ArticlePDF Available

Abstract and Figures

Post-ischemic inflammation is an essential step in the progression of brain ischemia-reperfusion injury. In this review, we focus on the post-ischemic inflammation triggered by infiltrating immune cells, macrophages, and T lymphocytes. Brain ischemia is a sterile organ, but injury-induced inflammation is mostly dependent on Toll-like receptor (TLR) 2 and TLR4. Some endogenous TLR ligands, high mobility group box 1 (HMGB1) and peroxiredoxin family proteins, in particular, are implicated in the activation and inflammatory cytokine expression in infiltrating macrophages. Following macrophage activation, T lymphocytes infiltrate the ischemic brain and regulate the delayed phase inflammation. IL-17-producing γδT lymphocytes induced by IL-23 from macrophages promote ischemic brain injury, whereas regulatory T lymphocytes suppress the function of inflammatory mediators. A deeper understanding of the inflammatory mechanisms of infiltrating immune cells may lead to the development of novel neuroprotective therapies.
This content is subject to copyright.
REVIEW ARTICLE
published: 31 May 2012
doi: 10.3389/fimmu.2012.00132
Post-ischemic inflammation in the brain
Takashi Shichita1,2 , Ryota Sakaguchi 1,3, Mayu Suzuki 1,3 and AkihikoYoshimura1,3 *
1Department of Microbiology and Immunology, School of Medicine, Keio University,Tokyo, Japan
2Department of Research Promotion, Precursory Research for Embryonic Science and Technology, Tokyo, Japan
3Core Research for Evolutional Science and Technology, Japan Science andTechnology Agency,Tokyo, Japan
Edited by:
Masaaki Murakami, Osaka University,
Japan
Reviewed by:
Daisuke Kamimura, Osaka University,
Japan
Yasunobu Arima, Osaka University,
Japan
*Correspondence:
Akihiko Yoshimura, Department of
Microbiology and Immunology,
School of Medicine, Keio University,
35 Shinanomachi, Shinjuku-ku,Tokyo
160-8582, Japan.
e-mail: yoshimura@a6.keio.jp
Post-ischemic inflammation is an essential step in the progression of brain
ischemia-reperfusion injury. In this review, we focus on the post-ischemic inflammation
triggered by infiltrating immune cells, macrophages, and T lymphocytes. Brain ischemia is
a sterile organ, but injury-induced inflammation is mostly dependent on Toll-like receptor
(TLR) 2 andTLR4. Some endogenousTLR ligands, high mobility group box 1 (HMGB1) and
peroxiredoxin family proteins, in particular, are implicated in the activation and inflamma-
tory cytokine expression in infiltrating macrophages. Following macrophage activation, T
lymphocytes infiltrate the ischemic brain and regulate the delayed phase inflammation. IL-
17-producing γδT lymphocytes induced by IL-23 from macrophages promote ischemic brain
injury, whereas regulatory T lymphocytes suppress the function of inflammatory media-
tors. A deeper understanding of the inflammatory mechanisms of infiltrating immune cells
may lead to the development of novel neuroprotective therapies.
Keywords: cytokine, inflammation, ischemia, brain, stroke,T cells, macrophages, DAMPs
INTRODUCTION
Stroke is a leading cause of death and disability worldwide. The
most common type of stroke is ischemic stroke (e.g., approxi-
mately 70% of strokes in Japan are ischemic). However, intra-
venous administration of tissue plasminogen activator (tPA) is
the only globally approved treatment for ischemic stroke, and it
is a time-dependent therapy that must be provided within 4.5 h
after stroke onset. Thus, there is a need for an efficacious therapy
that can be administered beyond this time window,one that targets
neuroprotection rather than clot dissolution (Lo, 2010;Moskowitz
et al., 2010).
Brain infarction is tissue death caused by ischemia due to
severe stenosis or occlusion of a cerebral artery. Ischemic brain
tissue is deprived of oxygen, glucose, and lipids, and eventually
becomes necrotic. Brain inflammation occurs in this necrotic
brain tissue, following the breakdown of the blood-brain barrier
(BBB) and infiltration of blood immune cells. Infiltrating immune
cells promote ischemic brain inflammation by producing vari-
ous inflammatory mediators, and also clear away necrotic debris.
After the demolition of necrotic debris has been completed, brain
inflammation subsides.
Despite intensive studies, the complexity of the brain inflam-
mation mechanism has thus far prevented sufficient clarification
(Eltzschig and Eckle, 2011;Iadecola and Anrather, 2011;Macrez
et al., 2011). Macrophages and neutrophils are pivotal players in
the various processes of brain inflammation, but the mechanism of
their activation is still unknown. In addition, T or B lymphocytes
have been also reported to participate in delayed brain inflam-
mation. This review focuses on the mechanism of ischemic brain
inflammation triggered and sustained by infiltrating immune cells.
POST-ISCHEMIC INFLAMMATION IN THE EARLY PHASE OF
BRAIN ISCHEMIA
Severe ischemia induces hypoxia and glucose deprivation in brain
tissue. Calcium and sodium ions are stored within brain cells and
glutamate is released into the extracellular compartment. The pro-
duction of reactive oxygen species (ROS) activates platelets and
endothelial cells, leading to microvascular occlusion. Oxidative
stress reduces the beneficial effects of nitric oxide (NO), a potent
vasodilator and inhibitor of platelet aggregation and leukocyte
adhesion, in endothelial cells. Oxidative stress and the inflam-
matory cascade alter the permeability of the BBB. The activation
of matrix metalloproteinases (MMPs) and the expression of var-
ious other proteases lead to BBB breakdown which exacerbates
leukocyte extravasation. Intravascular leukocytes firmly adhere to
activated endothelium by the interaction of endothelial expres-
sion of intercellular adhesion molecule-1 (ICAM-1) and leukocyte
β2 integrins (Iadecola and Anrather, 2011). The infiltration of
leukocytes is enhanced by BBB breakdown and by chemokines.
Brain cells, including astrocytes, oligodendrocytes, endothe-
lium, and pericytes, constitute a neurovascular network, which
is essential for metabolic requirement of neurons (Iadecola, 2004;
Fraser, 2011). These brain cells also contribute to triggering post-
ischemic inflammation by producing inflammatory mediators.
TNF-α,IL-1β, NOS (nitric oxide synthetase), and MMPs which
enhance cerebrovascular permeability and exaggerate brain edema
(Takano et al., 2009;Morancho et al., 2010). Thus, Infiltrat-
ing leukocytes and injured brain cells produce various inflam-
matory mediators, leading to the beginning of post-ischemic
inflammation (Barone and Feuerstein, 1999;Figure 1).
INNATE INFLAMMATORY CYTOKINES AND INFLAMMATORY
MEDIATORS
Various cytokines and mediators are produced from infiltrating
immune cells and brain cells as a result of ischemic changes of
brain. IL-1βis expressed in ischemic brain tissue within 30 min
after stroke onset. IL-1βdirectly induces apoptosis of neuronal
cells and enhances the expression of chemokine (RANTES, etc.)
in microglia and astrocytes. IL-1βis considered to be a neurotoxic
mediator,given that the loss of IL-1βfunction is reported to reduce
www.frontiersin.org May 2012 | Volume 3 | Article 132 | 1
Shichita et al. Mechanisms of cerebral post-ischemic inflammation
FIGURE1|Post-ischemic inflammation in the brain.Within 24h after
ischemic stroke onset, various inflammatory mediators are expressed in
ischemic brain tissue. ICAM-1 promotes leukocytes infiltration. Cytokines
activate infiltrating leukocytes and directly induce ischemic injury in brain
cells. Matrix metalloproteinases (MMPs) alter the permeability of epithelial
cells and are implicated in BBB breakdown. Endogenous TLR ligands
(DAMPs) are released from necrotic brain cells and activate infiltrating
immune cells. These inflammatory mediators trigger post-ischemic
inflammation by infiltrating leukocytes.There are currently few effective
therapies for this phase of leukocyte infiltration.
infarct size (Boutin et al., 2001). IL-1βis produced in an inactivate
form, pro-IL-1β, which is cleaved by caspase-1 to become an active
17 kDa form. Recently, the mechanism of IL-1βproduction and
caspase-1 activation mediated by inflammasome has been the sub-
ject of particular attention. Inflammasome has been shown to be
present in neurons, astrocytes, microglia, and macrophages in the
ischemic brain (Abulafia et al., 2009;Chakraborty et al., 2010).
Several types of inflammasome have been discovered, denoted
NALP1, NALP3, AIM, and so on, but the particular type most
important in ischemic brain injury remains unknown. Hypoxia
or ATP is reported to activate inflammasome,which then activates
caspase-1 and induces IL-1βproduction (Martinon et al., 2002).
In addition, IL-1βis mostly produced from monocytes which are
activated by endogenous Toll-like receptor (TLR) ligands, given
that IL-1βmRNA level in infiltrating mononuclear cells is drasti-
cally reduced in TLR2/4-double deficient mice after ischemic brain
injury.
TNF-αis another important mediator implicated in the pathol-
ogy of the ischemic brain. TNF-αis expressed in ischemic brain
tissue within 1 h after stroke onset, and upregulation of the TNF
receptors is observed thereafter. TNF-αexercises neurotoxic effects
by inducing apoptotic neuronal cell death and enhancing MHC
class II and ICAM-1 expression in astrocytes, leading to leukocyte
infiltration and BBB breakdown. TNF-αgene knockout (KO) mice
or anti-TNF-αneutralizing antibody administration has been
shown to reduce infarct volume, compared with that in control
mice. However, TNFR KO mice, which lack both p75 and p50
genes, exhibit enlargement of infarct volume on day 1 following
ischemic brain injury, indicating that TNF-αcan be considered
to function as both a neurotoxic and a neuroprotective mediator
(Hallenbeck, 2002). It appears that the opposing functions, toxic
or protective, depends on the type of brain cell involved. TNF-α
promotes post-ischemic inflammation but also participates in a
negative feedback loop to suppress inflammatory signal cascades,
and it controls the duration of post-ischemic inflammation by
regulating these two functions.
IL-6 is also expressed in ischemic brain tissue, but ischemic
brain damage is not attenuated in IL-6 KO mice or in anti-IL-
6R antagonistic antibody-treated mice (Yamashita et al., 2005).
However, it has been recently reported that IL-6 produced from
brain cells contributes to neoangiogenesis and neuronal sur-
vival through STAT3 activation (Jung et al., 2011;Gertz et al.,
2012). Consistent with this, the inhibition of JAK/STAT path-
way or the enhanced role of SOCS3 (negative regulator of
JAK/STAT pathway) has been reported to promote neuronal
cell death (Yadav et al., 2005;Yamashita et al., 2005). Thus, it
is possible that IL-6 protects neuron from cell death, although
a significant role of IL-6 in brain ischemia has not yet been
established.
Matrix metalloproteinases are essential neurotoxic mediators
that promote BBB breakdown and post-ischemic inflammation.
Functionally similar to IL-1β, MMPs induce apoptotic neuronal
cell death by TNF-αand FasL processing. The neurotoxic func-
tion of MMP-9 is particularly established, given that infarct size
is reduced in MMP-9-deficient mice compared to that in con-
trol mice (Asahi et al., 2000). Intercellular adhesion molecule-1
(ICAM-1) is another neurotoxic mediator. The increased expres-
sion of ICAM-1 observed in cerebrovascular endothelial cells is
implicated in the promotion of leukocyte infiltration. ICAM-
1-deficient mice reveal attenuated ischemic damage and the
administration of anti-ICAM-1 antibody decreases the number
of infiltrating immune cells (Connolly et al., 1996;Liesz et al.,
2011).
Chemokines are also important enhancers of post-ischemic
inflammation. Chemokines (RANTES, MCP-1, IL-8, etc.) have
been reported to promote leukocyte infiltration into the ischemic
brain (Terao et al., 2008, 2009;Strecker et al., 2011). Although
the chemokines for lymphocyte infiltration remain unknown,
CCL12, CCL20, and their receptor,CCR6, are essential for the exac-
erbation of experimental autoimmune encephalomyelitis (EAE;
Martin et al., 2009;Reboldi et al., 2009). Whether these lympho-
cyte chemokines also function in acute organ injury, such as brain
ischemia, should be elucidated in the future.
Sphingosine-1-phosphate (S1P) is a bioactive phospholipid.
At sites of tissue injury, S1P is mainly released from platelets
and mediates its effect via activation of cell-surface S1P recep-
tors, which are ubiquitously expressed in brain cells (Dev et al.,
2008). S1P receptors are also present on the surface of T lym-
phocytes; therefore, S1P has been thought to play an essential
role in T lymphocyte infiltration of inflammatory tissue. Recently,
the therapeutic effect of FTY720 (fingolimod), a functional S1P
receptor antagonist, has attracted attention. The administration of
FTY720 has been shown to attenuate ischemic brain damage and
decrease the number of infiltrating T lymphocytes in the ischemic
brain (Shichita et al., 2009;Hasegawa et al., 2010). Furthermore,
S1P receptors are expressed in neurons,astrocytes, and microglial
cells. S1P acts on these cells directly and exerts effects that include
astrocyte proliferation and migration, oligodendrocyte differenti-
ation and cell survival, and neurite outgrowth and neurogenesis
Frontiers in Immunology | Inflammation May 2012 | Volume 3 | Article 132 | 2
Shichita et al. Mechanisms of cerebral post-ischemic inflammation
(Dev et al., 2008). Thus, S1P is considered to be an important
inflammatory mediator in the ischemic brain.
ROLE OF TLR
Leukocyte infiltration is an essential step in the progression of
post-ischemic inflammation. However, the mechanisms that acti-
vate these infiltrating immune cells, macrophages, and lympho-
cytes, are not yet fully clarified. TLRs are an essential type of
receptor for innate and non-specific immune response to general
pathogens such as bacteria, viruses, and so on. The demonstration
that TLR2 and TLR4 are pivotal for sterile organ injury,including
ischemic brain injury, has recently attracted attention (Chen et al.,
2007;Tang et al., 2007).
Toll-like receptors are expressed on both leukocytes and brain
cells, although whether the effect of TLRs on brain cells is
neurotoxic or neuroprotective remains unclear. TLR stimulation
in macrophages and lymphocytes induces strong and various
inflammatory responses. In ischemic brain injury, post-ischemic
inflammation and subsequent ischemic damage depend on TLR2
and TLR4, but not TLR9 (Tang et al., 2007;Hyakkoku et al.,
2010). TLR2- or TLR4-deficient mice demonstrate both signifi-
cant reduction of infarct volume and suppression of neurotoxic
inflammatory responses. Analysis of bone marrow chimeric mice
indicates that TLRs in infiltrated immune cells, but not in residen-
tial microglia, have a neurotoxic effect on ischemic brain injury
(Yang et al., 2011). In addition, mice lacking MyD88, the adaptor
protein under almost all TLR signaling cascades other than TLR3,
are reported to show no improvement of ischemic brain injury
(Famakin et al., 2011). These results indicate that the function of
TLRs may be dependent on kind of cells in brain.
Toll-like receptor-2 or TLR4 deficiency suppresses inflamma-
tory cytokine expression in infiltrating immune cells on day
1 after brain ischemia (Shichita et al., 2012). Although both
TLR2 and TLR4 signaling cascades are essential triggers for post-
ischemic inflammation, and activators of infiltrating immune
cells, the particular molecules that activate TLR2 and TLR4 in
the ischemic brain remain unclear. Because the brain is a sterile
organ, pathogens derived from bacteria or viruses are completely
lacking in the normal and ischemic brain. This indicates that
certain endogenous molecules released from necrotic brain cells
become TLR stimulators, and several endogenous molecules have
indeed been reported to activate TLR signaling cascades. Such
endogenous molecules are called danger associated molecular pat-
terns (DAMPs), and are considered to be danger signals or alarm
molecules that warn immune cells of tissue and cellular injury.
DAMPs IN INJURED BRAIN
Several molecules in the brain have been reported as DAMPs.
Heat shock proteins (HSPs),β-amyloid (Aβ), hyaluronan, heparin
sulfate, DNA or RNA immune complex, oxidized low-density
lipoproteins (oxLDL),and others, can stimulate TLRs (Marsh et al.,
2009;Rivest, 2009;Yanai et al., 2009;Stewart et al., 2010;Zhang
et al., 2010). However, it remains unclear which molecule is the
most important for triggering post-ischemic inflammation and
inflammatory cytokine expression.
High mobility group box 1 (HMGB1) is a well-elucidated
DAMP and is also implicated in ischemic brain injury (Kim et al.,
2006;Liu et al., 2007;Hayakawa et al., 2010). HMGB1 increases
vascular permeability and promotes BBB breakdown (Zhang et al.,
2011). HMGB1, which is localized in cell nuclei in the normal
brain, translocates into the cytosolic compartment and is released
into the extracellular compartment in the ischemic condition.
The administration of anti-HMGB1-neutralizing antibody pro-
tects the BBB and reduces infarct volume. Thus, HMGB1 is an
essential DAMP in ischemic brain injury. Extracellular release of
HMGB1 is observed within 6 h after stroke onset, but is dimin-
ished by 12 h after the onset (Qiu et al., 2008;Zhang et al., 2011).
Thereafter, the infiltration of immune cells and the production
of inflammatory cytokines become evident. This indicates that
HMGB1 may not directly activate infiltrating immune cells in the
ischemic brain.
We have identified the peroxiredoxin (Prx) family proteins as
strong inducers of inflammatory cytokines in infiltrating immune
cells (Shichita et al., 2012). Prx family proteins are known to
exert a protective effect by catalyzing ROS. In the ischemic brain,
the expression of Prx within brain cells is increased by ischemic
stress, and such intracellular Prx is thought to be neuroprotective
(Patenaude et al., 2005;Rashidian et al., 2009;Figure 2). How-
ever, as necrosis occurs, this Prx is released into the extracellular
compartment where they induce inflammatory cytokine expres-
sion in infiltrating immune cells by stimulating TLR2 and TLR4
(Figure 2). It is interesting to note that the Prx family proteins
have a common active region for TLR activation and are extracel-
lularly released over 12h after stroke onset, which coincides with
the timing of leukocyte infiltration. Thus, Prx has two opposing
functions, one inside, and one outside, brain cells. Furthermore,
there is a time lag as well as functional differences between HMGB1
and Prx (Figure 3).
T LYMPHOCYTES IN ISCHEMIC BRAIN INJURY
It has been recently suggested that T lymphocytes play a role as
mediators in the delayed phase of brain ischemia (Yilmaz et al.,
2006). The number of infiltrating T lymphocytes in ischemic brain
tissue increases over 24h after stroke onset and reaches its peak
in the delayed phase (around day 3; Schroeter et al., 1994;Jan-
der et al., 1995). Infiltrating T lymphocytes appear to be localized
FIGURE 2 |Two opposing functions of Prx, one inside, and one outside,
brain cells. Ischemic stress increases Prx expression within brain cells,
which contributes to their survival by catabolizing reactive oxygen species
(ROS). When ischemic phenomena finally result in necrosis, the Prx
released from necrotic brain cells into the extracellular compartment then
functions as a strong TLR2 and TLR4 stimulator (DAMP) for the infiltrating
macrophages in ischemic brain tissue.
www.frontiersin.org May 2012 | Volume 3 | Article 132 | 3
Shichita et al. Mechanisms of cerebral post-ischemic inflammation
FIGURE3|Post-ischemic inflammation triggered by DAMPs and
infiltrating immune cells. At the hyperacute phase of brain ischemia
(within 6 h after stroke onset), HMGB1 is released from brain cells and
induces BBB breakdown. Following this, blood cells begin to infiltrate into
ischemic brain tissue via disrupted vessels during the acute phase of
ischemia (12–24h after stroke onset). Prx is extracellularly released from
necrotic brain cells and activates infiltrating macrophages via TLR2 and
TLR4. Activated macrophages produce inflammatory cytokines (IL-23, IL-1β,
TNF-α, etc.) which promote ischemic brain injury. At the delayed phase of
brain ischemia (more than 24h after stroke onset), IL-23 induces IL-17
production from γδT lymphocytes, which further enhances ischemic
damage. Thus, HMGB1 is a hyperacute DAMP, while Prx is secondarily
active in post-ischemic inflammation, during the acute phase.
to the infarct boundary zones, typically close to blood vessels.
By Percoll gradient centrifugation, infiltrating immune cells have
been analyzed (Shichita et al., 2009). The number of infiltrating
immune cells reaches the peak at day 3 after stroke onset and most
of these are macrophages. Approximately 1 1.5% of immune
cells are T lymphocytes which are consisted of 30 40% CD4+
helper T lymphocytes, 20 30% γδT lymphocytes, and 2030%
CD8+cytotoxic T lymphocytes. Inflammatory cytokines, IL-1β,
TNF-α, IL-23, and IL-12 have been shown to be produced from
macrophage and play important roles in promoting brain injury.
However, role of various cytokines from T lymphocytes, such as
IFNγand IL-17 in ischemic brain injury has not been clarified.
Although the function of these infiltrating T lymphocytes in
the ischemic brain is not yet clear, T lymphocytes, on the whole,
are considered to act as a neurotoxic effector. This is indicated
by the fact that severe combined immunodeficient (SCID) mice,
and recombination activating gene (RAG)-deficient mice,both of
which lack T and B lymphocytes, reveal significant reduction of
infarct volume (Yilmaz et al., 2006;Hurn et al., 2007). In addi-
tion, the depletion of CD4+(helper) or CD8+(cytotoxic) T
lymphocytes, but not one of the B lymphocytes, is reported to
attenuate ischemic brain damage. There is a report that regulatory
B lymphocytes protect brain from ischemic damages; however, the
neurotoxic effect of lymphocytes is considered to be majorly made
byTlymphocytes(Ren et al., 2011). It will be important to eluci-
date which subtype of lymphocytes is implicated in ischemic brain
injury (see T Lymphocyte Cytokines in Ischemic Brain Injury).
It is not yet clear whether a specific antigen in the brain is
involved in the activation of these infiltrating T lymphocytes.
Up to now, it has been thought likely that these T lymphocytes
mediate antigen-independent, innate inflammatory responses,
because post-ischemic inflammatory responses have been shown
to be driven by the innate immune system. However,some reports
suggest the importance of antigen recognition by T lymphocytes
in ischemic brain injury. Treatment with T cell receptor (TCR) lig-
ands, which are major histocompatibility complex (MHC) class II
molecules bound to myelin peptides, is protective against ischemic
brain injury (Subramanian et al., 2009). Infarct size in myelin basic
protein (MBP) tolerized animals has been shown to be reduced
compared to that in control mice. Thus, there is a possibility that
some T lymphocyte subsets specifically tolerized to brain proteins
could be protective to ischemic brain injury (Becker et al., 2003;
Becker, 2009). This idea is supported by the recent finding that
regulatory T lymphocytes are protective to ischemic brain injury
(Liesz et al., 2009).
T LYMPHOCYTE CYTOKINES IN ISCHEMIC BRAIN INJURY
T lymphocytes are considered to mediate ischemic brain injury
by producing various cytokines, and IFN-γand IL-4 are well-
known classical examples. In ischemic injury, IFN-γis thought
to be neurotoxic, as it acts on neurons directly and induces
apoptotic neuronal cell death in vitro (Lambertsen et al., 2004).
However, a protective effect by the IFN-γdeficiency has not been
observed, and the role of IFN-γin ischemic brain injury is con-
troversial (Lambertsen et al., 2004;Yilmaz et al., 2006). IL-12
is produced from myeloid cells such as macrophages, dendritic
cells, neutrophils, and so on, and is important for the differenti-
ation of IFN-γ-producing helper T lymphocytes (Th1). IL-12 is
expressed in ischemic brain tissue by infiltrating immune cells, but
its function has not been fully elucidated.
IL-4 may have the potential to attenuate ischemic damage and
promote tissue repair, since IL-4-deficient mice demonstrate exac-
erbated ischemic damage and neurological deficit (Xiong et al.,
2011). Although the emerging recognition of the function of IL-4
for tissue repair has recently attracted attention, whether or not
IL-4 is directly implicated in tissue repair in ischemic brain injury
is still unclear (Chen et al., 2012).
IL-10 is an immunosuppressive cytokine and is thought to
have a neuroprotective effect in ischemic brain injury. IL-10 is
produced from regulatory T lymphocytes (Treg) and suppresses
the neurotoxic function of TNF-αand IFN-γ(Liesz et al., 2009).
The overexpression of IL-10 by an adenovirus vector protects hip-
pocampal neurons against apoptotic cell death (Ooboshi et al.,
2006).
IL-17 is an emerging therapeutic target for various organ
injuries. IL-23 has been reported to be essential for IL-17 induc-
tion from T lymphocytes, and to play a critical role in EAE
(Cua et al., 2003). In ischemic brain injury, IL-23 is produced
by infiltrating macrophages on day 1, and it induces IL-17 pro-
duction from γδT lymphocytes in the delayed phase (Figure 3).
Both IL-23 KO mice and IL-17 KO mice show significantly atten-
uated ischemic brain damage on day 4. The IL-17 receptor is
ubiquitously expressed in brain cells and modifies various inflam-
matory responses in the central nervous system. IL-17 has been
reported to promote the expression of inflammatory cytokines
and chemokines from macrophages (Fossiez et al., 1996). IL-17
also modulates the epithelial barrier function by promoting MMPs
Frontiers in Immunology | Inflammation May 2012 | Volume 3 | Article 132 | 4
Shichita et al. Mechanisms of cerebral post-ischemic inflammation
and ICAM-1 expression (Kebir et al., 2007;Ifergan et al., 2008).
Although it remains unknown whether IL-17 directly affects
neurons, IL-17 is thought to be a promising therapeutic tar-
get for suppressing post-ischemic inflammation. Thus, IL-23-
induced IL-17-producing γδT lymphocytes play a pivotal role
in the delayed phase of brain ischemia (Figure 4). IL-17 pro-
duction from γδT lymphocytes requires only IL-1βand IL-23
stimulation, but not specific TCR stimulation,and IL-6 and TGFβ
stimulation are indispensable for IL-17-producing helper T lym-
phocyte (Th17) differentiation (Sutton et al., 2009). Thus, it is
reasonable that γδT lymphocytes mediate ischemic brain injury,
given that γδT lymphocytes produce IL-17 more rapidly than
does Th17.
THE POSSIBILITY OF MEDICAL INTERVENTION IN
POST-ISCHEMIC INFLAMMATION
Post-ischemic inflammation in the brain is considered to have two
opposing functions in stroke patients; one beneficial, the other
harmful. Post-ischemic inflammation promotes brain swelling
(brain edema) which leads to the compression of normal brain
tissue surrounding the ischemic core and the exacerbation of neu-
rological deficits. This undesirable effect of post-ischemic brain
inflammation should be suppressed by medical intervention when
possible. However, post-ischemic inflammation is also thought to
promote tissue repair in the recovery phase of ischemic stroke.
Thus, suppression of all inflammatory responses in ischemic
brain tissue is not always effective, due to their involvement
in both ischemic injury and tissue repair processes. To create
novel neuroprotective strategy, it may be possible to control
the balance between the neurotoxic and neuroprotective effects
of post-ischemic inflammation by targeting specific inflamma-
tory mediators. For example, therapy which both suppresses the
FIGURE4|Schematic model of IL-23/IL-17 inflammatory pathway in
ischemic brain tissue. Infiltrating macrophages produce IL-23 and IL-12,
which induce IL-17-producing γδT lymphocytes and IFN-γ-producing helperT
lymphocytes (Th1), respectively. IL-17 from γδT lymphocytes acts on
macrophages and brain cells directly, and promotes the expression of
inflammatory mediators that enhance apoptotic neuronal cell death and
BBB breakdown.
inflammatory subset of T lymphocytes (e.g., IL-17-producing γδT
lymphocytes) and promotes Treg function may be desirable.
It is possible that administration of γδTCR-depleting anti-
body or anti-IL-12/IL-23p40 antibody [p40 is a common sub-
unit of the IL-12 heterodimer (p35/p40) and IL-23 heterodimer
(p19/p40)] will become a therapeutic tool for ischemic stroke (Shi-
chita et al., 2009;Konoeda et al., 2010). Thus, neuroprotective
therapies for ischemic brain injury may be developed by targeting
the IL-23/IL-17 inflammatory pathway. Furthermore, one of the
most advantageous points of therapy targeting IL-17-producing
T lymphocytes is its long therapeutic time window. Since current
globally approved therapy is limited to intravenous administration
of tPA,which should be performed within 4.5 h after stroke onset,
further elucidation of the inflammatory mechanisms of the T lym-
phocytes that infiltrate the ischemic brain during the 24-h-period
after stroke onset is needed.
FTY720 is one of the most promising therapeutic tools for
ischemic stroke at this time (Wei et al., 2011). FTY720 decreases
the number of infiltrating T lymphocytes, including γδT lympho-
cytes. The most troublesome side effect of FTY720 administration
is the increased incidence of bacterial pneumonia after stroke
(Meisel and Meisel, 2011). It is possible that FTY720 interferes
with peripheral lymphocyte distribution in the body after stroke
and thus inhibits the protective function of peripheral T lympho-
cytes against bacterial infection. Although it has been reported that
FTY720 does not promote spontaneous bacterial infection after
experimental stroke in mice, future studies aimed at developing
effective clinical interventions for stroke patients should seek to
minimize this kind of detrimental effect of FTY720 (Pfeilschifter
et al., 2011).
In conclusion, research has gradually shed light on the mech-
anisms of post-ischemic inflammation. A deeper understanding
of these intricacies in the context of medical intervention should
enable the development of novel neuroprotective strategies that
are more effective and have a longer therapeutic time window
(Figure 5).
FIGURE 5 | Strategy for developing neuroprotective therapy by
suppressing neurotoxic inflammatory response.The targeting of specific
inflammatory mediators from macrophages and T lymphocytes can
attenuate neurotoxic inflammatory reactions.
www.frontiersin.org May 2012 | Volume 3 | Article 132 | 5
Shichita et al. Mechanisms of cerebral post-ischemic inflammation
REFERENCES
Abulafia, D. P., de Rivero Vaccari,
J. P., Lozano, J. D., Lotocki, G.,
Keane, R. W., and Dietrich, W. D.
(2009). Inhibition of the inflamma-
some complex reduces the inflam-
matory response after thromboem-
bolic stroke in mice. J. Cereb. Blood
Flow Metab. 29, 534–544.
Asahi, M., Asahi, K., Jung, J. C., del
Zoppo, G. J., Fini, M. E., and Lo, E.
H. (2000). Role for matrix metallo-
proteinase 9 after cerebral ischemia:
effects of gene knockout and enzyme
inhibition with BB-94. J. Cereb.
Blood Flow Metab. 20, 1681–1689.
Barone, F. C., and Feuerstein, G.
Z. (1999). Inflammatory mediators
and stroke: new opportunities for
novel therapeutics. J. Cereb. Blood
Flow Metab. 19, 819–834.
Becker, K., Kindrick, D., McCarron,
R., Hallenbeck, J., and Winn, R.
(2003). Adoptive transfer of myelin
basic protein-tolerized splenocytes
to naive animals reduces infarct size:
a role for lymphocytes in ischemic
brain injury? Stroke 34, 1809–1815.
Becker, K. J. (2009). Sensitization
and tolerization to brain anti-
gens in stroke. Neuroscience 158,
1090–1097.
Boutin, H., LeFeuvre, R. A., Horai, R.,
Asano,M., Iwakura, Y., and Rothwell,
N. J. (2001). Role of IL-1alpha and
IL-1beta in ischemic brain damage.
J. Neurosci. 21, 5528–5534.
Chakraborty, S., Kaushik, D. K., Gupta,
M., and Basu, A. (2010). Inflam-
masome signaling at the heart of
central nervous system pathology. J.
Neurosci. Res. 88, 1615–1631.
Chen, C. J., Kono, H., Golenbock, D.,
Reed, G., Akira, S., and Rock, K. L.
(2007). Identification of a key path-
way required for the sterile inflam-
matory response triggered by dying
cells. Nat. Med. 13, 851–856.
Chen, F.,Liu, Z., Wu, W., Rozo, C., Bow-
dridge, S., Millman, A., Van Rooi-
jen, N., Urban, J. F. Jr., Wynn, T. A.,
and Gause, W. C. (2012). An essen-
tial role for TH2-type responses in
limiting acute tissue damage dur-
ing experimental helminth infec-
tion. Nat. Med. 18, 260–266.
Connolly, E. S. Jr., Winfree, C. J.,
Springer, T. A., Naka, Y., Liao, H.,
Yan, S. D., Stern, D. M., Solomon, R.
A., Gutierrez-Ramos, J. C., and Pin-
sky, D. J. (1996). Cerebral protection
in homozygous null ICAM-1 mice
after middle cerebral artery occlu-
sion. Role of neutrophil adhesion in
the pathogenesis of stroke. J. Clin.
Invest. 97, 209–216.
Cua, D.J., Sherlock, J., Chen, Y., Murphy,
C. A., Joyce, B., Seymour, B., Lucian,
L., To, W., Kwan, S., Churakova, T.,
Zurawski, S., Wiekowski, M., Lira, S.
A., Gorman, D., Kastelein, R. A., and
Sedgwick, J. D. (2003). Interleukin-
23 rather than interleukin-12 is the
critical cytokine for autoimmune
inflammation of the brain. Natu re
421, 744–748.
Dev,K. K., Mullershausen, F., Mattes,H.,
Kuhn, R. R., Bilbe, G., Hoyer, D., and
Mir,A. (2008). Brain sphingosine-1-
phosphate receptors: implication for
FTY720 in the treatment of multi-
ple sclerosis. Pharmacol. Ther. 117,
77–93.
Eltzschig, H. K., and Eckle, T. (2011).
Ischemia and reperfusion from
mechanism to translation. Nat. Med.
17, 1391–1401.
Famakin, B. M., Mou, Y., Ruetzler, C.
A., Bembry, J., Maric, D., and Hal-
lenbeck, J. M. (2011). Disruption of
downstream MyD88 or TRIF Toll-
like receptor signaling does not pro-
tect against cerebral ischemia. Brain
Res. 1388, 148–156.
Fossiez, F., Djossou, O., Chomarat, P.,
Flores-Romo, L., Ait-Yahia, S., Maat,
C., Pin, J. J., Garrone, P., Garcia,
E., Saeland, S., Blanchard, D., Gail-
lard, C., Das Mahapatra, B., Rou-
vier, E., Golstein, P., Banchereau,
J., and Lebecque, S. (1996). T
cell interleukin-17 induces stromal
cells to produce proinflammatory
and hematopoietic cytokines. J. Exp.
Med. 183, 2593–2603.
Fraser, P. A. (2011). The role of radical
generation in increasing cerebrovas-
cular permeability. Free Radic. Biol.
Med. 51, 967–977.
Gertz, K., Kronenberg, G., Kälin, R. E.,
Baldinger, T., Werner, C., Balkaya,
M., Eom, G. D., Hellmann-Regen,
J., Kröber, J., Miller, K. R., Lindauer,
U., Laufs, U., Dirnagl, U., Hepp-
ner, F. L., and Endres, M. (2012).
Essential role of interleukin-6 in
post-stroke angiogenesis. Brain.doi:
10.1093/brain/aws075
Hallenbeck, J. M. (2002). The many
faces of tumor necrosis factor in
stroke. Nat. Med. 8, 1363–1368.
Hasegawa, Y., Suzuki, H., Sozen, T.,
Rolland, W., and Zhang, J. H.
(2010). Activation of sphingosine
1-phosphate receptor-1 by FTY720
is neuroprotective after ischemic
stroke in rats. Stroke 41, 368–374.
Hayakawa, K., Qiu, J., and Lo, E.
H. (2010). Biphasic actions of
HMGB1 signaling in inflammation
and recovery after stroke. Ann. N. Y.
Acad. Sci. 1207, 50–57.
Hurn, P. D., Subramanian, S., Parker,
S. M., Afentoulis, M. E., Kaler, L.
J., Vandenbark, A. A., and Offner,
H. (2007). T- and B-cell-deficient
mice with experimental stroke have
reduced lesion size and inflamma-
tion. J. Cereb. Blood Flow Metab. 27,
1798–1805.
Hyakkoku, K., Hamanaka, J., Tsuruma,
K., Shimazawa, M., Tanaka, H.,
Uematsu, S., Akira, S., Inagaki, N.,
Nagai, H., and Hara, H. (2010).
Toll-like receptor 4 (TLR4), but not
TLR3 or TLR9, knock-out mice have
neuroprotective effects against focal
cerebral ischemia. Neuroscience 171,
258–267.
Iadecola, C. (2004). Neurovascular reg-
ulation in the normal brain and in
Alzheimer’s disease. Nat. Rev. Neu-
rosci. 5, 347–360.
Iadecola, C., and Anrather, J. (2011).
The immunology of stroke: from
mechanisms to translation. Nat.
Med. 17, 796–808.
Ifergan, I., Kébir, H., Bernard, M.,
Wosik, K., Dodelet-Devillers, A.,
Cayrol, R., Arbour, N., and Prat,
A. (2008). The blood-brain barrier
induces differentiation of migrat-
ing monocytes into Th17-polarizing
dendritic cells. Brain 131, 785–799.
Jander, S., Karemer, M., Schroeter,
M., Witte, O. W., and Stoll, G.
(1995). Lymphocytic infiltration
and expression of intercellular adhe-
sion molecule-1 in photochemically
induced ischemia of the rat cortex. J.
Cereb. Blood Flow Metab. 15, 42–51.
Jung, J. E., Kim, G. S., and Chan,
P. H. (2011). Neuroprotection by
interleukin-6 is mediated by signal
transducer and activator of tran-
scription 3 and antioxidative signal-
ing in ischemic stroke. Stroke 42,
3574–3579.
Kebir, H., Kreymborg, K., Ifergan, I.,
Dodelet-Devillers, A., Cayrol, R.,
Bernard, M., Giuliani, F., Arbour,
N., Becher, B., and Prat, A. (2007).
Human TH17 lymphocytes promote
blood-brain barrier disruption and
central nervous system inflamma-
tion. Nat. Med. 13, 1173–1175.
Kim, J. B., Sig Choi, J., Yu, Y. M., Nam,
K., Piao, C. S., Kim, S. W.,Lee, M. H.,
Han, P. L., Park, J. S., and Lee, J. K.
(2006). HMGB1, a novel cytokine-
like mediator linking acute neuronal
death and delayed neuroinflamma-
tion in the postischemic brain. J.
Neurosci. 26, 6413–6421.
Konoeda, F., Shichita, T., Yoshida, H.,
Sugiyama, Y., Muto, G., Hasegawa,
E., Morita, R., Suzuki, N., and
Yoshimura, A. (2010). Therapeutic
effect of IL-12/23 and their signaling
pathway blockade on brain ischemia
model. Biochem. Biophys. Res. Com-
mun. 402, 500–506.
Lambertsen, K. L., Gregersen, R.,
Meldgaard, M., Clausen, B. H.,
Heibøl, E. K., Ladeby, R., Knudsen,
J., Frandsen, A., Owens, T., and Fin-
sen, B. (2004). A role for interferon-
gamma in focal cerebral ischemia in
mice. J. Neuropathol. Exp.Neurol. 63,
942–955.
Liesz, A., Suri-Payer, E., Veltkamp, C.,
Doerr, H., Sommer, C., Rivest, S.,
Giese, T., and Veltkamp, R. (2009).
Regulatory T cells are key cerebro-
protective immunomodulators in
acute experimental stroke. Nat. Med.
15, 192–199.
Liesz, A., Zhou, W., Mracskó, É.,
Karcher, S., Bauer, H., Schwart-
ing, S., Sun, L., Bruder, D., Stege-
mann, S., Cerwenka, A., Sommer,
C., Dalpke, A. H., and Veltkamp,
R. (2011). Inhibition of lympho-
cyte trafficking shields the brain
against deleterious neuroinflam-
mation after stroke. Brain 134,
704–720.
Liu, K., Mori, S., Takahashi, H. K.,
Tomono, Y., Wake, H., Kanke, T.,
Sato, Y., Hiraga, N., Adachi, N.,
Yoshino, T., and Nishibori, M.
(2007). Anti-high mobility group
box 1 monoclonal antibody ame-
liorates brain infarction induced by
transient ischemia in rats. FASEB J.
21, 3904–3916.
Lo, E. H. (2010). Degeneration and
repair in central nervous system dis-
ease. Nat. Med. 16, 1205–1209.
Macrez, R., Ali, C., Toutirais, O., Le
Mauff, B., Defer,G., Dir nagl, U., and
Vivien, D. (2011). Stroke and the
immune system: from pathophysi-
ology to new therapeutic strategies.
Lancet Neurol. 10, 471–480.
Marsh, B. J., Williams-Karnesky,
R. L., and Stenzel-Poore, M. P.
(2009). Toll-like receptor signaling
in endogenous neuroprotection
and stroke. Neuroscience 158,
1007–1020.
Martin, B., Hirota, K., Cua, D. J.,
Stockinger, B., and Veldhoen, M.
(2009). Interleukin-17-producing
gammadelta T cells selectively
expand in response to pathogen
products and environmental signals.
Immunity 31, 321–330.
Martinon, F., Burns, K., and Tschopp, J.
(2002). The inflammasome: a mole-
cular platform triggering activation
of inflammatory caspases and pro-
cessing of proIL-1beta. Mol. Cell 10,
417–426.
Meisel, C., and Meisel, A. (2011). Sup-
pressing immunosuppression after
stroke. N.Engl.J.Med.365,
2134–2136.
Morancho, A., Rosell, A., Garcia-
Bonilla, L., and Montaner, J. (2010).
Metalloproteinase and stroke infarct
size: role for anti-inflammatory
Frontiers in Immunology | Inflammation May 2012 | Volume 3 | Article 132 | 6
Shichita et al. Mechanisms of cerebral post-ischemic inflammation
treatment? Ann. N.Y. Acad. Sci. 1207,
123–133.
Moskowitz, M. A., Lo, E. H., and
Ladecola, C. (2010). The science
of stroke: mechanisms in search of
treatments. Neuron 67, 181–198.
Ooboshi, H., Ibayashi, S., Shichita,
T., Kumai, Y., Takada, J., Ago,
T., Arakawa, S., Sugimori, H.,
Kamouchi, M., Kitazono, T., and
Iida, M. (2006). Postischemic gene
transfer of interleukin-10 protects
against both focal and global brain
ischemia. Circulation 111, 913–919.
Patenaude, A., Murthy, M. R., and
Mirault, M. E. (2005). Emerging
roles of thioredoxin cycle enzymes
in the central nervous system. Cell.
Mol. Life Sci. 62, 1063–1080.
Pfeilschifter, W., Czech-Zechmeister, B.,
Sujak, M., Foerch, C., Wichelhaus, T.
A., and Pfeilschifter, J. (2011). Treat-
ment with the immunomodulator
FTY720 does not promote spon-
taneous bacterial infections after
experimental stroke in mice. Exp.
Transl. Stroke Med. 3, 2.
Qiu, J., Nishimura, M.,Wang, Y., Sims, J.
R., Qiu, S., Savitz, S. I., Salomone, S.,
and Moskowitz, M. A. (2008). Early
release of HMGB-1 from neurons
after the onset of brain ischemia.
J. Cereb. Blood Flow Metab. 28,
927–938.
Rashidian, J., Rousseaux, M. W.,
Venderova, K., Qu, D., Callaghan, S.
M., Phillips, M., Bland, R. J., Dur-
ing, M. J., Mao, Z., Slack, R. S., and
Park, D. S. (2009). Essential role of
cytoplasmic cdk5 and Prx2 in multi-
ple ischemic injury models, in vivo.
J. Neurosci. 29, 12497–12505.
Reboldi, A., Coisne, C., Baumjohann,
D., Benvenuto, F., Bottinelli, D.,
Lira, S., Uccelli, A., Lanzavecchia,
A., Engelhardt, B., and Sallusto, F.
(2009). C-C chemokine receptor
6-regulated entry of TH-17 cells
into the CNS through the choroid
plexus is required for the initia-
tion of EAE. Nat. Immunol. 10,
514–523.
Ren, X., Akiyoshi, K., Dziennis, S.,Van-
denbark, A. A., Herson, P. S., Hurn,
P. D., and Offner, H. (2011). Regula-
tory B cells limit CNS inflammation
and neurologic deficits in murine
experimental stroke. J. Neurosci. 31,
8556–8563.
Rivest, S. (2009). Regulation of innate
immune responses in the brain. Nat.
Rev. Immunol. 9, 429–439.
Schroeter, M., Jander, S., Witte, O. W.,
and Stoll, G. (1994). Local immune
responses in the rat middle cerebral
artery occlusion. J. Neuroimmunol.
55, 195–203.
Shichita, T., Hasegawa, E., Kimura, A.,
Morita, R., Sakaguchi, R., Takada,
I., Sekiya, T., Ooboshi, H., Kita-
zono, T., Yanagawa, T., Ishii, T., Taka-
hashi, H., Mori, S., Nishibori, M.,
Kuroda, K., Miyake, K., Akira, S.,
and Yoshimura, A. (2012). Peroxire-
doxin family proteins are key ini-
tiators of post-ischemic inflamma-
tion in the brain. Nat. Med. doi:
10.1038/nm.2749
Shichita, T., Sugiyama, Y., Ooboshi, H.,
Sugimori, H., Nakagawa, R., Takada,
I., Iwaki, T., Okada, Y., Iida, M., Cua,
D. J., Iwakura, Y., and Yoshimura,
A. (2009). Pivotal role of cere-
bral interleukin-17-producing gam-
madeltaT cells in the delayed phase
of ischemic brain injury. Nat. Med.
15, 946–950.
Stewart, C. R., Stuart, L. M., Wilkinson,
K., van Gils, J. M., Deng, J., Halle,
A., Rayner, K. J., Boyer, L., Zhong,
R., Frazier, W. A., Lacy-Hulbert,
A., El Khoury, J., Golenbock, D.
T., and Moore, K. J. (2010). CD36
ligands promote sterile inflamma-
tion through assembly of a Toll-like
receptor 4 and 6 heterodimer. Nat.
Immunol. 11, 155–161.
Strecker, J. K., Minnerup, J., Gess,
B., Ringelstein, E. B., Schäbitz,
W. R., and Schilling, M. (2011).
Monocyte chemoattractant protein-
1-deficiency impairs the expression
of IL-6, IL-1b and G-CSF after tran-
sient focal ischemia in mice. PLoS
ONE 6, e25863. doi:10.1371/jour-
nal.pone.0025863
Subramanian, S., Zhang, B., Kosaka, Y.,
Burrows, G. G., Grafe, M. R., Van-
denbark, A. A., Hurn, P. D., and
Offner, H. (2009). Recombinant T
cell receptor ligand treats experi-
mental stroke. Stroke 40, 2539–2545.
Sutton, C. E., Lalor, S. J., Sweeney, C.
M., Brereton, C. F., Lavelle, E. C.,
and Mills, K. H. (2009). Interleukin-
1 and IL-23 induce innate IL-
17 production from gammadelta
T cells, amplifying Th17 responses
and autoimmunity. Immunity 31,
331–341.
Takano, T., Oberheim, N., Cotrina,
M. L., and Nedergaard, M. (2009).
Astrocytes and ischemic injury.
Stroke 40, S8–S12.
Tang, S. C., Arumugam, T. V., Xu, X.,
Cheng, A., Mughal, M. R., Jo, D. G.,
Lathia, J. D.,Siler, D. A., Chigurupati,
S., Ouyang, X., Magnus, T., Caman-
dola, S., and Mattson, M. P. (2007).
Pivotal role for neuronal Toll-like
receptors in ischemic brain injury
and functional deficits. Proc. Natl.
Acad. Sci. U.S.A. 104, 13798–13803.
Terao, S., Yilmaz, G., Stokes, K. Y., Rus-
sell, J., Ishikawa, M., Kawase, T., and
Granger, D. N. (2008). Blood cell-
derived RANTES mediates cerebral
microvascular dysfunction, inflam-
mation, and tissue injury after
focal ischemia-reperfusion. Stroke
39, 2560–2570.
Terao, Y., Ohta, H., Oda, A., Nakagaito,
Y., Kiyota,Y., and Shintani, Y. (2009).
Macrophage inflammatory protein-
3alpha plays a key role in the inflam-
matory cascade in rat focal cerebral
ischemia. Neurosci. Res. 64, 75–82.
Wei, Y., Yemisci, M., Kim, H. H., Yung, L.
M., Shin,H. K., Hwang, S. K., Guo,S.,
Qin, T.,Alsharif, N., Brinkmann, V.,
Liao, J. K., Lo, E. H., and Waeber, C.
(2011). Fingolimod provides long-
term protection in rodent models of
cerebral ischemia. Ann. Neurol. 69,
119–129.
Xiong, X., Barreto, G. E., Xu,L., Ouyang,
Y. B., Xie, X., and Giffard, R. G.
(2011). Increased brain injury and
worsened neurological outcome in
interleukin-4 knockout mice after
transient focal cerebral ischemia.
Stroke 42, 2026–2032.
Yadav, A., Kalita, A., Dhillon, S., and
Banerjee, K. (2005). JAK/STAT3
pathway is involved in survival
of neurons in response to insulin
like growth factor and nega-
tively regulated by suppressor of
cytokine signal-3. J. Biol. Chem. 280,
31830–31840.
Yamashita, T., Sawamoto, K., Suzuki,
S., Suzuki, N., Adachi, K., Kawase,
T., Mihara, M., Ohsugi, Y., Abe,
K., and Okano, H. (2005). Block-
ade of interleukin-6 signaling aggra-
vates ischemic cerebral damage
in mice: possible involvement of
Stat3 activation in the protec-
tion of neurons. J. Neurochem. 94,
459–468.
Yanai, H., Ban, T.,Wang, Z., Choi, M. K.,
Kawamura,T.,Negishi, H., Nakasato,
M., Lu, Y., Hangai, S., Koshiba, R.,
Savitsky, D., Ronfani, L., Akira, S.,
Bianchi, M. E., Honda, K., Tamura,
T., Kodama, T., and Taniguchi, T.
(2009). HMGB proteins function as
universal sentinels for nucleic-acid-
mediated innate immune responses.
Nature 462, 99–103.
Yang, Q. W., Lu, F. L., Zhou, Y., Wang,
L., Zhong, Q., Lin, S., Xiang, J., Li,
J. C., Fang, C. Q., and Wang, J. Z.
(2011). HMBG1 mediates ischemia-
reperfusion injury by TRIF-adaptor
independent Toll-like receptor 4 sig-
naling. J. Cereb. Blood Flow Metab.
31, 593–605.
Yilmaz, G., Arumugam, T. V., Stokes, K.
Y., and Granger, D. N. (2006). Role
of T lymphocytes and interferon-
gamma in ischemic stroke. Circula-
tion 113, 2105–2112.
Zhang, J., Takahashi, H. K., Liu, K.,
Wake, H., Liu, R., Maruo, T.,
Date, I., Yoshino, T., Ohtsuka, A.,
Mori, S., and Nishibori, M. (2011).
Anti-high mobility group box-1
monoclonal antibody protects the
blood-brain barrier from ischemia-
induced disruption in rats. Stroke 42,
1420–1428.
Zhang, Q., Raoof, M., Chen, Y., Sumi,
Y., Sursal, T., Junger, W., Brohi,
K., Itagaki, K., and Hauser, C.
J. (2010). Circulating mitochon-
drial DAMPs cause inflammatory
responses to injury. Nature 464,
104–107.
Conflict of Interest Statement: The
authors declare that the research was
conducted in the absence of any com-
mercial or financial relationships that
could be construed as a potential con-
flict of interest.
Received: 14 April 2012; paper pend-
ing published: 30 April 2012; accepted:
08 May 2012; published online: 31 May
2012.
Citation: Shichita T, Sakaguchi R,
Suzuki M and Yoshimura A (2012)
Post-ischemic inflammation in the
brain. Front. Immun. 3:132. doi:
10.3389/fimmu.2012.00132
This article was submitted to Frontiers in
Inflammation, a specialty of Frontiers in
Immunology.
Copyright © 2012 Shichita, Sakaguchi,
Suzuki and Yoshimura. This is an open-
access article distributed under the terms
of the Creative Commons Attribution
Non Commercial License, which per-
mits non-commercial use, distribution,
and reproduction in other forums, pro-
vided the original authors and source are
credited.
www.frontiersin.org May 2012 | Volume 3 | Article 132 | 7
... Our CPB swine model offered an opportunity to critically explore this issue. We observed a significant increase in hematocrit during mild hypothermic CPB (median [IQR] = +58 [26,74] % relative to baseline), which remained high throughout the entire 24 h survival window. The increase was anticipated because baseline hematocrit (23 [20,27] %) was consistently lower than the target hematocrit goal during CPB. ...
... In healthy 1-month-old swine who underwent 2 h of mild hypothermic (30 • C) CPB, expression of IL-8 and TNF-α were observed to increase with CPB time, then remain elevated at 1 h after CPB, and finally resolve by 6 h [57]. The inflection of physiologic trends between the hyper-acute and acute survival periods that we observed are consistent with this and other prior evidence of the timing of an inflammatory response [73,74]. Concurrent MAP elevation with diminished CBF during the survival period lends further support to the hypothesized mechanism of increased intracranial pressure that may occur secondary to inflammation [75,76]. ...
Article
Full-text available
Cardiopulmonary bypass (CPB) provides cerebral oxygenation and blood flow (CBF) during neonatal congenital heart surgery, but the impacts of CPB on brain oxygen supply and metabolic demands are generally unknown. To elucidate this physiology, we used diffuse correlation spectroscopy and frequency-domain diffuse optical spectroscopy to continuously measure CBF, oxygen extraction fraction (OEF), and oxygen metabolism (CMRO2) in 27 neonatal swine before, during, and up to 24 h after CPB. Concurrently, we sampled cerebral microdialysis biomarkers of metabolic distress (lactate–pyruvate ratio) and injury (glycerol). We applied a novel theoretical approach to correct for hematocrit variation during optical quantification of CBF in vivo. Without correction, a mean (95% CI) +53% (42, 63) increase in hematocrit resulted in a physiologically improbable +58% (27, 90) increase in CMRO2 relative to baseline at CPB initiation; following correction, CMRO2 did not differ from baseline at this timepoint. After CPB initiation, OEF increased but CBF and CMRO2 decreased with CPB time; these temporal trends persisted for 0–8 h following CPB and coincided with a 48% (7, 90) elevation of glycerol. The temporal trends and glycerol elevation resolved by 8–24 h. The hematocrit correction improved quantification of cerebral physiologic trends that precede and coincide with neurological injury following CPB.
... TLRs play a crucial role in modulating ischemic brain damage post-stroke [177,178]. In neonatal hypoxic-ischemic brain damage in rats, inhibition of the TLR4/MyD88 signaling pathway activation led to improved hippocampal structure and decreased inflammatory markers, indicating neuroprotective effects [179]. ...
Article
Full-text available
Pregnane neuroactive steroids, notably allopregnanolone and pregnenolone, exhibit efficacy in mitigating inflammatory signals triggered by toll-like receptor (TLR) activation, thus attenuating the production of inflammatory factors. Clinical studies highlight their therapeutic potential, particularly in conditions like postpartum depression (PPD), where the FDA-approved compound brexanolone, an intravenous formulation of allopregnanolone, effectively suppresses TLR-mediated inflammatory pathways, predicting symptom improvement. Additionally, pregnane neurosteroids exhibit trophic and anti-inflammatory properties, stimulating the production of vital trophic proteins and anti-inflammatory factors. Androstane neuroactive steroids, including estrogens and androgens, along with dehydroepiandrosterone (DHEA), display diverse effects on TLR expression and activation. Notably, androstenediol (ADIOL), an androstane neurosteroid, emerges as a potent anti-inflammatory agent, promising for therapeutic interventions. The dysregulation of immune responses via TLR signaling alongside reduced levels of endogenous neurosteroids significantly contributes to symptom severity across various neuropsychiatric disorders. Neuroactive steroids, such as allopregnanolone, demonstrate efficacy in alleviating symptoms of various neuropsychiatric disorders and modulating neuroimmune responses, offering potential intervention avenues. This review emphasizes the significant therapeutic potential of neuroactive steroids in modulating TLR signaling pathways, particularly in addressing inflammatory processes associated with neuropsychiatric disorders. It advances our understanding of the complex interplay between neuroactive steroids and immune responses, paving the way for personalized treatment strategies tailored to individual needs and providing insights for future research aimed at unraveling the intricacies of neuropsychiatric disorders.
... The incidence of AIS increases with aging, and age is an important risk factor (2). Inflammation is known to occur in the pathophysiology of AIS. Necrotic cells, which are formed in the brain due to vascular occlusion, trigger inflammation in acut ischemic stroke (3). In addition, abnormal blood clotting, poor nutritional status and inflammation are associated with a poor prognosis of AIS (4,5). ...
Article
Full-text available
Aim: In this study, we aimed to evaluate the relationship between the HALP score, calculated by hemoglobin, albumin, lymphocyte and platelet values, and 28-day mortality in very elderly geriatric critically ill patients with acute ischemic stroke.Material and Method: The study was designed retrospectively and patients aged 85 years and older admitted to the general intensive care unit with the diagnosis of acute ischemic stroke were evaluated. Demographic data, laboratory data and HALP scores of these patients were recorded. Patients who died within 28 days in intensive care follow-up were defined as the Non-Survival group, and patients who did not die were defined as the survival group.Results:There was a statistically significant difference between the groups in terms of hemoglobin values admitted to the intensive care unit (p:0.00). For albumin, patients in the Non-Survival group had lower values, but there was no statistically significant difference between the groups (p: 0.054). Non-Survival group had lower values for lymphocytes and there was a statistically significant difference between the groups (p: 0.00). For platelet value, patients in the Non-Survival group had higher values and there was no statistically significant difference between the groups (p: 0.164). Patients in the Non-Survival group had lower values for HALP score and there was a statistically significant difference between the groups (p: 0.00)Conclusion: The HALP score is associated with 28-day mortality in very elderly geriatric critically ill patients with acute ischemic stroke. However, it has low sensitivity (30.1%) and specificity (27.9%).
... However, ischemia-reperfusion initiates several cellular responses in the brain. These cellular responses include the upregulation of inflammatory cytokines, the activation of endothelial cells, and the migration of immune cells to brain tissue [78][79][80][81]. These cellular responses can lead to brain cell damage, namely ischemia-reperfusion injury [82]. ...
Article
Full-text available
The efficacy and safety of treating ischemic stroke is still a challenging problem at this stage. Ischemic stroke has a special stroke microenvironment. In recent years, improving stroke microenvironment has become a new idea for treating ischemic stroke. At the same time, nanoparticles have unique physical and chemical properties and significant advantages in studying ischemic stroke. Therefore, in recent years, researchers have designed various types of nanoparticles in response to stroke microenvironment to treat ischemic stroke. In this review, we summarized and analyzed the ischemic areas targeted by nanoparticles in response to reactive oxygen species, pH, high expression of receptors in the blood–brain barrier, enrichment of molecules in the stroke microenvironment, light, magnetic harmony, and other stimuli. We analyze its advantages and disadvantages and look forward to the development prospect of this field. Hope to provide strategies for better treatment of ischemic stroke.
... The onset of stroke can initiate an inflammatory cascade in both the systemic immune system and central nervous system (CNS). This cascade plays a crucial role in the progression of cerebral ischemia pathology [2]. ...
Article
Full-text available
Interleukin (IL)-33 was initially recognized as a constituent of the IL-1 cytokine family in 2005. It exerts pleiotropic effects by regulating immune responses via its binding to the receptor ST2 (IL-33R). The IL-33/ST2 pathway has been linked to several inflammatory disorders. In human and rodents, the broad expression of IL-33 in spinal cord tissues and brain indicates its central nervous system (CNS)-specific functions. Growing evidence supports the protective effects of the IL-33/ST2 pathway in ischemic stroke, along with a better understanding of the underlying mechanisms. IL-33 plays a crucial role in the regulation of the release of inflammatory molecules from glial cells in response to neuropathological lesions. Moreover, IL-33/ST2-mediated neuroprotection following cerebral ischemia may be linked to T cell function, specifically regulatory T cells (Tregs). Soluble ST2 (sST2) acts as a decoy receptor in the IL-33/ST2 axis, blocking IL-33 signaling through the membrane ST2 receptor. sST2 has also been identified as a potential inflammatory biomarker of ischemic stroke. Targeting sST2 specifically to eliminate its inhibition of the protective IL-33/ST2 pathway in ischemic brain tissues is a promising approach for the treatment of ischemic stroke.
... Ischemia and the resultant hypoxia lead to cellular oxidative stress and remodeling of oxygen metabolism, resulting into cell death, release of cytokines, chemokines and "danger molecules" (4). Hypoxia and oxidative stress induce activation of hypoxia-inducible factor HIF-1a, which induces expression of VEGF, an activator of neovascularization (5). ...
Article
Full-text available
Ischemia causes an inflammatory response featuring monocyte-derived macrophages (MF) involved in angiogenesis and tissue repair. Angiogenesis and ischemic macrophage differentiation are regulated by Notch signaling via Notch ligand Delta-like 1 (Dll1). Colony stimulating factor 1 (CSF-1) is an essential MF lineage factor, but its role in ischemic macrophage development and the interaction with Notch signaling is so far unclear. Using a mouse model of hind limb ischemia with CSF-1 inhibitor studies and Dll1 heterozygous mice we show that CSF-1 is induced in the ischemic niche by a subpopulation of stromal cells expressing podoplanin, which was paralleled by the development of ischemic macrophages. Inhibition of CSF-1 signaling with small molecules or blocking antibodies impaired macrophage differentiation but prolonged the inflammatory response, resulting in impaired perfusion recovery and tissue regeneration. Yet, despite high levels of CSF-1, macrophage maturation and perfusion recovery were impaired in mice with Dll1 haploinsufficiency, while inflammation was exaggerated. In vitro , CSF-1 was not sufficient to induce full MF differentiation from donor monocytes in the absence of recombinant DLL1, while the presence of DLL1 in a dose-dependent manner stimulated MF differentiation in combination with CSF-1. Thus, CSF-1 is an ischemic niche factor that cooperates with Notch signaling in a non-redundant fashion to instruct macrophage cell fate and maturation, which is required for ischemic perfusion recovery and tissue repair.
... Invading leucocytes (and injured brain cells) produce various inflammatory mediators, causing inflammation. 77 MMP-12 aids in the release of IFN-α, which stimulates the generation of TNFα, interleukins (1, 2 and 6) and IFN-γ. 78 In addition, MMP-12 has been implicated in the release of the pro-inflammatory mediator, tumour necrosis factor-α (TNFα) and the activation of pro-TNFα. ...
Article
Full-text available
This article focuses on the emerging role of matrix metalloproteinase-12 (MMP-12) in ischaemic stroke (IS). MMP-12 expression in the brain increases dramatically in animal models of IS, and its suppression reduces brain damage and promotes neurological, sensorimotor and cognitive functional outcomes. Thus, MMP-12 could represent a potential target for the management of IS. This article provides an overview of MMP-12 upregulation in the brain following IS, its deleterious role in the post-stroke pathogenesis (blood-brain barrier disruption, inflammation, apoptosis and demyelination), possible molecular interactions and mechanistic insights, its involvement in post-ischaemic functional deficits and recovery as well as the limitations, perspectives, challenges and future directions for further research. Prior to testing any MMP-12-targeted therapy in patients with acute IS, additional research is needed to establish the effectiveness of MMP-12 suppression against IS in older animals and in animals with comorbidities. This article also examines the clinical implications of suppressing MMP-12 alone or in combination with MMP-9 for extending the currently limited tissue plasminogen activator therapy time window. Targeting of MMP-12 is expected to have a profound influence on the therapeutic management of IS in the future.
... Neutrophils are the most abundant immune cells in peripheral blood, playing important roles in the inflammatory response after ACI. Some studies revealed that neutrophils rapidly increased and migrated to the central nervous system (CNS) after ACI [7][8][9]. Cai et al. [10] established a mouse model of middle cerebral artery occlusion (MCAO) and reported that the levels of peripheral blood neutrophil chemokines (including CXCL1, CXCL2, CXCL3 and CXCL5) rapidly increase after cerebral I/R, reaching a peak at 12 h. The level of neutrophils in the peripheral blood and spleen increased rapidly after cerebral ischemia and peaked at 24 h. ...
Article
Full-text available
Background: Neutrophils play crucial roles in the inflammatory response after acute cerebral infarction (ACI). Previous studies revealed neutrophils are non-homogeneous and can be divided into at least two subtypes, pro-inflammatory and anti-inflammatory, correlated with patients' prognosis. Objective: We aimed to explore the correlation between disease severity and peripheral blood neutrophils in patients with ACI and determine whether remote ischemic postconditioning (RIPostC) exerts neuroprotective effects by regulating neutrophils. Methods: Patients (n = 38) with acute anterior circulation cerebral infarction were assigned to conventional treatment (n = 24; included aspirin, statins, neuro nutrition drugs, and circulation improvement drugs) or RIPostC (n = 14; 7-day ischemia adaptation [complete ischemia of both upper extremities for 5 minutes followed by remission for 5 minutes, 5 repeated cycles, twice a day, started from the morning of the second day of admission] based on conventional treatment) groups, based on their preference. General clinical data and peripheral blood samples were taken three times, in the morning before and 3 and 7 days after treatment. Fifteen adults with non-acute cerebral infarction matched for sex, age, and risk factors were recruited as controls; peripheral blood samples were only collected on the recruitment day. We used flow cytometry to detect the percentage of neutrophils and Real-Time PCR to detect the gene expression of interleukin (IL)-1β in the peripheral blood samples. Results: The percentage of neutrophils, pro-inflammatory neutrophils (IL-1β high expression in flow cytometry), and IL-1β mRNA expression increased after ACI (P = 0.01, P = 0.001, P < 0.001). The National Institutes of Health Stroke Scale (NIHSS) score of patients with ACI within one day of onset was positively correlated with the percentage of pro-inflammatory neutrophils (R = 0.618, P = 0.043). Pro-inflammatory neutrophils in the RIPostC group decreased compared with those in the conventional treatment group, with the most significant difference observed on Day 7 (P = 0.01). However, the percentage of neutrophils was not statistically different. IL-1β mRNA expression decreased, with the most significant difference on Day 3 (P = 0.004). The NIHSS and Modified Rankin Scale scores for RIPostC decreased more significantly than for conventional treatment (P = 0.002, P = 0.019). Conclusion: More severe cerebral infarction was associated with a higher percentage of pro-inflammatory neutrophils. The neuroprotective effect of RIPostC may partly be exerted through gene regulation to reduce pro-inflammatory neutrophils.
Chapter
Full-text available
Drug-resistant epilepsy (DRE) is characterized by the persistence of epileptic seizures despite the administration of antiepileptic treatments. The pathogenesis of this condition has been associated with many different molecular and cellular mechanisms. In this chapter, we describe how the CNS immune response is associated with the onset of epilepsy and the molecular mechanisms underlying brain injury associated with neuroinflammatory response triggered by epileptic seizures. Next, we describe the possible role of neuroinflammation in the progression from epilepsy to DRE.KeywordsDrug-resistant epilepsyInnate immune systemAdaptive immune systemBlood-brain barrierOxidative stress
Article
Full-text available
The development of next generation therapy for ischemic stroke is important for the aging society. To control post-ischemic inflammation could be a hopeful therapeutic approach. In this review, we focus on the triggers of inflammation after ischemic stroke. Various danger associated molecular patterns (DAMPs) activate pattern recognition receptors (PRRs) in immune cells and trigger the acute sterile inflammation in brain. The activation of inflammasome is also important for IL-1β production in ischemic brain. Ibrutinib can be a therapeutic agent for ischemic stroke by inhibiting the activity of Bruton’s tyrosine kinase (BTK) which promotes the inf lammasome activation. Thus, further investigation of inflammatory mechanisms will develop a novel therapeutic strategy for ischemic stroke.
Article
Full-text available
Monocyte chemoattractant protein-1 (MCP-1), a chemokine secreted by neurons and astrocytes following stroke is known to aggravate ischemia-related damage. Previous studies revealed that MCP-1-deficient mice develop smaller infarcts and have an improved neurological outcome, whereas mice overexpressing MCP-1 show worsened brain damage and impaired neurological function. The aim of the present study was to elucidate the molecular background of the enhanced recovery in MCP-1-deficient mice after stroke. For this purpose, we (1) performed expression analyses on crucial post-stroke related inflammatory genes in MCP-1-deficient mice compared to wildtype controls, (2) analyzed a possible impact of MCP-1 on astrocyte activation (3) investigated the cellular origin of respective inflammatory cytokines and (4) analyzed the impact of MCP-1 secretion on the migration of both neutrophil granulocytes and T-cells. Here we report that MCP-1-deficiency leads to a shift towards a less inflammatory state following experimental occlusion of the middle cerebral artery including an impaired induction of interleukin-6, interleukin-1β and granulocyte-colony stimulating factor expression as well as a subsequent diminished influx of hematogenous cells. Additionally, MCP-1-deficient mice developed smaller infarcts 36 hours after experimental stroke. Investigations revealed no differences in transcription of tumor necrosis factor-α and astrogliosis 12 and 36 hours after onset of ischemia. These novel results help to understand post ischemic, inflammatory mechanisms and might give further arguments towards therapeutical interventions by modulation of MCP-1 expression in post stroke inflammation.
Article
Full-text available
Post-ischemic inflammation is an essential step in the progression of brain ischemia-reperfusion injury. However, the mechanism that activates infiltrating macrophages in the ischemic brain remains to be clarified. Here we demonstrate that peroxiredoxin (Prx) family proteins released extracellularly from necrotic brain cells induce expression of inflammatory cytokines including interleukin-23 in macrophages through activation of Toll-like receptor 2 (TLR2) and TLR4, thereby promoting neural cell death, even though intracellular Prxs have been shown to be neuroprotective. The extracellular release of Prxs in the ischemic core occurred 12 h after stroke onset, and neutralization of extracellular Prxs with antibodies suppressed inflammatory cytokine expression and infarct volume growth. In contrast, high mobility group box 1 (HMGB1), a well-known damage-associated molecular pattern molecule, was released before Prx and had a limited role in post-ischemic macrophage activation. We thus propose that extracellular Prxs are previously unknown danger signals in the ischemic brain and that its blocking agents are potent neuroprotective tools.
Article
Full-text available
Ambivalent effects of interleukin-6 on the pathogenesis of ischaemic stroke have been reported. However, to date, the long-term actions of interleukin-6 after stroke have not been investigated. Here, we subjected interleukin-6 knockout (IL-6(-/-)) and wild-type control mice to mild brain ischaemia by 30-min filamentous middle cerebral artery occlusion/reperfusion. While ischaemic tissue damage was comparable at early time points, IL-6(-/-) mice showed significantly increased chronic lesion volumes as well as worse long-term functional outcome. In particular, IL-6(-/-) mice displayed an impaired angiogenic response to brain ischaemia with reduced numbers of newly generated endothelial cells and decreased density of perfused microvessels along with lower absolute regional cerebral blood flow and reduced vessel responsivity in ischaemic striatum at 4 weeks. Similarly, the early genomic activation of angiogenesis-related gene networks was strongly reduced and the ischaemia-induced signal transducer and activator of transcription 3 activation observed in wild-type mice was almost absent in IL-6(-/-) mice. In addition, systemic neoangiogenesis was impaired in IL-6(-/-) mice. Transplantation of interleukin-6 competent bone marrow into IL-6(-/-) mice (IL-6(chi)) did not rescue interleukin-6 messenger RNA expression or the early transcriptional activation of angiogenesis after stroke. Accordingly, chronic stroke outcome in IL-6(chi) mice recapitulated the major effects of interleukin-6 deficiency on post-stroke regeneration with significantly enhanced lesion volumes and reduced vessel densities. Additional in vitro experiments yielded complementary evidence, which showed that after stroke resident brain cells serve as the major source of interleukin-6 in a self-amplifying network. Treatment of primary cortical neurons, mixed glial cultures or immortalized brain endothelia with interleukin 6-induced robust interleukin-6 messenger RNA transcription in each case, whereas oxygen-glucose deprivation did not. However, oxygen-glucose deprivation of organotypic brain slices resulted in strong upregulation of interleukin-6 messenger RNA along with increased transcription of key angiogenesis-associated genes. In conclusion, interleukin-6 produced locally by resident brain cells promotes post-stroke angiogenesis and thereby affords long-term histological and functional protection.
Article
Full-text available
Helminths induce potent T helper 2 (TH2)-type immune responses that can mediate worm expulsion, but the role of this response in controlling the acute tissue damage caused by migrating multicellular parasites through vital tissues remains uncertain. We used a helminth infection model in which parasitic nematode larvae migrate transiently through the lung, resulting in hemorrhage and inflammation. We found that IL-17 initially contributed to inflammation and lung damage, whereas subsequent IL-4 receptor (IL-4R) signaling reduced elevations in IL-17 mRNA levels, enhanced the expression of insulin-like growth factor 1 (IGF-1) and IL-10 and stimulated the development of M2 macrophages, all of which contributed to the rapid resolution of tissue damage. These studies indicate an essential role for TH2-type immune responses in mediating acute wound healing during helminth infection.
Article
Full-text available
Ischemia and reperfusion-elicited tissue injury contributes to morbidity and mortality in a wide range of pathologies, including myocardial infarction, ischemic stroke, acute kidney injury, trauma, circulatory arrest, sickle cell disease and sleep apnea. Ischemia-reperfusion injury is also a major challenge during organ transplantation and cardiothoracic, vascular and general surgery. An imbalance in metabolic supply and demand within the ischemic organ results in profound tissue hypoxia and microvascular dysfunction. Subsequent reperfusion further enhances the activation of innate and adaptive immune responses and cell death programs. Recent advances in understanding the molecular and immunological consequences of ischemia and reperfusion may lead to innovative therapeutic strategies for treating patients with ischemia and reperfusion-associated tissue inflammation and organ dysfunction.
Article
Full-text available
Immunity and inflammation are key elements of the pathobiology of stroke, a devastating illness second only to cardiac ischemia as a cause of death worldwide. The immune system participates in the brain damage produced by ischemia, and the damaged brain, in turn, exerts an immunosuppressive effect that promotes fatal infections that threaten the survival of people after stroke. Inflammatory signaling is involved in all stages of the ischemic cascade, from the early damaging events triggered by arterial occlusion to the late regenerative processes underlying post-ischemic tissue repair. Recent developments have revealed that stroke engages both innate and adaptive immunity. But adaptive immunity triggered by newly exposed brain antigens does not have an impact on the acute phase of the damage. Nevertheless, modulation of adaptive immunity exerts a remarkable protective effect on the ischemic brain and offers the prospect of new stroke therapies. As immunomodulation is not devoid of deleterious side effects, a better understanding of the reciprocal interaction between the immune system and the ischemic brain is essential to harness the full therapeutic potential of the immunology of stroke.
Article
There is growing evidence that acute injury of the central nervous system, including stroke, impairs the immune system. A recent study implicates the postischemic activation of a particular type of lymphocyte, by noradrenergic signaling, as a mediator of impairment.
Article
Interleukin-6 (IL-6) has been shown to have a neuroprotective effect in brain ischemic injury. However, its molecular mechanisms are still poorly understood. In this study, we investigated the neuroprotective role of the IL-6 receptor (IL-6R) by IL-6 in the reactive oxygen species defense system after transient focal cerebral ischemia (tFCI). IL-6 was injected in mice before and after middle cerebral artery occlusion. Coimmunoprecipitation assays were performed for analysis of an IL-6R association after tFCI. Primary mouse cerebral cortical neurons were transfected with small interfering RNA probes targeted to IL-6Rα or gp130 and were used for chromatin-immunoprecipitation assay, luciferase promoter assay, and cell viability assay. Reduction in infarct volumes by IL-6 was measured after tFCI. IL-6R was disrupted through a disassembly between IL-6Rα and gp130 associated by protein oxidation after reperfusion after tFCI. This suppressed phosphorylation of signal transducer and activator of transcription 3 (STAT3) and finally induced neuronal cell death through a decrease in manganese-superoxide dismutase. However, IL-6 injections prevented disruption of IL-6R against reperfusion after tFCI, consequently restoring activity of STAT3 through recovery of the binding of STAT3 to gp130. Moreover, IL-6 injections restored the transcriptional activity of the manganese-superoxide dismutase promoter through recovery of the recruitment of STAT3 to the manganese-superoxide dismutase promoter and reduced infarct volume after tFCI. This study demonstrates that IL-6 has a neuroprotective effect against cerebral ischemic injury through IL-6R-mediated STAT3 activation and manganese-superoxide dismutase expression.