324
MDC (pg/ml)
1.001 (1.000 – 1.003)
0.071
MIP-1α (pg/ml)
1.069 (1.010 – 1.131)
0.021
Infract volume (cm3)
1.043 (0.997 – 1.090)
0.065
NIHSS score on admission, median (IQR)
2.148 (1.679 – 2.748)
<0.001
NIHSS score on discharge, median (IQR)
2.477 (1.855 – 3.307)
<0.001
2.494 (1.364 – 4.562)
0.003
Medications, no. (%)
Statin
0.283 (0.128 – 0.627)
0.002
Anticoagulation agents
2.135 (0.293 – 5.576)
0.455
Antiplatelet agents
0.304 (0.141 – 0.656)
0.002
Abbreviations: SBP, systolic blood pressure; DBP, diastolic blood pressure; BMI, body mass index; WBC, leukocyte; Hs-CRP, High-
sensitivity C-reactive protein; IL-1RA, interleukin-1 receptor antagonist; IL-1α, interleukin 1 alpha; IL-1β, interleukin 1 beta; IL-4,
interleukin-4; IL-5, interleukin-5; IL-6, interleukin-6; IL-7, interleukin-7; IL-8, interleukin-8; IL-9, interleukin-9; IL-10, interleukin-10; IL-
13, interleukin-13; IL-15, interleukin-15; Flt-3, Fms-like tyrosine kinase 3; EGF, epidermal growth factor ; G-CSF, granulocyte-colony
stimulating factor; GM-CSF, granulocyte-macrophage colony-stimulating factor; MDC, macrophage-derived chemokine; MIP-1α,
macrophage inflammatory protein-1 alpha; IQR, interquartile range; NIHSS, National Institutes of Health Stroke Scale.
Lin S., et al Serum cytokines and ischemic outcome
Aging and Disease Volume 10, Number 2, April 2019 8
Levels of serum cytokines in relation to cerebral
infarction volume
All AIS patients underwent cranial MRI scans and the
median (quartiles) infarct volume was 1.26 (IQR, 0.40
3.42). 29 patients belonged to large infarct volume group
(≥ 5cm³; median 3.07; IQR, 1.02 – 5.93) and 138 patients
were in the small infarct volume group (< 5 cm³; median
0.86; IQR, 0.29 2.06). Serum Hs-CRP level was
significantly higher in patients with large infarct volumes
(P = 0.014), while IL-9 level increased significantly in the
small infarct volume group (P = 0.034). Interestingly,
serum IL-4 was found to be decreased in the large infarct
volume group although it did not reach significance (P =
0.090) (Table 1).
Figure 3. ROC curve of IL-5 for predicting 3-months
outcome of AIS patients. The optimal cutoff value was 0.385
pg/mL with a sensitivity of 86.6% and a specificity of 37.7%
(AUC: 0.719, 95% CI (0.625 0.813; P < 0.001).
Levels of serum cytokines predicts functional outcome
167 patients completed the 3-month follow-up. Poor
prognosis was found in 54 (32.3%) patients with 27 of
them (50.0%) being male and mainly occurred in older
ages and patients with a history of smoking. We found that
serum IL-4 level was significantly lower in AIS patients
with poor outcome compared with those with a good
prognosis 3.10 (IQP 1.13 8.15) pg/mL vs. 0.53 (IQR
0.28 1.32) pg/mL, P < 0.001), IL-5 level also
significantly decreased in the poor outcome group [0.63
(IQR 0.48 0.86) pg/mL vs. 0.32 (IQR, 0.24 0.66)
pg/mL, P < 0.001] (Fig. 2). Serum IL-6 was significantly
higher in the poor outcome group (Table 3) while serum
levels of IL-1β, IL-7, IL-9, IL-10, IL-15, G-CSF, GM-
CSF remained relatively constant.
By univariate logistic regression analysis, we found
that the serum concentrations of neutrophils, IL-4, IL-5,
IL-6, and MIP- were significantly associated with
functional outcome of AIS patients (Table 3).
Multivariate logistic regression was used to further
analysis these parameters in unadjusted models (including
age, gender, SBP, DM, cardiac disease, smoking, alcohol
drinking, WBCs, neutrophils, serum levels of IL-4, IL-5,
IL-6, MDC and MIP-1α, infract volume, NIHSS scores on
admission and on discharge, the use of antiplatelet agents
and statin). We found that serum concentration of IL-5
functioned as a protective factor and was an independent
predictor for functional outcome of AIS patients with an
adjusted OR of 0.039 (95% CI, 0.003 0.475, P = 0.011),
whereas IL-6 was an independent risk factor in functional
outcome with an adjusted OR of 1.329 (95% CI,1.095
1.612, P = 0.004). Furthermore, NIHSS score on
discharge has alsobeen used as an indpdendent predictor
of AIS patient outcome. Based on the ROC curve analysis,
the optimal cutoff value of IL-5 for predicting AIS
prognosis was projected to be 0.385 pg/mL (with a
sensitivity of 86.6% and a specificity of 37.7%) and AUC:
0.719, 95% CI (0.625 0.813, P < 0.001, Fig. 3).
DISCUSSION
In this study, we found that IL-1RA, IL-1β, IL-4, IL-5, IL-
6, IL-7, IL-9, IL-10, IL-13, IL-15, EGF, G-CSF, Flt-3L,
GM-CSF and Fractalkine levels were significantly
decreased in the severe stroke group. While in AIS
patients with poor outcome, IL-1β, IL-4, IL-5, IL-7, IL-9,
IL-10, IL-15, G-CSF and GM-CSF were significantly
reduced. We also found that IL-5 was an independent
protective factor for prognosis and IL-6 was an
independent risk predictor for AIS patients. Our data
suggest that the levels of serum cytokines were related to
stroke severity, short-term prognosis and cerebral infarct
volume of AIS patients.
A large body of evidence suggests that inflammation
is a major contributor to the pathophysiologic processes
of ischemic stroke. The intimate balance between pro- and
anti-inflammatory cytokines are relevant to the
susceptibility and functional outcome of patients with AIS
[5, 37]. IL-4 is a product of selective immune cells, acting
as a pleiotropic regulator of numerous immune and
inflammatory responses [38, 39]. It has been confirmed
that IL-4 could polarize macrophages from a
proinflammatory M1 phenotype to a “healing” M2
phenotype, which would play an anti-inflammatory role
in tissue repair [40, 41]. Zhao et al. reported that IL-4
secreted by neuronal cells in ischemia acts as a
Lin S., et al Serum cytokines and ischemic outcome
Aging and Disease Volume 10, Number 2, April 2019 9
neuroprotective mechanism to aid in the regulation of
intracerebral cleanup and repair after stroke [42]. IL-5, a
primary T-cell-derived cytokine, regulates eosinophil
development and is regarded as a significant contributor
to atopic diseases such as asthma [43, 44]. An increasing
number of studies support the idea that IL-5 induces the
production of anti-Ox-LDL antibodies and plays an anti-
atherosclerotic role by enhancing Th2-type immune
responses [45, 46]. mpi et al. suggested that IL-5 level
was related to antibodies binding to the Ox-LDL and
inversely connected with carotid intima-media thickness
[47]. Studies have found that the expression levels of IL-
4, IL-5, IL-6, and IL-10 are increased remarkably in the
damaged hemisphere after ischemic stroke. IL-4 is mostly
known as an anti-inflammatory cytokine and IL-5 have
been shown to suppress post-stroke inflammation [16]. In
our study, we found that serum concentrations of IL-4, IL-
5 and IL-9 were significantly elevated in minor stroke
patients and negatively related to the NIHSS score. These
biomarkers were also associated with functional outcome
of AIS patients and markedly decreased in AIS patients
with a poor prognosis. Moreover, we showed that IL-5
possessed a protective role and was an independent factor
that could predict AIS patients’ prognosis. Thus, we
speculate that IL-4, IL-5 and IL-9 could act as protective
factors for AIS. In fact, Xiong et al. demonstrated that IL-
4 was somehow involved in cerebral ischemic outcome as
IL-4 deficiency resulted in a greater degree of ischemic
brain damage [15]. Similarly, Sheikh et al. demonstrated
that pretreatment of HMO6 cells with IL-5 could suppress
the expression of IL-and IFNγ-induced mRNA, and
IL-5 had the capability to inhibit focal ischemia-induced
inflammation [17]. Therefore, the regulation of IL-4 or
IL-5 and its associated pathways are potential targets for
the treatment of cerebral ischemia. As a pro-inflammatory
cytokine, IL-9 can promote CCL20 release from
astrocytes and the migration of Th17 cells into the central
nervous system [48]. The mechanisms underlying IL-9
and Th9 cells-mediated ischemic injury are largely
unknown. Like TNF-α and IL-1β, IL-9 may directly
damage brain tissue as they perpetuate pro-inflammatory
actions [49, 50]. Tan et al. demonstrated that the
expression level of IL-9 and percentages of Th9 and Tc9
cells were notably higher in (PBMCs) derived from
ischemic stroke patients. Moreover, an increased level of
IL-9 may compromise the BBB’s integrity through IL-
9R/STAT1,3 pathways [51], but this was negative
correlated with stroke severity.
Notably, there was a controversy regarding the
source of elevated IL-6 levels in the early stroke phase
[52]; IL-6 possessed multipotent functions and was
upregulated during brain injury or the repair process [53].
IL-6 plays a dual role in the inflammatory response
induced by cerebral ischemia. On one hand, IL-6 would
further aggravate the deterioration of brain damage and
counteract neural stem cell proliferation in the acute phase
of cerebral ischemia. On the other hand, IL-6 would
restore and enhance glial cells to prevent collagen
deposition and repair brain damage in the subacute phase
[54]. Previous studies reported that circulating IL-6 was
closely correlated with brain infarct volume or stroke
severity [53, 55], although contradicting results were
reported [25]. Besides, Smith et al. revealed that peak
plasma IL-6 level in stroke patients was correlated with
poor prognosis [55]. A recent report recommended that
IL-6 induced by IL-1 was associated with worse prognosis
in stroke patients, and that clinical outcome would be
improved after the inflammatory factor was reduced by
IL-1 receptor antagonist [56]. Consistently, our study
showed that IL-6 level was significantly elevated in AIS
patients with poor outcome. However, Karen et al.
suggested that IL-6 is beneficial for long-term prognosis
as IL-6 could promote early transcriptional changes in
angiogenesis-related genes after cerebral ischemia, which
afford long-term histological and functional protection
[57]. Therefore, this phenomenon needs to be further
studied. We found that the level of serum IL-7 was
notably reduced in patients with severe stroke and poor
outcome. As a pleiotropic cytokine, IL-7 is capable of
multiple effects [58]. IL-7 could act as a regulator of
growth or anti-apoptosis in pre-B cells, and maintain the
steady state proliferation of mature T cells, which might
effectively reduce neuroinflammatory and some
autoimmune responses through the inhibition of signal
transduction [20, 59]. Cagnin and Damas reported that IL-
7 was upregulated in patients with cardiovascular
atherosclerosis (particularly among patients with acute
myocardial infarction and angina pectoris) compared with
controls [60, 61]. Controversially, other studies showed
that IL-7 level was significantly reduced in stroke patients
compared to controls [62]. Lawson et al. speculated that
IL-7 signaling was a prerequisite for activating optimal
CD4+ T cell and that IL-7R antagonism could be an
effective treatment for CD4+ T cell-mediated
neuroinflammation and several inflammatory
autoimmune diseases [59].
Here, we also found that levels of IL-1β, IL-1RA, IL-
10, IL-13, IL-15, Flt-3L, Fractalkine, EGF, G-CSF and
GM-CSF were lower in the severe stroke group. IL-1β is
an important mediator of the inflammatory response and
involved in a variety of cellular activities, including cell
proliferation, differentiation, and apoptosis. Several
studies found that IL- were elevated in the AIS group
[63, 64]. Similarly, IL-1RA levels were found to be
elevated in stroke patients in previous studies [65, 66]. IL-
10 is an anti-inflammatory cytokine produced by T cells
and monocytes, suggesting its participation in vascular
protection, although the exact mechanisms are unclear. In
Lin S., et al Serum cytokines and ischemic outcome
Aging and Disease Volume 10, Number 2, April 2019 10
an animal model, cerebral infarct volume have been
reduced through intraventricular or systemic
administration of IL-10 [67], and IL-10 has been
suggested to be neuroprotective [67, 68]. IL-13 is
a cytokine mainly secreted by Th2 cells [22]. The
secondary structural features of IL-13 are like IL-4 and
share similar functions with IL-4. IL-13 possess anti-
inflammatory properties and is mainly associated with
diseases involving the airway. However, there are few
reports regarding IL-13’s participation in stroke. IL-15 is
a cytokine with structural similarity to IL-2. Lee et al.
found that IL-15 promote astrocyte survival in response to
OGD, thus can be beneficial to ischemic stroke [69].
However, there is no clinical data showing a link between
IL-15 and stroke. Flt3L is a growth factor, which can
stimulate not only the proliferation and differentiation of
hematopoietic progenitor cells, leading to increased
numbers of pre-B cells, but also the maturation of T, B
and NK cells [70, 71]. Fractalkine was initially discovered
as an adhesion molecule for lymphocytes and monocytes,
natural killer cells, and microglia, indicating its role for
regulating the inflammatory response [72]; Fractalkine
can also decrease microglial activation and release pro-
inflammatory cytokines [73, 74]. EGF, G-CSF and CM-
CSF are trophic factors and capable of neuroprotection.
Administration of EGF and G-CSF in a rat MCAO model
could reduce infarct volume [75, 76]. Navarro et al. found
that the level of GM-CSF was significantly higher in
stroke patients than in healthy controls, and was positively
correlated with NIHSS score [77].
MDC is a member of the CC-chemokine family and
is mainly produced by macrophages and dendritic cells.
There is an increasing number of reports regarding the
involvement of MDC in a variety of diseases, ranging
from allergic reactions to HIV infection and neoplasia
[78]. Kimura et al. suggested that MDC promotes
atherosclerosis by migration or recruitment of monocyte-
derived cells and the stimulation of platelet activity [79].
MIP- belong to the CC-chemokine subfamily,
possessing inflammatory and neutrophil chemokinetic
properties [80]. Gourmala et al. revealed an early increase
in macrophage inflammatory protein-and macrophage
inflammatory protein- messenger RNA levels in a rat
MCAO model [81].
Several limitations in the study should not be
neglected. First, this is a single center study with a small
number of patients. As stroke patients are mostly elderly
persons, it was difficult to find age-matched healthy
subjects. Second, serum cytokine levels were tested only
at admission. Monitoring the dynamic change of serum
biomarker levels will be essential in further studies. Last,
a minor stroke is generally defined as a NIHSS of 5 or
less, which only considers certain deficits but not the fact
that some patients can have a more profound impact on
the quality of life versus others. Therefore, the scale does
not linearly correlate deficit and disability. Recent studies
suggest that score of an NIHSS of 3 or less may be a better
definition of a minor stroke [82, 83].
In conclusion, the present study is the first report to
demonstrate the simultaneous measurement of 35 serum
cytokines, chemokines, and growth factors in patients
with AIS. We found that the serum concentration of these
factors is significantly associated with the outcome for the
AIS patient. Indeed, an increased knowledge of
inflammatory mediators in response to AIS may provide
a basis for the design and development of new
pharmacological approaches to treat stroke.
Acknowledgments
This work is supported by US Public Health Service
Grants NS57186 and AG21980 (to K.J.)
References
[1] Collaborators GBDCoD (2017). Global, regional, and
national age-sex specific mortality for 264 causes of
death, 1980-2016: a systematic analysis for the Global
Burden of Disease Study 2016. Lancet, 390:1151-1210.
[2] Benjamin EJ, Blaha MJ, Chiuve SE, Cushman M, Das
SR, Deo R, et al. (2017). Heart Disease and Stroke
Statistics-2017 Update: A Report From the American
Heart Association. Circulation, 135:e146-e603.
[3] Johnston SC, Mendis S, Mathers CD (2009). Global
variation in stroke burden and mortality: estimates from
monitoring, surveillance, and modelling. Lancet Neurol,
8:345-354.
[4] Allen CL, Bayraktutan U (2009). Oxidative stress and its
role in the pathogenesis of ischaemic stroke. Int J Stroke,
4:461-470.
[5] Chamorro A, Hallenbeck J (2006). The harms and
benefits of inflammatory and immune responses in
vascular disease. Stroke, 37:291-293.
[6] McColl BW, Allan SM, Rothwell NJ (2009). Systemic
infection, inflammation and acute ischemic stroke.
Neuroscience, 158:1049-1061.
[7] Wang R, Li J, Duan Y, Tao Z, Zhao H, Luo Y (2017).
Effects of Erythropoietin on Gliogenesis during Cerebral
Ischemic/Reperfusion Recovery in Adult Mice. Aging
Dis, 8:410-419.
[8] Tang Y, Wang L, Wang J, Lin X, Wang Y, Jin K, et al.
(2016). Ischemia-induced Angiogenesis is Attenuated in
Aged Rats. Aging Dis, 7:326-335.
[9] McCombe PA, Read SJ (2008). Immune and
inflammatory responses to stroke: good or bad? Int J
Stroke, 3:254-265.
[10] Castellon X, Bogdanova V (2016). Chronic
Inflammatory Diseases and Endothelial Dysfunction.
Aging Dis, 7:81-89.
[11] Denes A, Thornton P, Rothwell NJ, Allan SM (2010).
Inflammation and brain injury: Acute cerebral ischaemia,
Lin S., et al Serum cytokines and ischemic outcome
Aging and Disease Volume 10, Number 2, April 2019 11
peripheral and central inflammation. Brain Behavior and
Immunity, 24:708-723.
[12] Reaux-Le Goazigo A, Van Steenwinckel J, Rostene W,
Melik Parsadaniantz S (2013). Current status of
chemokines in the adult CNS. Prog Neurobiol, 104:67-
92.
[13] Qian L, Yuanshao L, Wensi H, Yulei Z, Xiaoli C, Brian
W, et al. (2016). Serum IL-33 Is a Novel Diagnostic and
Prognostic Biomarker in Acute Ischemic Stroke. Aging
Dis, 7:614-622.
[14] Chen C, Chu SF, Liu DD, Zhang Z, Kong LL, Zhou X,
et al. (2018). Chemokines play complex roles in cerebral
ischemia. Neurochem Int, 112:146-158.
[15] Xiong X, Barreto GE, Xu L, Ouyang YB, Xie X, Giffard
RG (2011). Increased brain injury and worsened
neurological outcome in interleukin-4 knockout mice
after transient focal cerebral ischemia. Stroke, 42:2026-
2032.
[16] Pena-Philippides JC, Caballero-Garrido E,
Lordkipanidze T, Roitbak T (2016). In vivo inhibition of
miR-155 significantly alters post-stroke inflammatory
response. Journal of Neuroinflammation, 13.
[17] Sheikh AM, Nagai A, Wakabayashi K, Narantuya D,
Kobayashi S, Yamaguchi S, et al. (2011). Mesenchymal
stem cell transplantation modulates neuroinflammation
in focal cerebral ischemia: contribution of fractalkine
and IL-5. Neurobiol Dis, 41:717-724.
[18] Suzuki S, Tanaka K, Suzuki N (2009). Ambivalent
aspects of interleukin-6 in cerebral ischemia:
inflammatory versus neurotrophic aspects. Journal of
Cerebral Blood Flow and Metabolism, 29:464-479.
[19] Erta M, Quintana A, Hidalgo J (2012). Interleukin-6, a
major cytokine in the central nervous system. Int J Biol
Sci, 8:1254-1266.
[20] Arya AK, Pokharia D, Bhan S, Tripathi R, Tripathi K
(2012). Correlation between IL-7 and MCP-1 in diabetic
chronic non healing ulcer patients at higher risk of
coronary artery disease. Cytokine, 60:767-771.
[21] Han HS, Yenari MA (2003). Cellular targets of brain
inflammation in stroke. Curr Opin Investig Drugs, 4:522-
529.
[22] Rael EL, Lockey RF (2011). Interleukin-13 signaling
and its role in asthma. World Allergy Organ J, 4:54-64.
[23] Feuerstein GZ, Wang X, Barone FC (1998). The role of
cytokines in the neuropathology of stroke and
neurotrauma. Neuroimmunomodulation, 5:143-159.
[24] Fassbender K, Rossol S, Kammer T, Daffertshofer M,
Wirth S, Dollman M, et al. (1994). Proinflammatory
cytokines in serum of patients with acute cerebral
ischemia: kinetics of secretion and relation to the extent
of brain damage and outcome of disease. J Neurol Sci,
122:135-139.
[25] Tarkowski E, Rosengren L, Blomstrand C, Wikkelso C,
Jensen C, Ekholm S, et al. (1995). Early intrathecal
production of interleukin-6 predicts the size of brain
lesion in stroke. Stroke, 26:1393-1398.
[26] (1989). Stroke--1989. Recommendations on stroke
prevention, diagnosis, and therapy. Report of the WHO
Task Force on Stroke and other Cerebrovascular
Disorders. Stroke, 20:1407-1431.
[27] Adams HP, Jr., Bendixen BH, Kappelle LJ, Biller J, Love
BB, Gordon DL, et al. (1993). Classification of subtype
of acute ischemic stroke. Definitions for use in a
multicenter clinical trial. TOAST. Trial of Org 10172 in
Acute Stroke Treatment. Stroke, 24:35-41.
[28] Brott T, Adams HP, Jr., Olinger CP, Marler JR, Barsan
WG, Biller J, et al. (1989). Measurements of acute
cerebral infarction: a clinical examination scale. Stroke,
20:864-870.
[29] Sulter G, Steen C, De Keyser J (1999). Use of the Barthel
index and modified Rankin scale in acute stroke trials.
Stroke, 30:1538-1541.
[30] Warnecke T, Im S, Kaiser C, Hamacher C, Oelenberg S,
Dziewas R (2017). Aspiration and dysphagia screening
in acute stroke - the Gugging Swallowing Screen
revisited. Eur J Neurol, 24:594-601.
[31] Yakhkind A, McTaggart RA, Jayaraman MV, Siket MS,
Silver B, Yaghi S (2016). Minor Stroke and Transient
Ischemic Attack: Research and Practice. Front Neurol,
7:86.
[32] Bamford JM, Sandercock PA, Warlow CP, Slattery J
(1989). Interobserver agreement for the assessment of
handicap in stroke patients. Stroke, 20:828.
[33] Kim J, Song TJ, Park JH, Lee HS, Nam CM, Nam HS, et
al. (2012). Different prognostic value of white blood cell
subtypes in patients with acute cerebral infarction.
Atherosclerosis, 222:464-467.
[34] Huybrechts KF, Caro JJ (2007). The Barthel Index and
modified Rankin Scale as prognostic tools for long-term
outcomes after stroke: a qualitative review of the
literature. Curr Med Res Opin, 23:1627-1636.
[35] Sims JR, Gharai LR, Schaefer PW, Vangel M, Rosenthal
ES, Lev MH, et al. (2009). ABC/2 for rapid clinical
estimate of infarct, perfusion, and mismatch volumes.
Neurology, 72:2104-2110.
[36] Li Q, Lin YS, Huang WS, Zhou YL, Chen XL, Wang BA,
et al. (2016). Serum IL-33 Is a Novel Diagnostic and
Prognostic Biomarker in Acute Ischemic Stroke. Aging
and Disease, 7:614-622.
[37] Vila N, Castillo J, Davalos A, Esteve A, Planas AM,
Chamorro A (2003). Levels of anti-inflammatory
cytokines and neurological worsening in acute ischemic
stroke. Stroke, 34:671-675.
[38] Luzina IG, Keegan AD, Heller NM, Rook GA, Shea-
Donohue T, Atamas SP (2012). Regulation of
inflammation by interleukin-4: a review of "alternatives".
J Leukoc Biol, 92:753-764.
[39] Wang HW, Joyce JA (2010). Alternative activation of
tumor-associated macrophages by IL-4: priming for
protumoral functions. Cell Cycle, 9:4824-4835.
[40] Sica A, Mantovani A (2012). Macrophage plasticity and
polarization: in vivo veritas. J Clin Invest, 122:787-795.
[41] Balce DR, Li BQ, Allan ERO, Rybicka JM, Krohn RM,
Yates RM (2011). Alternative activation of macrophages
by IL-4 enhances the proteolytic capacity of their
phagosomes through synergistic mechanisms. Blood,
118:4199-4208.
[42] Zhao XR, Wang H, Sun GH, Zhang J, Edwards NJ,
Aronowski J (2015). Neuronal Interleukin-4 as a
Modulator of Microglial Pathways and Ischemic Brain
Lin S., et al Serum cytokines and ischemic outcome
Aging and Disease Volume 10, Number 2, April 2019 12
Damage. Journal of Neuroscience, 35:11281-11291.
[43] Stein ML, Villanueva JM, Buckmeier BK, Yamada Y,
Filipovich AH, Assa'ad AH, et al. (2008). Anti-IL-5
(mepolizumab) therapy reduces eosinophil activation ex
vivo and increases IL-5 and IL-5 receptor levels. J
Allergy Clin Immunol, 121:1473-1483, 1483 e1471-
1474.
[44] Luk AO, Wang Y, Ma RC, Tam CH, Ng MC, Lam V, et
al. (2011). Predictive role of polymorphisms in
interleukin-5 receptor alpha-subunit, lipoprotein lipase,
integrin A2 and nitric oxide synthase genes on ischemic
stroke in type 2 diabetes--an 8-year prospective cohort
analysis of 1327 Chinese patients. Atherosclerosis,
215:130-135.
[45] Taleb S (2016). Inflammation in atherosclerosis. Arch
Cardiovasc Dis, 109:708-715.
[46] Binder CJ, Hartvigsen K, Chang MK, Miller M, Broide
D, Palinski W, et al. (2004). IL-5 links adaptive and
natural immunity specific for epitopes of oxidized LDL
and protects from atherosclerosis. J Clin Invest, 114:427-
437.
[47] Sampi M, Ukkola O, Paivansalo M, Kesaniemi YA,
Binder CJ, Horkko S (2008). Plasma Interleukin-5
Levels Are Related to Antibodies Binding to Oxidized
Low-Density Lipoprotein and to Decreased Subclinical
Atherosclerosis. Journal of the American College of
Cardiology, 52:1370-1378.
[48] Zhou Y, Sonobe Y, Akahori T, Jin S, Kawanokuchi J,
Noda M, et al. (2011). IL-9 promotes Th17 cell
migration into the central nervous system via CC
chemokine ligand-20 produced by astrocytes. J Immunol,
186:4415-4421.
[49] Yang GY, Gong C, Qin Z, Ye W, Mao Y, Bertz AL (1998).
Inhibition of TNFalpha attenuates infarct volume and
ICAM-1 expression in ischemic mouse brain.
Neuroreport, 9:2131-2134.
[50] Barone FC, Arvin B, White RF, Miller A, Webb CL,
Willette RN, et al. (1997). Tumor necrosis factor-alpha.
A mediator of focal ischemic brain injury. Stroke,
28:1233-1244.
[51] Tan S, Shan Y, Wang Y, Lin Y, Liao S, Deng Z, et al.
(2017). Exacerbation of oxygen-glucose deprivation-
induced blood-brain barrier disruption: potential
pathogenic role of interleukin-9 in ischemic stroke. Clin
Sci (Lond), 131:1499-1513.
[52] Dziedzic T, Slowik A, Szczudlik A (2003). Interleukin-6
and stroke: cerebral ischemia versus nonspecific factors
influencing interleukin-6. Stroke, 34:e229-230; author
reply e229-230.
[53] Acalovschi D, Wiest T, Hartmann M, Farahmi M,
Mansmann U, Auffarth GU, et al. (2003). Multiple levels
of regulation of the interleukin-6 system in stroke. Stroke,
34:1864-1869.
[54] Park SY, Kim J, Kim OJ, Kim JK, Song J, Shin DA, et
al. (2013). Predictive value of circulating interleukin-6
and heart-type fatty acid binding protein for three months
clinical outcome in acute cerebral infarction: multiple
blood markers profiling study. Crit Care, 17:R45.
[55] Smith CJ, Emsley HC, Gavin CM, Georgiou RF, Vail A,
Barberan EM, et al. (2004). Peak plasma interleukin-6
and other peripheral markers of inflammation in the first
week of ischaemic stroke correlate with brain infarct
volume, stroke severity and long-term outcome. BMC
Neurol, 4:2.
[56] Smith CJ, Hulme S, Vail A, Heal C, Parry-Jones AR,
Scarth S, et al. (2018). SCIL-STROKE (Subcutaneous
Interleukin-1 Receptor Antagonist in Ischemic Stroke):
A Randomized Controlled Phase 2 Trial. Stroke.
[57] Gertz K, Kronenberg G, Kalin RE, Baldinger T, Werner
C, Balkaya M, et al. (2012). Essential role of interleukin-
6 in post-stroke angiogenesis. Brain, 135:1964-1980.
[58] Vignali D, Cantarelli E, Bordignon C, Canu A, Citro A,
Annoni A, et al. (2018). Detection and Characterization
of CD8+ Autoreactive Memory Stem T Cells in Patients
with Type 1 Diabetes. Diabetes.
[59] Lawson BR, Gonzalez-Quintial R, Eleftheriadis T, Farrar
MA, Miller SD, Sauer K, et al. (2015). Interleukin-7 is
required for CD4(+) T cell activation and autoimmune
neuroinflammation. Clinical Immunology, 161:260-269.
[60] Cagnin S, Biscuola M, Patuzzo C, Trabetti E, Pasquali A,
Laveder P, et al. (2009). Reconstruction and functional
analysis of altered molecular pathways in human
atherosclerotic arteries. BMC Genomics, 10:13.
[61] Damas JK, Waehre T, Yndestad A, Otterdal K,
Hognestad A, Solum NO, et al. (2003). Interleukin-7-
mediated inflammation in unstable angina - Possible role
of chemokines and platelets. Circulation, 107:2670-2676.
[62] Oberheiden T, Nguyen XD, Fatar M, Elmas E, Blahak C,
Morper N, et al. (2012). Platelet and Monocyte
Activation in Acute Ischemic Stroke-Is There a
Correlation With Stroke Etiology? Clinical and Applied
Thrombosis-Hemostasis, 18:87-91.
[63] Licata G, Tuttolomondo A, Di Raimondo D, Corrao S,
Di Sciacca R, Pinto A (2009). Immuno-inflammatory
activation in acute cardio-embolic strokes in comparison
with other subtypes of ischaemic stroke. Thromb
Haemost, 101:929-937.
[64] Mazzotta G, Sarchielli P, Caso V, Paciaroni M, Floridi A,
Floridi A, et al. (2004). Different cytokine levels in
thrombolysis patients as predictors for clinical outcome.
Eur J Neurol, 11:377-381.
[65] Emsley HC, Smith CJ, Gavin CM, Georgiou RF, Vail A,
Barberan EM, et al. (2007). Clinical outcome following
acute ischaemic stroke relates to both activation and
autoregulatory inhibition of cytokine production. BMC
Neurol, 7:5.
[66] Beamer NB, Coull BM, Clark WM, Hazel JS, Silberger
JR (1995). Interleukin-6 and interleukin-1 receptor
antagonist in acute stroke. Ann Neurol, 37:800-805.
[67] Spera PA, Ellison JA, Feuerstein GZ, Barone FC (1998).
IL-10 reduces rat brain injury following focal stroke.
Neurosci Lett, 251:189-192.
[68] Rodriguez-Yanez M, Sobrino T, Arias S, Vazquez-
Herrero F, Brea D, Blanco M, et al. (2011). Early
biomarkers of clinical-diffusion mismatch in acute
ischemic stroke. Stroke, 42:2813-2818.
[69] Lee GA, Lai YG, Chen RJ, Liao NS (2017). Interleukin
15 activates Akt to protect astrocytes from oxygen
glucose deprivation-induced cell death. Cytokine, 92:68-
74.
Lin S., et al Serum cytokines and ischemic outcome
Aging and Disease Volume 10, Number 2, April 2019 13
[70] Parajuli P, Mosley RL, Pisarev V, Chavez J, Ulrich A,
Varney M, et al. (2001). Flt3 ligand and granulocyte-
macrophage colony-stimulating factor preferentially
expand and stimulate different dendritic and T-cell
subsets. Exp Hematol, 29:1185-1193.
[71] Namikawa R, Muench MO, de Vries JE, Roncarolo MG
(1996). The FLK2/FLT3 ligand synergizes with
interleukin-7 in promoting stromal-cell-independent
expansion and differentiation of human fetal pro-B cells
in vitro. Blood, 87:1881-1890.
[72] Bazan JF, Bacon KB, Hardiman G, Wang W, Soo K,
Rossi D, et al. (1997). A new class of membrane-bound
chemokine with a CX3C motif. Nature, 385:640-644.
[73] Pabon MM, Bachstetter AD, Hudson CE, Gemma C,
Bickford PC (2011). CX3CL1 reduces neurotoxicity and
microglial activation in a rat model of Parkinson's
disease. J Neuroinflammation, 8:9.
[74] Zujovic V, Benavides J, Vige X, Carter C, Taupin V
(2000). Fractalkine modulates TNF-alpha secretion and
neurotoxicity induced by microglial activation. Glia,
29:305-315.
[75] Jin K, Sun Y, Xie L, Childs J, Mao XO, Greenberg DA
(2004). Post-ischemic administration of heparin-binding
epidermal growth factor-like growth factor (HB-EGF)
reduces infarct size and modifies neurogenesis after focal
cerebral ischemia in the rat. J Cereb Blood Flow Metab,
24:399-408.
[76] Schabitz WR, Kollmar R, Schwaninger M, Juettler E,
Bardutzky J, Scholzke MN, et al. (2003).
Neuroprotective effect of granulocyte colony-
stimulating factor after focal cerebral ischemia. Stroke,
34:745-751.
[77] Navarro-Sobrino M, Rosell A, Penalba A, Ribo M,
Alvarez-Sabin J, Fernandez-Cadenas I, et al. (2009).
Role of endogenous granulocyte-macrophage colony
stimulating factor following stroke and relationship to
neurological outcome. Curr Neurovasc Res, 6:246-251.
[78] Mantovani A, Gray PA, Van Damme J, Sozzani S (2000).
Macrophage-derived chemokine (MDC). J Leukoc Biol,
68:400-404.
[79] Kimura S, Tanimoto A, Wang KY, Shimajiri S, Guo X,
Tasaki T, et al. (2012). Expression of macrophage-
derived chemokine (CCL22) in atherosclerosis and
regulation by histamine via the H2 receptor. Pathol Int,
62:675-683.
[80] Wolpe SD, Davatelis G, Sherry B, Beutler B, Hesse DG,
Nguyen HT, et al. (1988). Macrophages secrete a novel
heparin-binding protein with inflammatory and
neutrophil chemokinetic properties. J Exp Med,
167:570-581.
[81] Gourmala NG, Limonta S, Bochelen D, Sauter A,
Boddeke HW (1999). Localization of macrophage
inflammatory protein: macrophage inflammatory
protein-1 expression in rat brain after peripheral
administration of lipopolysaccharide and focal cerebral
ischemia. Neuroscience, 88:1255-1266.
[82] Fischer U, Baumgartner A, Arnold M, Nedeltchev K,
Gralla J, De Marchis GM, et al. (2010). What is a minor
stroke? Stroke, 41:661-666.
[83] Park TH, Hong KS, Choi JC, Song P, Lee JS, Lee J, et al.
(2013). Validation of minor stroke definitions for
thrombolysis decision making. J Stroke Cerebrovasc Dis,
22:482-490.
... As for the analyzed cytokines at baseline (Fig. 3), we only detected a notable correlation between MPO-histone complexes and IL-5. IL-5 has been discussed as a potential predictive factor for positive functional outcome after stroke [33]. Interestingly, our results indicated a similar, although confounded, association between MPO-histone complexes and recanalization, as the amount of MPOhistone complexes was higher in patients with favorable recanalization outcome. ...
Article
Full-text available
Inflammation including immunothrombosis by neutrophil extracellular traps (NETs) has important implications in acute ischemic stroke and can affect reperfusion status, susceptibility to stroke associated infections (SAI) as well as functional clinical outcome. NETs were shown to be prevalent in stroke thrombi and NET associated markers were found in stroke patients’ blood. However, little is known whether blood derived NET markers reflect the amount of NETs in thrombi. Conclusions from blood derived markers to thrombus composition might open avenues for novel strategies in diagnostic and therapeutic approaches. We prospectively recruited 166 patients with acute ischemic stroke undergoing mechanical thrombectomy between March 2018 and May 2021. Available thrombi (n = 106) were stained for NET markers DNA-histone-1 complexes and myeloperoxidase (MPO). Cell free DNA (cfDNA), deoxyribonuclease (DNase) activity, MPO-histone complexes and a cytokine-panel were measured before thrombectomy and after seven days. Clinical data, including stroke etiology, reperfusion status, SAI and functional outcome after rehabilitation, were collected of all patients. NET markers were present in all thrombi. At onset the median concentration of cfDNA in blood was 0.19 µg/ml increasing to 0.30 µg/ml at 7 days. Median DNase activity at onset was 4.33 pmol/min/ml increasing to 4.96 pmol/min/ml at 7 days. Within thrombi DNA-histone-1 complexes and MPO correlated with each other (ρ = 0.792; p < 0.001). Moreover, our study provides evidence for an association between the amount of NETs and endogenous DNase activity in blood with amounts of NETs in cerebral thrombi. However, these associations need to be confirmed in larger cohorts, to investigate the potential clinical implications for individualized therapeutic and diagnostic approaches in acute ischemic stroke. Graphical Abstract
... Inflammation is an important variable for explaining the pathophysiology of acute ischemic stroke. 1,2 The levels of inflammatory mediators have been measured in normal brain tissue. Proinflammatory cytokines are released from ischemic tissue, thus continuously releasing immune cells. ...
Article
Full-text available
Background & Objectives: To evaluate immature granulocytes, a new inflammatory biomarker, and other markers in short- and long-term prognosis in patients with acute ischemic stroke (AIS). Methods: Laboratory information system data from a tertiary hospital in Turkiye were used in this retrospective study. Of the 327 patients with the diagnosis of AIS, 275 recovered, and 52 died. It was determined that 31 of these 275 patients, who were followed up retrospectively, died within 12 months after discharge. Routinely measured immature granulocyte (IG), other hemogram parameters in the Sysmex XN 1000 (XN-1000-Hematology-Analyzer-Sysmex Corporation, Japan), and demographic data were statistically compared in both groups. We tried to estimate the short- and long-term mortality from the blood samples of these patients at their first admission to the hospital. Results: Of the patients included in the study, 150 (45.9%) were female, and 177 (54.1%) were male. National Institutes Of Health Stroke Scale (NIHSS) (AUC=960), length of stay (AUC=791), red blood cell distribution width – standard deviation (RDW-CV) (AUC=728), and IG (AUC=712) were the most effective parameters in predicting short-term mortality, while age (AUC=764) in predicting long-term mortality was the most effective parameters. Conclusion: IG, together with NIHSS and length of stay, shows moderate and high predictive properties in prognosticating short-term mortality but is ineffective in prognosticating long-term mortality. Age was found to be the most predictive marker for long-term mortality.
Article
Full-text available
Cannabidiol (CBD) is the most relevant nonpsychostimulant phytocompound found in Cannabis sativa. CBD has been extensively studied and has been proposed as a therapeutic candidate for neuroinflammation-related conditions. However, being a highly lipophilic drug, it has several drawbacks for pharmaceutical use, including low solubility and high permeability. Synthetic polymers can be used as drug delivery systems to improve CBD’s stability, half-life, and biodistribution. Here, we propose using a synthetic polymer as a nanoparticulate vehicle for CBD (NPCBD) to overcome the pharmacological drawbacks of free drugs. We tested the NPCBDengineered system in the context of ischemic events in a relevant oxygen and glucose deprivation (OGD) model in primary cortical cells (PCC). Moreover, we have characterized the inflammatory response of relevant cell types, such as THP-1 (human monocytes), HMC3 (human microglia), and PCC, to NPCBD and observed a shift in the inflammatory state of the treated cells after the ischemic event. In addition, NPCBD exhibited a promising ability to restore mitochondrial function after OGD insult in both HMC3 and PCC cells at low doses of 1 and 0.2 μM CBD. Taken together, these results suggest the potential for preclinical use.
Article
Full-text available
Acute ischemic stroke (AIS) is one of the most common strokes posing a grave threat to human life and health. Predicting the prognosis of AIS allows for an understanding of disease progress, and a better quality of life by making individualized treatment scheme. In this paper, we conducted a systematic search on PubMed, focusing on the relevant literature in the last 5 years. Summarizing the candidate prognostic biomarkers of AIS in body fluids such as blood, urine, saliva and cerebrospinal fluid is often of great significance for the management of acute ischemic stroke, which has the potential to facilitate early diagnosis, treatment, prevention and long-term outcome improvement.
Article
Full-text available
Background Decreased eosinophil level is associated with poor outcomes after mechanical thrombectomy in patients with acute ischemic stroke (AIS), but the underlying mechanism of this association is elusive. We aimed to assess the mediation effect of symptomatic intracranial hemorrhage (sICH) on the aforementioned association. Methods A total of 328 consecutive AIS patients undergoing mechanical thrombectomy between May 2017 and March 2021 were analyzed. SICH was defined as any evidence of brain hemorrhage on CT scan with neurological deterioration. Regression analysis was used to assess the effect of eosinophils on sICH, and its effect on poor outcome. Mediation analysis was performed to assess the proportion of total effect by sICH on the association between eosinophils and poor outcome. Results Multivariate analysis revealed an independent association between eosinophil count and sICH after adjusting for potential confounders (odds ratio, 0.00; 95% CI, 0.00–0.01; P = 0.0141), which is consistent with the result of eosinophil count (dichotomous) as a categorical variable (odds ratio, 0.22; 95% CI, 0.11–0.46; P < 0.0001). Eosinophil count was negatively associated with poor outcome (odds ratio, 0.00; 95% CI, 0.00–0.02; P = 0.0021). Mediation analysis revealed that sICH partially mediated the negative relationship between eosinophil count and poor outcome (indirect effect=-0.1896; 95%CI: -0.3654 – -0.03, P < 0.001). Conclusion This study showed an important effect of sICH on the association between eosinophils and poor outcome.
Article
Full-text available
Cytokines and growth factors contribute to nerve growth and angiogenesis and are associated with the development of vascular disease. This Mendelian randomization (MR) study was designed to examine the causal relationship between factors associated with stem cell paracrine mechanisms and with stroke and its subtypes. We used pooled statistics on cytokine levels from three studies (INTERIAL, Olink Proseek CVD array, and KORA) encompassing 7795 participants in Europe. Data for stroke and its subtypes were pooled from these European populations (40,585 cases and 406,111 controls) in a multiprogenitor genome-wide association study (GWAS). MR was performed using established analytical methods, including inverse variance weighting (IVW), weighted median (WM), and MR-Egger. Genetically determined high IGF-1 levels were found to associate negatively with risk of stroke, ischemic stroke (large-artery atherosclerosis), and ischemic stroke (cardiogenic embolism). Meanwhile, high IL-13 levels had a positive causal relationship with ischemic stroke (large-artery atherosclerosis). An additional 27 cytokines were found to have a causal association with stroke or its subtypes. However, these results should be interpreted with caution given that the power efficacy was <80%. This MR study supports the concept of a causal relationship of 29 cytokines with stroke or its subtypes. Our genetic analysis provides new insights into stroke prevention and treatment by demonstrating an association of stem cell paracrine-related cytokines with stroke risk.
Article
OBJECTIVE Although tirofiban and endovascular thrombectomy have been widely used in the treatment of acute ischemic stroke (AIS) patients, the effectiveness of their combined application remains a subject of debate. This study aimed to assess the efficacy and safety of tirofiban in direct thrombectomy for AIS with anterior circulation vessel occlusion. METHODS A total of 204 patients undergoing direct thrombectomy between January 2020 and December 2021 for AIS with anterior circulation vessel occlusion from four hospitals were included in this study. Patients at high risk of reocclusion with severe atherosclerosis, those who achieved successful recanalization for ≥ 3 stent retriever passes, or those who underwent emergency stenting or balloon angioplasty for severe residual stenosis were treated with tirofiban. Following a low-dose intra-arterial bolus (0.25–1 mg) immediately after endovascular treatment, tirofiban was administered continuously through intravenous infusion (0.1 μg/kg/min) for 12–24 hours. The primary efficacy outcome was evaluated using the 90-day modified Rankin Scale score. The safety outcome was assessed using symptomatic intracerebral hemorrhage (sICH) and mortality rates. RESULTS The tirofiban group and nontirofiban group each included 102 patients. The favorable outcome rate in the tirofiban group was significantly higher than that in the nontirofiban group (53.9% vs 35.3%, p = 0.007). However, the sICH and 90-day mortality rates were lower in the tirofiban group, despite a lack of statistical significance (sICH: 15.7% vs 16.7%, p = 0.849; 90-day mortality: 16.67% vs 24.51%, p = 0.166). Finally, it was found that older patients (> 72 years), male patients, patients with admission National Institutes of Health Stroke Scale scores > 14, patients with a time from onset to reperfusion > 327 minutes, and patients with a medical history of diabetes tend to benefit from tirofiban treatment. CONCLUSIONS This study suggests that tirofiban combined with direct thrombectomy improves functional outcomes of AIS and reduces the 90-day mortality rate. Therefore, it could be considered as a suitable treatment option for AIS patients with anterior circulation vessel occlusion.
Article
Full-text available
Prenatal alcohol exposure (PAE) impairs recovery from cerebrovascular ischemic stroke in adult rodents. Since the gut becomes dysbiotic following stroke, we assessed links between PAE and enteric portal inflammation. Adult control and PAE rat offspring received a unilateral endothelin-1-induced occlusion of the middle cerebral artery. Post-stroke behavioral disabilities and brain cytokines were assessed. Mesenteric adipose and liver transcriptomes were assessed from stroke-exposed and stroke-naive offspring. We identified, in the liver of stroke-naive animals, a moderate correlation between PAE and a gene network for inflammatory necroptosis. PAE inhibited the acute-phase brain inflammatory cytokine response to stroke. Post-stroke neurological function was correlated with an adipose gene network associated with B-lymphocyte differentiation and nuclear factor κB (NF-κB) signaling and with a liver pro-inflammatory gene network. Collectively, PAE inhibits brain inflammation but results in an inflammatory signature in enteric portal tissues after stroke, suggesting that PAE persistently and adversely impacts the gut-brain axis following adult-onset disease.
Article
Full-text available
Stem memory T cells (Tscm) constitute the earliest developmental stage of memory T cells, displaying stem cell-like properties such as self-renewal capacity. Their superior immune reconstitution potential has sparked interest in cancer immune-therapy, vaccine development and immune reconstitution, whereas their role in autoimmunity is largely unexplored. Here we show that autoreactive CD8+ Tscm specific for β cell antigens GAD65, insulin and IGRP are present in patients with type 1 diabetes (T1D).In vitro,generation of autoreactive Tscm from naïve precursors required the presence of the homeostatic cytokine interleukin-7 (IL-7). IL-7 promotes glucose uptake via overexpression of the glucose transporter GLUT-1 and up-regulation of the glycolytic enzyme hexokinase II. Even though metabolism depends on glucose uptake, the subsequent oxidation of pyruvate in the mitochondria was necessary for Tscm generation from naïve precursors. In patients with T1D, high expression of GLUT1 was a hallmark of circulating Tscm and targeting glucose uptake via GLUT-1 using the selective inhibitor WZB117 resulted in inhibition of Tscm generation and expansion. Our results suggest that autoreactive Tscm are present in patients with T1D and can be selectively targeted by inhibition of glucose metabolism.
Article
Full-text available
Background: Monitoring levels and trends in premature mortality is crucial to understanding how societies can address prominent sources of early death. The Global Burden of Disease 2016 Study (GBD 2016) provides a comprehensive assessment of cause-specific mortality for 264 causes in 195 locations from 1980 to 2016. This assessment includes evaluation of the expected epidemiological transition with changes in development and where local patterns deviate from these trends. Methods: We estimated cause-specific deaths and years of life lost (YLLs) by age, sex, geography, and year. YLLs were calculated from the sum of each death multiplied by the standard life expectancy at each age. We used the GBD cause of death database composed of: vital registration (VR) data corrected for under-registration and garbage coding; national and subnational verbal autopsy (VA) studies corrected for garbage coding; and other sources including surveys and surveillance systems for specific causes such as maternal mortality. To facilitate assessment of quality, we reported on the fraction of deaths assigned to GBD Level 1 or Level 2 causes that cannot be underlying causes of death (major garbage codes) by location and year. Based on completeness, garbage coding, cause list detail, and time periods covered, we provided an overall data quality rating for each location with scores ranging from 0 stars (worst) to 5 stars (best). We used robust statistical methods including the Cause of Death Ensemble model (CODEm) to generate estimates for each location, year, age, and sex. We assessed observed and expected levels and trends of cause-specific deaths in relation to the Socio-demographic Index (SDI), a summary indicator derived from measures of average income per capita, educational attainment, and total fertility, with locations grouped into quintiles by SDI. Relative to GBD 2015, we expanded the GBD cause hierarchy by 18 causes of death for GBD 2016. Findings: The quality of available data varied by location. Data quality in 25 countries rated in the highest category (5 stars), while 48, 30, 21, and 44 countries were rated at each of the succeeding data quality levels. Vital registration or verbal autopsy data were not available in 27 countries, resulting in the assignment of a zero value for data quality. Deaths from non-communicable diseases (NCDs) represented 72·3% (95% uncertainty interval [UI] 71·2–73·2) of deaths in 2016 with 19·3% (18·5–20·4) of deaths in that year occurring from communicable, maternal, neonatal, and nutritional (CMNN) diseases and a further 8·43% (8·00–8·67) from injuries. Although age-standardised rates of death from NCDs decreased globally between 2006 and 2016, total numbers of these deaths increased; both numbers and age-standardised rates of death from CMNN causes decreased in the decade 2006–16—age-standardised rates of deaths from injuries decreased but total numbers varied little. In 2016, the three leading global causes of death in children under-5 were lower respiratory infections, neonatal preterm birth complications, and neonatal encephalopathy due to birth asphyxia and trauma, combined resulting in 1·80 million deaths (95% UI 1·59 million to 1·89 million). Between 1990 and 2016, a profound shift toward deaths at older ages occurred with a 178% (95% UI 176–181) increase in deaths in ages 90–94 years and a 210% (208–212) increase in deaths older than age 95 years. The ten leading causes by rates of age-standardised YLL significantly decreased from 2006 to 2016 (median annualised rate of change was a decrease of 2·89%); the median annualised rate of change for all other causes was lower (a decrease of 1·59%) during the same interval. Globally, the five leading causes of total YLLs in 2016 were cardiovascular diseases; diarrhoea, lower respiratory infections, and other common infectious diseases; neoplasms; neonatal disorders; and HIV/AIDS and tuberculosis. At a finer level of disaggregation within cause groupings, the ten leading causes of total YLLs in 2016 were ischaemic heart disease, cerebrovascular disease, lower respiratory infections, diarrhoeal diseases, road injuries, malaria, neonatal preterm birth complications, HIV/AIDS, chronic obstructive pulmonary disease, and neonatal encephalopathy due to birth asphyxia and trauma. Ischaemic heart disease was the leading cause of total YLLs in 113 countries for men and 97 countries for women. Comparisons of observed levels of YLLs by countries, relative to the level of YLLs expected on the basis of SDI alone, highlighted distinct regional patterns including the greater than expected level of YLLs from malaria and from HIV/AIDS across sub-Saharan Africa; diabetes mellitus, especially in Oceania; interpersonal violence, notably within Latin America and the Caribbean; and cardiomyopathy and myocarditis, particularly in eastern and central Europe. The level of YLLs from ischaemic heart disease was less than expected in 117 of 195 locations. Other leading causes of YLLs for which YLLs were notably lower than expected included neonatal preterm birth complications in many locations in both south Asia and southeast Asia, and cerebrovascular disease in western Europe. Interpretation: The past 37 years have featured declining rates of communicable, maternal, neonatal, and nutritional diseases across all quintiles of SDI, with faster than expected gains for many locations relative to their SDI. A global shift towards deaths at older ages suggests success in reducing many causes of early death. YLLs have increased globally for causes such as diabetes mellitus or some neoplasms, and in some locations for causes such as drug use disorders, and conflict and terrorism. Increasing levels of YLLs might reflect outcomes from conditions that required high levels of care but for which effective treatments remain elusive, potentially increasing costs to health systems.
Article
Full-text available
Erythropoietin (EPO) promotes oligodendrogenesis and attenuates white matter injury in neonatal rats. However, it is unknown whether this effect extends to adult mice and whether EPO regulate microglia polarization after ischemic stroke. Male adult C57BL/6 mice (25–30g) were subjected to 45 min of middle cerebral artery occlusion (MCAO). EPO (5000 IU/kg) or saline was injected intraperitoneally every other day after reperfusion. Neurological function was evaluated using the rotarod test at 1, 3, 7 and 14 days after MCAO. Brain tissue loss volume was determined by hematoxylin-eosin staining. Immunofluorescence staining and Western blot were also used to assess the severity of white matter injury and phenotypic changes in microglia/macrophages. Bromodeoxyuridine (BrdU) was injected intraperitoneally daily for 1 week to analyze the number of newly proliferating glia cells (oligodendrocytes, microglia, and astrocytes). We found that EPO significantly reduced Brain tissue loss volume, ameliorated white matter injury, and improved neurobehavioral outcomes at 14 days after MCAO (P<0.05). In addition, EPO also increased the number of newly generated oligodendrocytes and attenuated the rapid hypertrophy and hyperplasia of microglia and astrocytes after ischemic stroke (P<0.05). Furthermore, EPO reduced M1 microglia and increased M2 microglia (P<0.05). Taken together, our results suggest that EPO treatment improves white matter integrity after cerebral ischemia, which could be attributed to EPO attenuating gliosis and facilitating the microglial polarization toward the beneficial M2 phenotype to promote oligodendrogenesis.
Article
Full-text available
Each year, the American Heart Association (AHA), in conjunction with the Centers for Disease Control and Prevention, the National Institutes of Health, and other government agencies, brings together in a single document the most upto-date statistics related to heart disease, stroke, and the factors in the AHA's Life's Simple 7 (Figure1), which include core health behaviors (smoking, physical activity [PA], diet, and weight) and health factors (cholesterol, blood pressure [BP], and glucose control) that contribute to cardiovascular health. The Statistical Update represents a critical resource for the lay public, policy makers, media professionals, clinicians, healthcare administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions. Cardiovascular disease (CVD) and stroke produce immense health and economic burdens in the United States and globally. The Update also presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure (HF), valvular disease, venous disease, and peripheral arterial disease) and the associated outcomes (including quality of care, procedures, and economic costs). Since 2006, the annual versions of the Statistical Update have been cited >20000 times in the literature. In 2015 alone, the various Statistical Updates were cited '4000 times.
Article
Full-text available
Background MicroRNA miR-155 is implicated in modulation of the inflammatory processes in various pathological conditions. In our previous studies, we demonstrated that in vivo inhibition of miR-155 promotes functional recovery after mouse experimental stroke. In the present study, we explored if this beneficial effect is associated with miR-155 inhibition-induced alterations in post-stroke inflammatory response. Methods Intravenous injections of a specific miR-155 inhibitor were initiated at 48 h after mouse distal middle cerebral artery occlusion (dMCAO). Temporal changes in the expression of cytokines and key molecules associated with cytokine signaling were assessed at 7, 14, and 21 days after dMCAO, using mouse cytokine gene and protein arrays and Western blot analyses. Electron and immunofluorescence confocal microscopy techniques were used to evaluate the ultrastructural changes, as well as altered expression of specific phenotypic markers, at different time points after dMCAO. ResultsIn the inhibitor-injected mice (inhibitor group), there was a significant decrease in CCL12 and CXCL3 cytokine expression at 7 days and significantly increased levels of major cytokines IL-10, IL-4, IL-6, MIP-1α, IL-5, and IL-17 at 14 days after dMCAO. These temporal changes correlated with altered expression of miR-155 target proteins SOCS-1, SHIP-1, and C/EBP-β and phosphorylation levels of cytokine signaling regulator STAT-3. Electron microscopy showed decreased number of phagocytically active peri-vascular microglia/macrophages in the inhibitor samples. Immunofluorescence and Western blot of these samples demonstrated that expression of leukocyte/ macrophage marker CD45 and phagocytosis marker CD68 was reduced at 7 days, and in contrast, significantly increased at 14 days after dMCAO, as compared to controls. Conclusions Based on our findings, we propose that in vivo miR-155 inhibition following mouse stroke significantly alters the time course of the expression of major cytokines and inflammation-associated molecules, which could influence inflammation process and tissue repair after experimental cerebral ischemia.
Article
Ischemic stroke (IS) is a disease caused by deficiency of blood and oxygen in focal or complete brain, followed by inflammation cascade and other pathological reactions, which finally lead to irreversible damage to the cerebrum. For the inflammation is a key progress at the initiation of ischemia and poststroke, and chemokines work as vital cytokines in inflammation, we focus the roles of chemokines in IS. Studies have shown cerebral ischemia is associated with marked induction of both CXC and CC chemokines which resulting in extensive leukocyte infiltration in the ischemic brain, and neutrophil infiltration may increase cerebral edema inducing injury in the ischemic area. In addition, chemokines also shows other functions such as promote neuroblast migration, hematogenous cell recruitment and functional brain repair. Thus, a similar chemokine ligand/chemokine receptor pair can mediate both beneficial and detrimental effects depending on the window of observation and pathophysiological conditions. This manuscript reviews the studies about chemokine-mediated effects in cerebral ischemia/reperfusion and discusses the potential significance of these interactions in injury and repair of ischemic tissues. We also refer drug development based on the chemokines and clinical applications using chemokines as diagnostic or prognostic biomarkers in ischemic stroke.
Article
Interleukin (IL) -9 exerts a variety of functions in autoimmune diseases. However, its role in ischemic brain injury remains unknown. The present study explored the biological effects of IL-9 in ischemic stroke. We recruited 42 patients newly diagnosed with ischemic stroke and 22 age- and sex-matched healthy controls. The expression levels of IL-9 and percentages of IL-9-producing T cells, including CD3+CD4+IL-9+ and CD3+CD8+IL-9+ cells, were determined in peripheral blood mononuclear cells (PBMCs) obtained from patients and control individuals. We also investigated the effects of IL-9 on the blood brain barrier (BBB) following oxygen-glucose deprivation and the potential downstream signaling pathways. We found that patients with ischemic stroke had higher IL-9 expression levels and increased percentages of IL-9-producing T cells in their PBMCs. The percentages of CD3+CD4+IL-9+ and CD3+CD8+IL-9+ T cells were positively correlated with the severity of illness. In invitro experiments using bEnd.3 cells, exogenously administered IL-9 exacerbated the loss of tight junction proteins in cells subjected to oxygen-glucose deprivation plus reoxygenation. This effect was mediated via activation of IL-9 receptors, which increased the level of endothelial nitric oxide synthase (eNOS), as well as through upregulated phosphorylation of signal transducer and activator of transcription 1 and 3 and downregulated phosphorylated protein kinase B/phosphorylated phosphatidylinositol 3-kinase signaling. These results indicate that IL-9 has a destructive effect on the BBB following oxygen-glucose deprivation, at least in part by inducing eNOs production, and raise the possibility of targeting IL-9 for therapeutic intervention in ischemic stroke.
Article
Background and purpose: The Gugging Swallowing Screen (GUSS) is a tool to screen aspiration risk in acute stroke. We aimed to replicate its validity in a larger second cohort of patients with acute stroke, including the more severe with a National Institutes of Health Stroke Scale (NIHSS) ≥ 15. Methods: In a prospective, double-blind design, the GUSS was validated with the Fiberoptic Endoscopic Evaluation of Swallowing scale. Patients were categorized into different stroke severities as assessed by the NIHSS, and the diagnostic properties were calculated separately for each subgroup. Results: A total of 100 patients with acute stroke were evaluated consecutively at a mean 1.7 ± 2.2 days after stroke. With the GUSS cut-off value of 14 points, the GUSS screened aspiration risk with a 96.5% sensitivity and 55.8% specificity (area under the curve, 0.76; 95% CI, 0.67-0.84), which corresponded well with the original publication. In the NIHSS < 5 group, the sensitivity and specificity levels were 71.4% and 88.8%, respectively. In the NIHSS ≥ 15 group, these levels changed to 100% and 20%, respectively. The high failure rate in completing the first part of the GUSS in the latter group was related to the low specificity. Diet recommendations following the GUSS were more conservative than those after Fiberoptic Endoscopic Evaluation of Swallowing. In particular, the GUSS overestimated the need for nasogastric tube feeding. Conclusions: This is the first time that a swallowing screening tool for patients with acute stroke has been revalidated in a larger population from another stroke center. The validity of a swallow screening test may vary according to different stroke severities.