Neurological Sciences

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 21

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/316171830

Pathogenic mechanisms following ischemic stroke

Article  in  Neurological Sciences · April 2017


DOI: 10.1007/s10072-017-2938-1

CITATIONS READS

58 631

5 authors, including:

Seyed Esmaeil Khoshnam William Winlow


Ahvaz Jondishapour University of Medical Sciences University of Naples Federico II
37 PUBLICATIONS   162 CITATIONS    336 PUBLICATIONS   2,426 CITATIONS   

SEE PROFILE SEE PROFILE

Maryam Farzaneh Yaghoob Farbood


Royan Institute Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
18 PUBLICATIONS   147 CITATIONS    74 PUBLICATIONS   886 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

M. Sc Disertatios of my students. View project

The Effects of Hydroalcoholic Extract of Raspberry fruit on Pain and Passive Avoidance Memory Following Transient Global Ischemia/Reperfusion in Wistar Rats View
project

All content following this page was uploaded by Seyed Esmaeil Khoshnam on 23 September 2017.

The user has requested enhancement of the downloaded file.


Neurol Sci (2017) 38:1167–1186
DOI 10.1007/s10072-017-2938-1

REVIEW ARTICLE

Pathogenic mechanisms following ischemic stroke


Seyed Esmaeil Khoshnam 1 & William Winlow 2,3 & Maryam Farzaneh 4 &
Yaghoob Farbood 1 & Hadi Fathi Moghaddam 1

Received: 28 November 2016 / Accepted: 25 March 2017 / Published online: 17 April 2017
# Springer-Verlag Italia 2017

Abstract Stroke is the second most common cause of death pathophysiology of ischemic stroke and reveal the intertwined
and the leading cause of disability worldwide. Brain injury pathways that are promising therapeutic targets.
following stroke results from a complex series of pathophys-
iological events including excitotoxicity, oxidative and Keywords Ischemia . Stroke . Oxidative stress .
nitrative stress, inflammation, and apoptosis. Moreover, there Inflammation . Apoptosis
is a mechanistic link between brain ischemia, innate and adap-
tive immune cells, intracranial atherosclerosis, and also the gut
microbiota in modifying the cerebral responses to ischemic Introduction
insult. There are very few treatments for stroke injuries, partly
owing to an incomplete understanding of the diverse cellular Stroke occurs due to disruption in the blood supply to a brain
and molecular changes that occur following ischemic stroke region, resulting in death or permanent neurological deficits
and that are responsible for neuronal death. Experimental dis- [1]. It is the second leading cause of death and the major cause
coveries have begun to define the cellular and molecular of adult physical disability in the world [2]. Neurological def-
mechanisms involved in stroke injury, leading to the develop- icits which occur following stroke include balance problems,
ment of numerous agents that target various injury pathways. hemiplegia, loss of sensory and vibratory sensation, numb-
In the present article, we review the underlying ness, decreased reflexes, ptosis (of the eyelid), visual field
defects, aphasia, and apraxia [3]. Stroke accounts for 44 mil-
lion physical disabilities annually, with 5.5 million deaths
worldwide. There are very few treatments for stroke, and the
development of new therapeutic agents or combined therapies
* Seyed Esmaeil Khoshnam
Esmaeil.khoshnam1392@gmail.com is imperative. At present the only effective treatment for stroke
is limited to recombinant tissue plasminogen activator (tPA)
* William Winlow
bill.winlow@gmail.com [4]. tPA is the only appropriate thrombolytic agent available
for stroke treatment [5, 6]. However, due to a narrow time
window, this treatment is only suitable for a minority
1
Department of Physiology, Faculty of Medicine, Physiology (<10%) of stroke patients [7].
Research Center, Ahvaz Jundishapur University of Medical
Sciences, Ahvaz, Iran
2
Dipartimento di Biologia, Università degli Studi di Napoli, Federico
II, Via Cintia 26, 80126 Napoli, Italy
Ischemic stroke pathogenesis
3
Honorary Research Fellow, Institute of Ageing and Chronic
Diseases, The APEX building, 6 West Derby Street, University of
Stroke can be classified as ischemic or hemorrhagic stroke,
Liverpool, Liverpool L7 8TX, UK depending on the underlying pathology [8]. Ischemic stroke
4
Department of Stem Cells and Developmental Biology, Cell Science
accounts for 85% of all strokes, while hemorrhagic stroke is
Research Center, Royan Institute for Stem Cell Biology and responsible for 15% of them [9]. Cerebral ischemia results
Technology, ACECR, Tehran, Iran from cerebral artery occlusion obstructing blood flow to a
1168 Neurol Sci (2017) 38:1167–1186

portion of the brain [10]. According to the etiologies, ischemic sodium ions [27], but is not affected by removal of calcium
stroke can be further classified into four main categories as ions [28]. Furthermore, delayed neuronal cell death that oc-
follows: small vessel disease, athero–thrombotic, cardio–em- curs following glutamate treatment is abolished by calcium
bolic, and undetermined causes [8]. removal and potentiated by increased calcium concentrations
Following ischemic stroke, neurons are deprived of oxygen in the bathing medium [29]. Moreover, the initial calcium
and energy with detrimental effects on energy-dependent pro- influx is known to trigger a secondary intracellular, toxic cal-
cesses in neuronal cells [11]. Immediately after ischemia, neu- cium overload, which is strongly correlated with neuronal
rons are unable to sustain their normal transmembrane ionic death [30]. Thus, calcium ions are essential to glutamate
gradient and homoeostasis. This elicits several processes that neurotoxicity.
lead to cell death: excitotoxicity, oxidative and nitrative stress,
inflammation, and apoptosis. These pathophysiological pro- Glutamate receptors: the gateway to excitotoxicity
cesses are seriously injurious to neurons, glia, and endothelial
cells [12–15] and are interlinked, triggering each other in a Several types of ionotropic and metabotropic glutamate recep-
positive feedback loop that terminates in neuronal destruction tors have been identified in the central nervous system
[16]. [31–33]. Activation of the N-methyl-D-aspartate (NMDA)
A spectrum of severity is observed in the affected region of and a-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
the brain, owing to differential lessening of blood supply to (AMPA) type of glutamate receptors leads to membrane de-
different zones. Thus, part of the brain tissue (core) undergoes polarization and facilitates influx of calcium ions into neurons,
irreversible neuronal damage due to necrotic cell death, while which may lead to excitotoxicity [34–36]. There is good evi-
the surrounding tissues contain salvageable and metabolically dence that NMDA receptors (NMDARs) are responsible for
active cells (penumbra), in which cell death occurs less rapid- the neuronal damage after ischemic stroke, and these receptors
ly. Most therapeutic methods established in the laboratory are the most calcium-permeable ionotropic glutamate recep-
have focused on safeguarding neurons from the main patho- tors [24]. Several studies have revealed that inhibition of the
physiological processes, such as inflammation and apoptosis, NMDARs and AMPA receptors (AMPARs) prevent calcium
that might otherwise occur in the penumbra, and this area is entry, resulting in neuro-protective effects in models of focal
the target for therapeutic agents [17–20]. ischemia [37].
Expression of the sodium–calcium exchanger (NCX) gene
is influenced by cerebral ischemia, which is an important reg-
Postischemic excitotoxicity ulator of intracellular calcium and sodium homeostasis in the
brain [38]. Following ischemia, activation of the NCX could
The brain oxygen consumption relative to its weight is very ameliorate the consequences of ischemic brain injury [39]. It
high (approximately 20% of the body’s oxygen) and must has been demonstrated that NCX dysfunction is mediated by
generate sufficient ATP through mitochondrial electron trans- NMDARs, which explains the subsequent calcium overload
port chain to maintain and restore ionic gradients [21]. At a following the excitotoxic stimulus [40]. During excitotoxicity,
cellular level, ischemia causes failure of oxidative phosphor- increasing the mitochondrial calcium concentration leads to
ylation and ATP synthesis, which leads to ATP being con- the production of reactive oxygen species (ROS) [41], mito-
sumed within 2 min. This adversely affects Na+/K+ ATPase chondrial depolarization via opening of the permeability tran-
pump, resulting in plasma membrane depolarization, release sition pore [42], induction of calcium deregulation [43], and
of potassium into the extracellular space, and entry of sodium finally induction of neuronal death [44]. Moreover, during
into cells [22]. In addition, the Ca2+ pump also fails [23] caus- excitotoxicity, the mitochondrial NCX (mNCX) partially con-
ing a dramatic rise in intracellular calcium concentration dur- tributes to delayed cytoplasmic calcium loading [45, 46]. The
ing ischemia. This activates several death-signaling proteins essential role of the NMDAR in calcium-mediated death sig-
such as calcium-dependent proteases, lipases, and DNases naling led to the theory that some calcium-dependent death-
resulting in cell death in the ischemic core [21]. signaling proteins must be closely associated with the
One reason for the particular susceptibility of the brain to NMDAR [47].
ischemic injury is that brain tissue contains high levels of Metabotropic glutamate (mGlu) receptors also contribute
glutamate, and many neurons in the brain are activated by to excitotoxicity. mGlu receptors belong to the G-protein
glutamate receptors. Following a stroke, glutamate is critical coupled receptor (GPCR) family of receptors that modulate
for neuron degeneration [24] when it acts as a neurotoxic excitatory synaptic transmission. There are three groups of
excitatory neurotransmitter playing a pivotal role in ischemia metabotropic glutamate receptors, (groups I, II, and III
via excitotoxic pathogenesis [25, 26]. In vivo, it has been mGluRs); group I mGluR receptors comprise mGluR1 and
shown that the rapid neuronal swelling that occurs after glu- mGluR5, which are predominantly found at the postsynaptic
tamate treatment is entirely abolished by removal of external membrane of glutamatergic synapses where they increase
Neurol Sci (2017) 38:1167–1186 1169

neuronal excitability by modulating NMDA and AMPA re- injury induced by ischemic stroke [66, 68]. Thus, the NR2B
ceptors [48]. There is substantial evidence that in vivo subunit is a major hub for NDC formation [68–70].
mGluR1 activation is protective against NMDA-induced Pro-survival effects of synaptic NMDAR result in activa-
excitotoxicity [40]. Several studies reported that the neuropro- tion of the CREB (cyclic-AMP response element-binding pro-
tective effects of group I mGluRs were mediated by the acti- tein) signaling pathway while pro-death effects of extra syn-
vation of the PI3K-Akt signaling pathway [49–51]. Activation aptic NMDARs are mediated by diminishing the activity of
of other mGlu receptors (Group II and group III) by their the CREB pathway [67]. It is interesting to note that
specific agonists demonstrates neuroprotective effects by lim- memantine, a low affinity uncompetitive NMDAR antagonist,
iting induction of excitotoxicty. These mGlu receptors are has neuroprotective effects in stroke models [71]. Also, in a
mainly found at presynaptic terminals where they inhibit the mouse model of Huntington’s disease, memantine can selec-
release of glutamate [52]. Following ischemic stroke, inhibi- tively block extra synaptic NMDARs, thereby protecting
tion of expression of the glutamate transporter-1 (GLT-1) pro- against neuronal death [72]. The NR2B–PSD95–nNOS sig-
motes excitotoxicity [53]. Moreover, postischemic downreg- naling complex was identified to be the primary set of death-
ulation of GLT-1 is closely associated with glutamate accumu- signaling proteins [69, 73, 74]. PSD95 is a synaptic scaffold-
lation and promotion of excitotoxicity, suggesting that ing protein that binds to carboxyl terminus of the NMDAR
excitotoxicity could be controlled by GLT-1 expression [54]. NR2B subunit (NR2B-CT) and nNOS which results in the
production of the neurotoxic molecule nitric oxide (NO)
Dual roles of NMDARs in neuronal survival and death [73–75], and suppression of nNOS following stroke led to
inhibition of NMDAR-dependent neuronal death [74].
The mechanisms participating in stroke injury are possibly Following a stroke insult, excitotoxic neuronal death me-
multifactorial [55], with NMDAR-mediated excitotoxicity be- diated by the NMDARs is not only limited to the NDC but
ing a key factor [55, 56]. Many important neurological func- also involves further death-signaling proteins, such as the
tions require normal NMDAR activity, including neuronal calpains (a family of calcium-activated cysteine proteases)
plasticity, brain formation, and neuronal survival [57, 58]. which play a major role in turning the calcium influx via the
However, NMDAR over-activation results in excitotoxicity NMDAR into neuronal damage [62, 63, 76, 77]. Calpains are
and neuronal death [55, 56]. A number of studies have found only activated by extra synaptic activation of the NR2BRs
that NMDAR blockers have neuro-protective effects against [63, 69, 77, 78], so in vitro and in vivo inhibition of these
ischemic damages both in vitro [56, 59] and in vivo [59, 60]. proteins mediates strongly neuroprotective effects against
Subfamilies of NMDAR are multimeric assemblies whose NMDAR-mediated neuronal injury [76, 78, 79].
subunits include those classed as NR1, NR2, and NR3. In the
cytoplasmic terminus, different NR2 subunits have structural
diversity that couple the receptor to different signaling ma- Postischemic oxidative and nitrative stress
chineries [57, 61]. In the stroke and traumatic models,
NR2AR and NR2BR subunits are necessary for glutamate- Oxidative and nitrative stress occurs when the production of
mediated neuronal survival and death [59, 60, 62–64]. It has free radicals such as reactive oxygen/nitrogen species (ROS/
been suggested that inhibition of NR2BR subunits protects the RNS) overpowers the endogenous scavenging capacity of an-
ischemic brain from excitotoxic and neuronal cell death [65]. tioxidant defenses systems [80]. Substantial experimental ev-
Excessive activation of NMDARs contributes to neuronal idence indicates that in the all forms of stroke injuries free
death following stroke [66]. During basal synaptic transmis- radical formation was increased [81, 82], and both ROS and
sion, activation of the synaptic NMDAR (predominantly RNS molecules have been clearly shown to be important me-
NR2A-containing) stimulates the signaling components of diators of tissue injury in acute ischemic stroke [83–85].
the neuronal survival signaling complex (NSC) that promote During ischemia, several mechanisms lead to the free radical
neuronal survival [59, 67]. However, under pathological con- formation, including NMDAR-mediated excitotoxicity, ex-
ditions such as stroke, elevation of the extracellular glutamate cessive of Ca2+ influx, mitochondrial dysfunction, and neuro-
concentration causes excitotoxic activation of extrasynaptic nal nitric oxide synthase (nNOS) activation [86–90].
NMDARs (predominantly NR2B-containing). The NR2B ac- Excessive ROS cause the devastation of cellular macromole-
tivation increased Ca2+ influx and promotes aDAPK (active cules and contribute to the signaling processes that result in
death-associated protein kinase) to bind with NR2B [59, 67, apoptotic cell death [91].
68]. aDAPK recruitment activates the neuronal death- After ischemic stroke, energy depletion causes lactic acid
signaling complex (NDC) that in turn suppresses synaptic to accumulate and this leads to acidosis in the neurons. The
NSC activity [67] and mediates neuronal death. It is demon- acidosis has a pro-oxidant effect via increasing H+ concentra-
strated that inhibition of aDAPK binding to the NR2B reduces tions, increasing the rate of superoxide anion (O−2) conversion
activation of NDC and prevents the excitotoxic neuronal to hydrogen peroxide (H2O2), and the hydroperoxyl radical
1170 Neurol Sci (2017) 38:1167–1186

(HO2) [92]. Unlike other organs, the brain is especially vul- by free radical scavengers and antioxidant enzymes [83, 106].
nerable to free radical-mediated attack due to the highly oxy- The activity of antioxidant and detoxifying enzymes maintain
genated environment of neurons, coupled with highly redox homeostasis. These enzymes include superoxide dis-
peroxidisable lipids and low levels of endogenous antioxi- mutase (SOD), glutathione peroxidase (GSHPx), glutathione
dants [92, 93]. reductase, and glutathione-S-transferase (GST), and their ac-
Stroke risk factors and ischemia lead to the activation of tivity is studied in ischemic stroke progression [107, 108]. The
several ROS-generating enzymatic systems including SOD enzymes including manganese SOD (MnSOD) and ex-
NADPH oxidase (NOX) [94, 95] mitochondrial depolariza- tracellular SOD (EcSOD) assist in brain recovery following
tion [96], xanthine oxidase (XO) [97], and nitric oxide syn- ischemic reperfusion damage [109, 110].
thase (NOS) [98]. Following ischemia and NMDAR activa- Antioxidant enzymes are encoded by specific genes that
tion, NADPH oxidase produces the majority of superoxide bear an antioxidant response element (ARE) within their pro-
anions [94, 95]. It has been shown that following activation moters. The activation of this element is mainly regulated by
of NMDARs the mitochondrion ceases to be the major source nuclear factor erythroid-2 related factor 2 (Nrf2) [111].
of free radicals. However, the proximity of neuronal mito- Postischemic oxidative stress induces antioxidant responses
chondria to the NADPH oxidase pathway increases the prob- that are mediated by Nrf2, and this factor is neuroprotective
ability that this enzyme produces Bkindling^ ROS that elevate against stroke injuries [112, 113]. It has been found that ex-
mitochondrial uncoupling, initiating a secondary ROS surge pression of Nrf2 is upregulated in the penumbra, indicating
from mitochondria [94]. the value and contribution of this factor in neuronal protection
ROS have substantial cellular effects, causing tissue dev- and survival [114]. Nrf2 regulates several downstream protec-
astation and cell death by processes including DNA damage, tive proteins, including glutathione-S transferase (GST),
protein destruction, lipid peroxidation, release of Ca2+ from NAD(P)H: quinone oxidoreductase-1 (NQO1) and
intracellular stores, cytoskeletal structural injury, and chemo- hemeoxygenase-1 (HO-1). These proteins are all involved in
taxis. Some of these effects lead to the activation of antioxi- cytoprotection against various oxidative insults in the brain
dant defense mechanisms to counterbalance the ROS devas- [115, 116]. Thus, Nrf2 is located in the center of the neuro-
tation [82]. Furthermore, ROS have profound effects on the protective pathway [117]. Studies have demonstrated that
cerebral vasculature which ultimately influence cerebral blood Nrf2 also contributes to protective effects against mitochon-
flow. Hence, the H2O2, O2, and ONOO exert their effects by drial dysfunction, which occur during ischemic stroke [118].
increasing vasodilation, aggregation of the platelets, increased
endothelial permeability, and focal lesions in endothelial cells
[99]. Postischemic inflammation
The NO molecule which is generated by the ischemia-
activated NOS combines with superoxide to produce Inflammation is a critical step in the pathophysiology of cere-
peroxynitrite, a potent oxidant [23]. RNS have important cel- brovascular diseases, especially ischemic stroke [119]. Many
lular effects, such as facilitating the opening of the mitochon- studies have indicated that postischemic neuro-inflammation
drial permeability transition pore (mPTP), inhibition of impor- is an important factor for ischemic long-term prognoses [120,
tant mitochondrial enzymes, DNA damage, and activation of 121]. Brain inflammation is a therapeutic target for some brain
the transient receptor potential cation channel, subfamily M, diseases such as ischemia and trauma [122]. Following brain
member 7 (TRPM7) channels that are permeable to the Ca2+ ischemia, inflammatory responses are initiated as a result of
ions [100, 101]. Also, NO modifies protein groups through several agents such as ROS formation, necrotic cells, and im-
post-translational alterations that affect cell survival by chang- paired tissues. These agents cause activation of inflammatory
ing the functions of critical proteins such as caspases, cells [123–126]. Moreover, these activated inflammatory cells
metalloproteases, and the glycolytic enzyme GAPDH [102, participate in tissue remodeling after cerebral injuries [127].
103]. In particular, NO by covalently attaching to cysteine The inflammatory process involves several different cell
residues forms S-nitrosothiol derivatives, which impact pro- types, inflammatory cytokines, and cellular receptors such as
tein function, thus affecting S-nitrosylation, but this aspect is toll-like receptors (TLRs) (Fig. 1).
underexplored in ischemic mechanisms [20].
Despite the deleterious effects of reactive radical overpro- The cellular neuro-inflammatory response
duction which are destructive to brain cells, at homeostatic
levels, these radicals are required signaling molecules contrib- Following brain injury, the initial inflammatory response is
uting to normal neuronal function [104]. Therefore, it is im- mediated by the activation and recruitment of microglial cells
portant that novel therapies only scavenge deleterious radicals [129–131]. Microglial cells are the resident macrophages of
without interfering with endogenous signaling [105]. The ef- the brain, and they are highly activated after brain insult [132].
fects of free radical injuries are normally inhibited or lessened Studies have demonstrated that suppression of the microglial
Neurol Sci (2017) 38:1167–1186 1171

Fig. 1 Schematic of the effects of cerebral ischemia on the inflammatory from ischemic neurons recruits NK cells into the ischemic zones [128].
cytokines and immune cells and their contribution to neurotoxicity. These NK cells affect ischemic neurons by releasing cytokines, mainly
Cerebral ischemia causes lessening of blood supply in the penumbra TNF, that increase glutamate release and result in neuronal hyperactivity
zone, and triggers a complex cascade of events that include and excitotoxicity [128]. NK cells also release cytokines, such as IFN-γ
excitotoxicity and oxidative stress in the ischemic neurons. These and GM-CSF, that activate microglia and macrophages and condition
cascades lead to microglial activation and release of pro-inflammatory astrocytes, which in turn secrete inflammatory mediators such as IL-1β,
cytokines such as TNF-α, IL-1β, and IL-6. These cytokines can potenti- IL-6, and NO [128]. Thus, excitotoxicity and inflammation induced by
ate inflammation by recruitment and infiltration of the neutrophils, mono- microglia and NK cells contribute to neurotoxicity and apoptotic cell
cytes, and T cells into the brain lesion. After core zone ischemia, neurons death. Abbreviations: DAMPs damage-associated molecular patterns,
undergo irreversible neuronal damage due to necrotic cell death. DAMPs TLRs toll-like receptors, NK cells natural killer cells, TNF tumor necrosis
released by necrotic cells are detected by receptors such as TLRs, which factor, GM-CSF granulocyte macrophage colony-stimulating factor, NO
are expressed by immune cells such as NK cells, which are used as an nitric oxide
example of lymphocytes. In the early stages of stroke, fractalkine released
1172 Neurol Sci (2017) 38:1167–1186

cells reduced postischemic damage, and these cells may be- hyperactivity, and finally catalyze neuronal death [128, 156].
come part of an attractive therapeutic strategy for ischemic Data from experimental models showed that lymphocytes in-
stroke [133, 134]. Ischemia activates microglia and transform filtrate the brain immediately after ischemic stroke, with neu-
them into the phagocytic cells that release a variety of cyto- trophils migrating into the brain parenchyma immediately af-
toxic and/or cytoprotective substances. The neuroprotective ter the initial injury, followed by macrophages and natural
effects of microglia are exerted by generating neurotrophic killer cells [128, 157–159]. Subsequently, T and B lympho-
factors such as insulin-like growth factor I (IGF-I) and brain- cytes enter the lesion [157].
derived neurotrophic factor (BDNF). In response to the ische- In ischemic brain tissue, ROS induce the expression of the
mia, activated microglial cells distribute pro-inflammatory cy- pro-inflammatory genes such as nuclear factor kappa B (NF-
tokines including tumor necrosis factor α (TNF-α), interleu- kB), interferon regulator factor 1 (IRF 1), hypoxia inducible
kin-1β(IL-1β), interleukin-6 (IL-6), as well as other potential factor 1 (HIF 1), and STAT3. Thus, these inflammatory factors
cytotoxic molecules such as prostanoids, ROS, and NO increase the expression of cytokines and also of adhesion
[135–137]. While the initial purpose of microglial activation molecules. The adhesion molecules are upregulated in the first
is the brain’s effort to protect neurons, the over-activation of few days following ischemic stroke, and they are responsible
microglia results in deleterious inflammation, increasing the for the leukocyte migration through the brain endothelium
probability of neuronal death [138–140]. [134, 160]. Neutrophils also infiltrate the brain facilitated by
Previously, it was accepted that blood brain barrier (BBB) disruption of the BBB disruption, and it has been mediated by
prevented blood inflammatory cells (monocytes and neutro- cell adhesion molecules including selectins, integrins, and im-
phils) from entering the brain and the predominant opinion munoglobulins [161]. When neutrophils infiltrate the ische-
was that only microglial cells were involved in inflammation mic brain, they release oxygen-free radicals and proteolytic
of the brain. However, infiltration of neutrophils and mono- enzymes that damage to the tissue. However, in animals, de-
cytes into spinal cord injury sites has been described and their ficient in protein kinase C infarct volumes are reduced, be-
contribution to the inflammation has been demonstrated cause protein kinase C has significant roles in neutrophil ad-
[141]. Also, an increase in neutrophil and monocyte counts hesion, degranulation, and superoxide generation [161, 162].
has been reported in transient ischemia and in stroke. [142]. While the initial infiltration of immune cells into the ischemic
Leukocyte transmigration through BBB appears to be regulat- brain exacerbates tissue damage and worsening of neurologi-
ed by CAMs (ICAM-1, VCAM-1) and chemokine signaling cal deficits, there is a subsequent beneficial effect, including
processes [143]. Moreover, astrocytes and even neurons con- the release of cytokines that promote glial scar formation ac-
tribute to brain inflammation, and these cells produce several companied by phagocytosis of cellular debris that is essential
anti-inflammatory factors and chemokines that recruit mono- for effective wound healing [23].
cytes [144–149]. Inflammation is regulated both negatively
and positively by neuronal cells [150–152], and the number Cytokines and the neuro-inflammatory response
of circulating monocytes is increased following stroke.
Neutrophilia and lymphocytopenia have also been observed Cytokines are key players in the inflammatory mechanism and
in cerebral ischemic patients [142]. The distinct aspects of contribute stroke-related brain injury [163]. During ischemia,
ischemic stroke are characterized by neuronal necrosis and pro-inflammatory cytokines and chemokines appear to exac-
excess infiltration of immune cells [20, 153]. Hence, brain erbate stroke-related brain injury. The major pro-
inflammation is a complex process in which several immune inflammatory cytokines are TNF-α, IL-1β, IL-6, and
cells play a role. chemokines. However, anti-inflammatory cytokines such as
Astrocytes, similar to the microglial cells, could deliver IL-10 and TGF-β may have neuroprotective effects [161,
inflammatory factors such as chemokines, cytokines, and 164, 165]. Pro-inflammatory and anti-inflammatory cytokines
NO [154]. Furthermore, there are some cytokines delivered are secreted from different cells in the brain, including microg-
from several peripheral cells such as T lymphocytes, mono- lia, astrocytes, endothelial cells, and neurons [155]. In animal
nuclear phagocytes, natural killer (NK) cells, and poly morph models, it has been demonstrated that there is an increase in
nuclear leukocytes, and they are involved in the ischemic in- the production of pro-inflammatory cytokines and a lessening
flammation [155]. NK cells are innate lymphocytes that can in production of anti-inflammatory cytokines associated with
be rapidly mobilized in the primary phases of immune re- increasing infarct size [166].
sponses. During stroke, NK cells infiltrate ischemic lesions The use different ischemia models has resulted in the ob-
of the brain. Following cerebral ischemia, fractalkine is de- servation that production of IL-1 is increased after permanent
rived from the ischemic neurons, which subsequently recruit or transient cerebral ischemia in microglia, astrocytes, and
NK cells, and determine the size of brain lesions in a T and B neurons [161]. Calcium entry increases due to the actions of
cell-independent manner. Also, NK cells exacerbate the brain IL-1 on the NMDARs, resulting in neuronal cell death [167].
infarction by increasing local inflammation and neuronal Furthermore, IL-1 can potentiate inflammation by recruitment
Neurol Sci (2017) 38:1167–1186 1173

and adhesion of neutrophils. IL-1 has been shown to increases NF-kB is a heteromeric transcription factor widely known
leukocyte infiltration by increasing the expression of adhesion to be associated with inflammatory responses following ische-
molecules such as E-selectin, ICAM-1, ICAM-2, and VCAM- mia [191–193]. It is involved in activation of several pro-
1 on the endothelial cells and causing breakdown of the blood inflammatory genes including TNF-α, ICAM-1, IL-6, iNOS,
brain barrier [161, 168–170]. Adhesion molecules such as and cyclooxygenase 2 (COX2) [194, 195]. Several studies
ICAM-1 and VCAM-1 are the major ligands for leukocyte have revealed that inhibition of the NF-kB pathway prevents
integrins to attach to endothelial cells. Under normal condi- production of pro-inflammatory cytokines [196–198].
tions, ICAM-1 is detected on a small number of CNS Matrix metalloproteinases (MMPs) are a family of proteo-
microvessels and massively upregulated by inflammatory lytic enzymes that are responsible for remodeling the extracel-
stimuli [171]. lular matrix and that can degrade all its components.
It has also been shown that administration of recombinant Expression of MMPs is increased in the brain in response to
IL-1 receptor antagonists decreases tissue necrosis and infarct injury [199]. Following injury, expression of MMPs has been
volume [172], and mouse models with IL1-αβ knock-out shown in some cells such as neurons, astrocytes, microglia,
have smaller infarct volumes than controls [173]. and endothelial cells. Experimental studies have shown that
Furthermore, intra-cerebroventricular administration of IL- MMPs such as MMP-9 and MMP-2 are responsible for dis-
1β increased infiltration of neutrophils into the brain, as well ruption of BBB and hemorrhagic transformation following
as increasing cerebral edema and infarct size [161]. ischemic stroke [200–202]. MMP-9 is associated with
In rats subjected to experimental ischemia, IL-6 is elevated neuro-inflammation, and its inhibition could have a beneficial
within 3–3.5 h of stroke, with peak expression at 12 h and effect on the outcome of stroke [203].
continuing for at least 24 h [174]. IL-6 is a pro-inflammatory
cytokine whose serum levels correlate with larger infarct size The TLRs and the neuro-inflammatory response
and poorer clinical outcome [161, 175]. However, following
stroke, IL-6 signaling is important for tissue remodeling and Activation of toll-like receptors (TLRs) leads to cytokine and
recovery via activation of transcription factor signal transduc- chemokine production, which initiates the inflammatory re-
er and activator of transcription (STAT) [176–178]. In addi- sponse. In the central nervous system, TLRs are expressed
tion, IL-6 demonstrates anti-inflammatory properties by in- on endothelial cells, microglia, astrocytes, oligodendrocytes,
ducing IL-1 receptor antagonist synthesis, inhibiting TNF-α, and neurons [204–207]. Thirteen TLRs have been identified,
and inducing apoptosis in neutrophils [23, 179]. However, IL- and TLR4 signaling contributes to postischemic inflammatory
6 also increases brain damage by elevating body temperature injuries [208, 209]. In response to hypoxia, TLR4 expression
after stroke [180]. Thus, IL-6 has both beneficial and detri- is upregulated on the surface of microglial cells [210].
mental effects in the context of stroke. After cerebral ischemia in mice, TLR4 mRNA expression
Several studies demonstrate a positive correlation between increased between 1 and 22 h post-stroke [211]. Furthermore,
TNF-α and the degree of ischemic damage [181]. Hence, in TLR2 mRNA expression also increased following brain is-
rat ischemic models, administration of anti-TNF antibody and chemia [212]. Endogenous ligands such as Hsp70 are upreg-
TNF-α binding protein decreases infarct volume [182, 183]. ulated in the brain tissue following ischemia and can cause a
TNF-α promotes neutrophil infiltration into the brain by in- detrimental response to cerebral ischemia by TLR activation.
ducing expression of adhesion molecules in cerebral endothe- In animals missing either TLR2 or TLR4, there are smaller
lial cells. Additionally, TNF-α disrupts the BBB and stimu- infarct volumes than in wild-type animals following experi-
lates the induction of other inflammatory mediators [184, mental ischemia [213]. In TLR4 and TLR2 knockout models,
185]. lesion size is reduced between 1 and 3 days after stroke [211,
IL-10 is an anti-inflammatory cytokine that can inhibit pro- 214]. TLRs can activate NF-κB which induces the expression
inflammatory cytokine production and chemokine secretion of pro-inflammatory genes, cytokines, and adhesion mole-
and prevents antigen delivery by macrophages and microglia cules [215].
[186]. Moreover, IL-10 can neutralize the deleterious effects
of TNF-α by preventing activation of signaling pathways trig-
gered by TNF-α [187]. Also, TGF-β is a neuroprotective Innate and adaptive immune responses following
factor against neuro-inflammation [188] and it has neuropro- stroke
tective effects against ischemic insult [161, 189]. Moreover,
administration of TGF-β reduces the number of circulating In response to injuries, the first line of defense is the innate
neutrophils, which improves postischemic injury [190]. immune system in which macrophages, neutrophils, dendritic
After ischemic stroke, chemokines attract neutrophils to ische- cells, natural killer cells, and γδT cells are involved in the
mic tissue, and blocking these chemokines decreases cerebral innate immune response to cerebral damage. Following ische-
edema and infarction [161]. mic brain injury, a variety of danger signals are released from
1174 Neurol Sci (2017) 38:1167–1186

intracellular compartments or generated by the action of lytic T cells have differential effects on stroke outcome, which have
enzymes escaping from dead cells on matrix proteins, which been associated with increased inflammatory damage and
are usually called danger-associated molecular pattern mole- even worse outcomes [158, 187, 238, 239]. Studies showed
cules (DAMPs) [216]. These signals include purines (ATP, that following transient cerebral ischemia, B cell deficiency
UTP, and their catabolites), cytokines, and chemokines, caused further deterioration of histological damage and func-
among other molecules. DAMPs activate their receptors, in- tional outcomes, while adoptive transfer of B cells into these
cluding TLRs and scavenger receptors, on microglia, astro- mice improved neurological function and reduced ischemic
cytes, perivascular macrophages, and brain endothelial cells infarct size. The authors also showed that IL-10 was respon-
[217–221]. These signals activate microglia that then release sible for the B cell-mediated neuroprotection [240]. Therefore,
inflammatory cytokines, such as IL-1β and TNF-α. Also, in cerebral ischemia/reperfusion injury, the role of B lympho-
activated microglia induce release of chemokines, which in- cytes might be protective rather than pathogenic.
clude CCL2 and CCL3, to recruit leukocyte infiltration [222].
Following cerebral ischemia, the CCL2 and receptor CC che- The gut microbiota and ischemic stroke
mokine receptor 2 (CCR2) axis is responsible for the recruit-
ment of macrophages [223]. Ischemia also activates macro- Recent studies demonstrated that antibiotic-induced alter-
phages that are present in the perivascular space, and which ations in the intestinal flora reduce ischemic brain injury in
release pro-inflammatory cytokines, chemokines, ROS, and mice. Intestinal dysbiosis causes an increase in regulatory T
NO [224, 225]. Cellular adhesion molecules, including cells and a reduction in IL-17+ γδ T cells [241]. Hence, large
ICAM-1, P-selectin, and E-selectin, are expressed in the in- stroke lesions cause gut microbiota dysbiosis via stress-
flamed endothelial cells of ischemic brain. Adhesion mole- mediated intestinal paralysis, which in turn affects stroke out-
cules appear to be involved in the trans-endothelial migration come via changes in T cell homeostasis, induction of a pro-
of neutrophils into the brain. Following acute stroke, neutro- inflammatory response, and deterioration of stroke outcome.
phil infiltration and infarct volumes were reduced in ICAM-1- Therapeutic transplantation of fecal microbiota to normalize
deficient or P-selectin-deficient mice compared with wild- poststroke dysbiosis normalizes the brain lesion and improves
type mice [226–228]. Many studies have highlighted that fol- stroke outcome. Therefore, a target of stroke-induced systemic
lowing cerebral ischemia, activated neutrophils greatly con- alterations is the gut microbiome, which is an effector with
tribute to elevation of MMP-9 [229], which, in turn, causes substantial impact on stroke outcome [242]. Also, several
BBB breakdown, stimulates peripheral leukocyte infiltration, studies describe changes in intestinal microbiota composition
and ultimately results in neuronal damage [230]. Therefore, in stroke patients [243, 244]. Recent studies show that consid-
inhibition of neutrophils could be a reasonable peripheral tar- erable numbers of T cells migrate from the intestine to the
get for stroke treatment. poststroke brain [242]. The gut microbiota is a key regulator
Adaptive immunity that requires a relatively long time to of T cell homeostasis [245]. After the first few days of stroke
become protective is an antigen-specific defense mechanism, insult, T cells are recruited to the injured brain [246].
which includes the T and B lymphocytes that are related to the
post-injury inflammatory response. Antibodies against brain-
derived antigens develop following ischemic stroke, leading Post-ischemic apoptosis
to the sensitization of circulating T cells against brain antigens.
These antigens can be processed by antigen-presenting cells Apoptosis is involved in the pathogenesis of ischemic stroke
(APC), such as dendritic cells, with MHC molecules on the and is known as programmed cell death. It is activated during
cell surface. T cell receptors can recognize the MHC and development and physiological cellular turnover and in path-
brain–antigen complex and activate the adaptive immune sys- ological conditions such as stroke [247–249]. The apoptotic
tem. In rodent and human stroke, APC numbers are increased intrinsic stimuli are activated by the mitochondrial signaling
in the ischemic brain and reduced in the periphery [158, pathways, while extrinsic stimuli trigger cell surface death
231–233]. Following stroke, both CD4+ T-helper cells and receptors such as TNF-α, Fas (CD95/APO1), and TRAIL
larger numbers of CD8+ cytotoxic T cells are recruited into (TNF related apoptosis inducing ligand) receptors [250, 251].
the ischemic brain [158, 234]. The CD4+ helper T cells help to During ischemia, activation of NMDARs leads to accumu-
activate and direct other immune cells, including CD8+ cyto- lation of the intracellular Ca2+, causing cleavage of Bcl-2
toxic T cells, which are capable of directly inducing cell death interacting domain (BID) to truncated Bid (tBid) [252]. At
[235]. The regulatory T cells (Treg) exert their neuroprotective the mitochondrial membrane, tBid interacts by pro-apoptotic
function by secretion of inhibitory cytokines, such as TGF-β, proteins such as Bad-Box and opens the mitochondrial transi-
IL-10 [187], and IL-35 that is a novel anti-inflammatory cy- tion pores (MTP). MTP opening promotes release of mito-
tokine [236, 237]. Several studies show that T helper cell chondrial cytochrome c (Cytc) or apoptosis-inducing factor
subpopulations such as pro-inflammatory Th1, Th17, and γδ (AIF) [253]. Cytc in the presence of ATP/dATP binds to the
Neurol Sci (2017) 38:1167–1186 1175

Apaf-1 (apoptotic protease activating factor 1) and inactivating anti-apoptotic protein BCL2 and activating apo-
procaspase-9 to form an apoptosome. Cytc release activates ptotic protein BAX-BAK1 [271, 272]. Following ischemia,
downstream caspases of the intrinsic pathway through forma- reducing Bcl-2 and Bcl-w increased ischemic neuronal cell
tion of the apoptosome that activates caspase-9 and conse- death [273]. Decreasing of the Bcl-2 proteins also resulted in
quently caspase-3. Activated caspase-3 cleaves nuclear DNA astrocyte dysfunction [274]. Moreover, following oxygen and
(nDNA) repair enzymes, causing nDNA damage and apopto- glucose deprivation, ischemic injuries were alleviated by en-
tic cell death. Moreover, AIF is translocated to the nucleus and hancement of Bcl-2 expression in the neurons [275]. Several
causes DNA fragmentation and cell death in a caspase- studies have shown that activation of transcription factors
independent pathway [254]. It has been reported that caspase such as CREB and NFkB are related to cell survival and phos-
proteins are activated in the penumbra zone, and their inhibi- phorylate Bax and Bad thus inhibiting the release of Cytc. In
tion could protect against focal ischemia injuries [255]. addition, inhibition of caspase 3, gene deletions of Bid, and
In response to ischemia, release of the Cytc from outer the use of viral vector-mediated gene transfer of Bcl-2 and
mitochondrial membrane plays a pivotal role in apoptosis ini- Bcl-XL are approaches that are identified to be neuroprotec-
tiation. As mentioned, release of Cytc caused MTP formation, tive [276–280].
in which proteins of the Bcl-2 family could either promote A summary of the most important genes involved in neu-
(Bax, Bak, Bad, Bim, Bid) or prevent (Bcl-2, Bcl-XL, Bcl- ronal degradation and neuronal protection following ischemic
w) MTP formation. In the outer membrane of the mitochon- stroke is provided in Tables 1 and 2.
dria, oligomerization of Bax and/or Bak causes MTP forma-
tion, while anti-apoptotic proteins such as Bcl-2 and Bcl-XL
prevent pore formation via heterodimers forming the Bax pro- Intracranial atherosclerosis and ischemic stroke
teins [256–259]. Bax and Bak directly mediate release of the
Cytc by forming a pore in the mitochondrial membrane. Several lines of evidence show that intracranial atherosclerosis
Meanwhile, Bid, Bad, and Bim appear to function as sensors is a common cause of ischemic stroke [281–283]. Progression
of cell stress and promote apoptosis by antagonizing the func- of intracranial atherosclerosis is accelerated in association
tion of Bcl-2/Bcl-XL, as anti-apoptotic proteins, and/or acti- with several risk factors such as hypertension and diabetes
vating the function of apoptotic function of the Bax and Bak mellitus [282, 283]. Advanced atherosclerosis can affect
[257, 258, 260]. downstream or collateral perfusion because of stenosis of
The extrinsic pathway is initiated via ligand-receptor inter- blood vessels [284, 285]. Advanced atherosclerosis leads to
actions, and it can induce caspase activation independent of vasoconstriction, platelet activation, and thrombosis and intra-
the release of Cytc. The TNF family of ligands such as FASL, cranial atherosclerosis, which potentially alters hemodynam-
TNF, LT-alpha, LT-beta, CD40L, LIGHT, RANKL, and ics and may cause thrombosis that has been linked to
TRAIL bind to the TNF-receptor, Fas receptor (FasR), and Alzheimer’s disease (AD) [286, 287]. Atherosclerosis of ce-
TRAIL receptor that leads to the activation of death receptors rebral vessels may enhance the production of amyloid-β pep-
[261, 262]. These bindings result in activation of caspase-8 tide (Aβ), a peptide central to AD pathology, through induc-
and caspase-10, which in turn can activate effector caspase 3 tion of hypoperfusion and hypoxia. In turn, Aβ may promote
[263, 264]. Activation of caspase-3 results in the mitochon- formation of atherosclerotic lesions through vascular oxida-
drial membrane permeabilization, chromatin condensation, tive stress and endothelial dysfunction. Furthermore, athero-
DNA fragmentation, and eventually cell death [265]. sclerosis could contribute to AD pathophysiology by
Increasing the Fas expression by the extrinsic caspase- impairing Aβ clearance and elevating brain levels of Aβ
dependent pathway has been involved in the ischemic stroke. [288]. Antioxidants are known to affect the progression of
Further studies have revealed that expression of caspase-1, experimental atherosclerosis [289], which may be why of in-
caspase-3, caspase-8, and caspase-9 and cleaved caspase-8 tracranial arteries are less susceptible to hypercholesterolemia
are increased in the ischemic penumbra zone [266]. Fas/ and ox-LDL [290, 291]. Furthermore, the rate of progression
FasL belong to the tumor necrosis factor receptor/ligand su- of atherosclerosis can be affected by the gut microbiota [292,
perfamily of costimulatory molecules and play an important 293] and gut microbes can modify important factors in hemo-
role in induction of apoptosis [267]. Caspase-1 and caspase-3 stasis and thrombosis, including platelet responsiveness and
activation occurred only 2 h after ischemia and reperfusion clot formation [294].
[268].
Several studies have revealed that inhibition of apoptosis
decreases ischemic injury [269]. For example, inhibition of Future therapies
BCL2L11 led to reduction of apoptosis, restoration of neuro-
logical scores, and reduction of behavioral abnormalities fol- Despite progress in understanding the pathophysiologic
lowing stroke [270]. BCL2L11 induce apoptosis by mechanisms of ischemic stroke over the last few decades,
1176 Neurol Sci (2017) 38:1167–1186

Table 1 Specific genes involved in neurodegeneration following ischemic stroke

Pathogenic category Genes Expression Function Ref.

Excitotoxicity NR2B Increased Calcium influx, stimulates the signaling components of NDC [37, 67]
AMPAR Increased Calcium influx [37]
NCX Decreased Dis-regulation of intracellular calcium and sodium homeostasis [38, 40]
mNCX Increased Cytoplasmic calcium loading [45, 46]
Calpains Increased Turns the calcium influx via the NMDAR into neuronal damage [62, 63, 76, 77]
Oxidative and NADPH oxidase Increased Produces the majority of superoxide anions [94, 95]
nitrative stress xanthine oxidase Increased ROS formation [97]
NOS Increased RNS formation [23]
Inflammation IL-1 Increased Increases leukocyte infiltration, breakdown of BBB [161, 168–170]
IL-6 Increased Elevates body temperature [180]
TNF-α Increased Neutrophil infiltration, expression of adhesion molecules, BBB [184, 185]
disruption
CAM Increased Leukocyte transmigration [143]
NF-kB Increased Activation of pro-inflammatory genes and adhesion molecules [194, 195]
MMP Increased Disruption of BBB and hemorrhagic transformation [200–202]
TLR Increased Activation of NF-κB, Cytokine and chemokine production [204–207, 215]
Apoptosis Bax, Bak Increased Promotes MTP formation, [257, 258, 260].
Bad, Bim, Bid Increased Sensors of cell stress [257, 258, 260].
Caspase-1, caspase-3, Increased DNA fragmentation and cell death [265, 266]
caspase-8, caspase-9
Fas Increased Induces caspase activation [266]
BCL2L11 Increased Inactivates BCL2 and activating BAX-BAK1 [271, 272]

NR2B N-methyl-D-aspartate receptor (NMDAR) subunit, NDC neuronal death-signaling complex, AMPAR a-amino-3-hydroxy-5-methyl-4-
isoxazolepropionic acid receptor, NCX sodium–calcium exchanger, mNCX mitochondrial NCX, calpains a family of calcium-activated cysteine prote-
ases, ROS reactive oxygen species, NOS nitric oxide synthase, RNS reactive nitrative species, TNF-α tumor necrosis factor α, IL-1β interleukin-1β, IL-6
interleukin-6, BBB blood brain barrier, CAM cell adhesion molecule, NF-kB nuclear factor kappa B, IRF 1 interferon regulator factor 1, HIF 1 hypoxia
inducible factor 1, MMP matrix metalloproteinases, TLR toll-like receptor, MTP mitochondrial transition pores, Fas tumor necrosis factor receptor, Bcl-2
B cell lymphoma 2

thrombolytic therapy with tPA and endovascular stroke. Due to a narrow time window and a hemorrhagic risk,
recannulation are only FDA-approved treatments for acute this treatment is only suitable for a minority (4%) of stroke

Table 2 Specific genes involved in neuroprotection following ischemic stroke

Pathogenic category Genes Expression Function Ref.

Excitotoxicity NR2A Decreased Stimulates the signaling components of NSC [59, 67]
mGluR (group I) Decreased Activation of the PI3K-Akt signaling pathway [49–51]
mGluR (group II and Decreased Inhibition of presynaptic glutamate release [52]
group III)
GLT-1 Decreased Inhibits glutamate accumulation [54]
Oxidative and nitrative SOD enzymes Increased Maintains redox homeostasis [109, 110]
stress Nrf2 Increased Antioxidant responses [112, 113]
Inflammation IGF-I Increased Neurogenesis [135–137]
BDNF Increased Neurogenesis [135–137]
IL-10 Increased Inhibition of pro-inflammatory cytokine [186]
production and chemokine secretion
TGF-β Increased Reduces the number of circulating neutrophils [190]
Apoptosis Bcl-2, Bcl-w Decreased Prevents MTP formation [273]

NR2A NMDAR subunit, mGluR metabotropic glutamate receptors, GLT-1 glutamate transporter-1, NSC neuronal survival signaling complex, SOD
superoxide dismutase, Nrf2 nuclear factor erythroid-2 related factor 2, IGF-I insulin-like growth factor I, BDNF brain-derived neurotrophic factor, TGF-
β transforming growth factor beta, MTP mitochondrial transition pores, Bcl-2 B cell lymphoma 2
Neurol Sci (2017) 38:1167–1186 1177

patients [295]. Therefore, therapies that can benefit more Combination therapy
stroke patients are urgently needed.
MK-801, an NMDA receptor antagonist, confers neuroprotec-
tion by attenuating the NMDARs and excitotoxicity [310],
Normobaric oxygenation therapy failed in clinical use due to the short therapeutic window.
Moreover, maslinic acid treatment promoted the expression
Currently, stroke researchers are competing to improve neu- of the astrocytic glutamate transporter GLT-1 post-ischemia.
roprotective strategies that guard the brain from ischemia- In ischemic rats, the combination of maslinic acid and MK-
induced injury. As yet, neuroprotective agents that have 801 clearly induced neuroprotection. Furthermore, the syner-
shown protective potential in animal models have failed in gistic effect provided by maslinic acid on the therapeutic time
clinical trials. Since the underlying pathophysiology of stroke window and dosage range for MK-801 is possibly attributed
is highly complicated and exploits numerous deleterious path- to the modulation of glutamate excitotoxicity by increasing
ways, a combination of neuroprotective agents may offer sub- astrocytic glutamate transporter GLT-1 levels and stimulating
stantially better results than a single agent alone, by interven- astrocyte activation. Therefore, a combination of drugs
ing in multiple mechanisms. After stroke onset, normobaric targeting astrocytic glutamate transporters, and also NMDA
oxygenation (NBO) therapy increases tissue oxygenation, receptor antagonists may be a potential treatment for ischemic
thereby decreasing the extent of hypoxic injury. Studies have stroke [311].
also shown that low to moderate levels of ethanol not only
reduce the risk of stroke but also decrease its postischemic Cerebral collateral flow
consequences [296]. In rats following ischemic stroke, NBO
administration salvages ischemic brain tissue by diminishing Cerebral collateral flow is a powerful determinant of ischemic
apoptotic cell death. Further, combination therapy with NBO stroke outcome, which is recruited after arterial occlusion.
and ethanol greatly improves both the degree of neuroprotec- Approaches to support collateral flow in stroke models may
tion and associated apoptosis [297, 298]. Other studies dem- promote the translational development of therapeutic strate-
onstrate that acute ethanol administration reduces lactic acido- gies for enhancing collateral flow, and also may have a role
sis after ischemic stroke. This beneficial effect of ethanol was in the hyper-acute phase of ischemic stroke, prior to recanali-
shown to be potentiated by NBO therapy in permanent ische- zation therapies [312]. Exploring the modulatory mechanisms
mia. Because both NBO and ethanol are available, inexpen- of cerebral collateral flow may improve the outcome of ische-
sive, and easy to administer, their combination could be ap- mic stroke by rescuing cerebral perfusion in the penumbra
plied in clinics shortly after ischemic stroke [299]. region, a potentially viable region of the ischemic area.
These protective mechanisms may have been impaired by
vascular risk factors such as hypertension, diabetes, and acute
Hypothermia ischemia. These risk factors result in insufficient collateral
flow arising from adjacent normally perfused territories, and
Studies have shown that therapeutic hypothermia (TH) is a increase susceptibility to focal ischemic injury. The fate of the
promising treatment for stroke. TH therapy protects brain tis- ischemic penumbra is mainly dependent on the efficiency of
sue via inhibition of several pathways including oxidative collateral flow following vascular occlusion [313]. In stroke
stress, inflammatory responses, metabolic disruption, and cell patients, the degree of collateralization has been revealed to be
death signals [300–302]. In animal models and human pa- a predictor of damage [314] and functional outcome [315].
tients of stroke, TH therapy improves functional and behav- Research has shown that intravenous and intra-arterial reper-
ioral outcomes [300, 303, 304]. TH therapies also affect in- fusion therapy with the presence of good collaterals leads to
flammatory responses after a hypoxic/ischemic insult in cul- better clinical outcomes. Important advances in the imaging of
tured cells as well as in a stroke model of adult mice and can collateral blood vessels and collateral blood flow by multi-
inhibit the expression of inflammatory factors, reduce both modal CT and MRI techniques seem to be the most hopeful
microglial activation and migration as well as promoting func- methods for the routine assessment and quantification of col-
tional recovery following stroke [305]. lateral blood flow in patients with acute ischemic stroke [316].
Additional studies demonstrate that the effect of a single
drug can be improved or the therapeutic time window avail- Pre- and post-conditioning
able for administration extended if applied in combination
with hypothermia. For example, the synergistic effects of S- Conditioning consists of premeditated application of a non-
emopamil, which reduces brain edema, and nimopidine with detrimental physiologic or pharmacologic stimulus or stimu-
hypothermia. The benefit of hypothermia is to extend the time lus train pre- and post-stroke (pre- and post-conditioning, re-
window available for Bcl-2 gene therapy [306–309]. spectively) with the intention that the brain becomes
1178 Neurol Sci (2017) 38:1167–1186

transiently more resistant to ischemia [317–319]. It has been monocyte activation were lessened in postconditioned ani-
shown that pre- and post-conditioning with an ischemic stim- mals [328]. Furthermore, angiogenic responses were in-
ulus are both able to produce neuroprotection in stroke models creased in the conditioned brain following stroke. Indeed, in
[320]. Ischemic pre- and post-conditioning, as promising en- the neonatal rat brain, angiogenesis-related genes were upreg-
dogenous mechanisms of neuroprotection [321], have the po- ulated in response to preconditioning, so postischemic lessen-
tential to affect several protective pathways simultaneously ing in capillary density was ameliorated in these animals
[320]. Several studies showed that patients with a history of [329]. Post-conditioning could thus be seen as a strategy for
transient ischemic attack and who have undergone precondi- neuroprotection in the reperfused brain following stroke
tioning may have reduced morbidity following stroke [322]. [330].
Studies demonstrate that postischemic inflammatory ele-
ments are reduced or abolished in the stroke-tolerant brain.
In particular, preconditioning attenuates the numbers of acti- Conclusion
vated neutrophils that are induced by ischemia, and also de-
creases plasma levels of monocytes and T lymphocytes [323]. Ischemic stroke-induced brain injury results from the interac-
In addition, expression of ICAM-1 and VCAM-1 in cerebral tion of complex pathophysiological processes such as
endothelial cells are reduced by conditioning, [324, 325]. excitotoxicity, oxidative stress, inflammation, and apoptosis
Hence, leukocyte movements along inflamed endothelia and (Fig. 2). Current treatments for ischemic stroke injuries have
leukocyte diapedesis into the brain parenchyma were quanti- proved inadequate, owing to the complexity of events in ce-
tatively reduced in preconditioned animals [326, 327]. rebral ischemia and the disappointing results from single agent
Following stroke, the extent of peripheral lymphopenia and trials. Therefore, it may be unrealistic to hope that a single

Fig. 2 Important pathways in ischemic stroke. Following ischemia, the deprivation of oxygen and glucose to the brain can trigger a cascade of events
that leads to neuronal cell death
Neurol Sci (2017) 38:1167–1186 1179

neuroprotective drug will be beneficial in treatment of stroke. third international stroke trial [IST-3]): a randomised controlled
trial. Lancet (London, England) 379(9834):2352–2363
Despite several agents that can prevent the cascade of events
8. Amarenco P, Bogousslavsky J, Caplan L et al (2009)
leading to ischemic injuries, there is no obvious neuroprotec- Classification of stroke subtypes. Cerebrovasc Dis 27(5):493–501
tive agent yet available to improve stroke outcome in humans. 9. Beal CC (2010) Gender and stroke symptoms: a review of the
Given the complexity of ischemic stroke injuries, an effec- current literature. J Neurosci Nurs 42(2):80–87
tive stroke treatment will probably need a combined approach. 10. Hatano S (1976) Experience from a multicentre stroke register: a
preliminary report. Bull World Health Organ 54(5):541
Fortunately, several experimental discoveries of combination 11. Murphy TH, Li P, Betts K et al (2008) Two-photon imaging of
drug therapy for stroke have revealed drugs that target cellular stroke onset in vivo reveals that NMDA-receptor independent is-
and molecular mechanisms involved in ischemic stroke injury. chemic depolarization is the major cause of rapid reversible dam-
These drugs can act together either synergistically or additive- age to dendrites and spines. J Neurosci 28(7):1756–1772
12. Besancon E, Guo S, Lok J et al (2008) Beyond NMDA and
ly. Combination therapy for cerebral ischemia can be more
AMPA glutamate receptors: emerging mechanisms for ionic im-
effective and has fewer adverse effects than single therapies. balance and cell death in stroke. Trends Pharmacol Sci 29(5):268–
Further studies have shown that single-drug effects can be 275
improved or their time window for administration prolonged 13. Bretón RR, Rodríguez JCG (2012) Excitotoxicity and oxidative
stress in acute ischemic stroke. Stroke 8:9
in combination with hypothermia, a potent therapy that targets
14. Ouyang Y-B, Voloboueva LA, Xu L-J et al (2007) Selective dys-
multiple mechanisms of injury. Furthermore, cerebral collat- function of hippocampal CA1 astrocytes contributes to delayed
eral flow and pre- and post-conditioning are a valuable new neuronal damage after transient forebrain ischemia. J Neurosci
strategies for neuroprotection following ischemic stroke. 27(16):4253–4260
Therefore, due to the extensive overlap of the detrimental 15. Xu L, Emery JF, Ouyang YB et al (2010) Astrocyte targeted over-
expression of Hsp72 or SOD2 reduces neuronal vulnerability to
mechanisms triggered by brain ischemia, combination thera- forebrain ischemia. Glia 58(9):1042–1049
py, cerebral collateral flow, and pre- and post-conditioning 16. Siesjö B (1992) Pathophysiology and treatment of focal cerebral
may represent a promising therapeutic approaches for the ischemia. II: Mechanisms of damage and treatment. J Neurosurg
treatment of stroke. 77(3):337–354
17. Bandera E, Botteri M, Minelli C et al (2006) Cerebral blood flow
threshold of ischemic penumbra and infarct core in acute ischemic
Acknowledgements This work was supported by Ahvaz Jundishapur stroke a systematic review. Stroke 37(5):1334–1339
University Grant. 18. Baron J-C (1999) Mapping the ischaemic penumbra with PET:
implications for acute stroke treatment. Cerebrovasc Dis 9(4):
Compliance with ethical standards 193–201
19. Jung S, Gilgen M, Slotboom J et al (2013) Factors that determine
penumbral tissue loss in acute ischaemic stroke. Brain 136(Pt 12):
Conflict of interest The authors declare that they have no conflict of 3554–3560 awt246
interest.
20. Moskowitz MA, Lo EH, Iadecola C (2010) The science of stroke:
mechanisms in search of treatments. Neuron 67(2):181–198
21. Edvinsson L, Krause DN (2002) Cerebral blood flow and metab-
References olism. Eur J Neurol 9(5):550–550
22. Caplan L (2000) Caplan’s stroke: a clinical approach, 3rd edn.
Butterworth Heinemann, Boston
1. Hossmann K-A (2006) Pathophysiology and therapy of experi- 23. Doyle KP, Simon RP, Stenzel-Poore MP (2008) Mechanisms of
mental stroke. Cell Mol Neurobiol 26(7–8):1055–1081 ischemic brain damage. Neuropharmacology 55(3):310–318
2. Heron M (2007) Deaths: leading causes for 2004. Natl Vital Stat 24. Lai TW, Zhang S, Wang YT (2014) Excitotoxicity and stroke:
Rep 56(5):1–96 identifying novel targets for neuroprotection. Prog Neurobiol
3. Tsuchiya M, Sako K, Yura S et al (1992) Cerebral blood flow and 115:157–188
histopathological changes following permanent bilateral carotid 25. Krnjević K (2008) Electrophysiology of cerebral ischemia.
artery ligation in Wistar rats. Exp Brain Res 89(1):87–92 Neuropharmacology 55(3):319–333
4. Seto S-W, Chang D, Jenkins A et al (2016) Angiogenesis in is- 26. Mehta SL, Manhas N, Raghubir R (2007) Molecular targets in
chemic stroke and angiogenic effects of Chinese herbal medicine. cerebral ischemia for developing novel therapeutics. Brain Res
Journal of clinical medicine 5(6):56 Rev 54(1):34–66
5. Fonarow GC, Zhao X, Smith EE et al (2014) Door-to-needle times 27. Olney JW, Price MT, Samson L et al (1986) The role of specific
for tissue plasminogen activator administration and clinical out- ions in glutamate neurotoxicity. Neurosci Lett 65(1):65–71
comes in acute ischemic stroke before and after a quality improve- 28. Rothman SM (1985) The neurotoxicity of excitatory amino acids
ment initiative. JAMA 311(16):1632–1640 is produced by passive chloride influx. J Neurosci 5(6):1483–
6. Del Zoppo GJ, Saver JL, Jauch EC et al (2009) Expansion of the 1489
time window for treatment of acute ischemic stroke with intrave- 29. Choi DW (1985) Glutamate neurotoxicity in cortical cell culture is
nous tissue plasminogen activator. A science advisory from the calcium dependent. Neurosci Lett 58(3):293–297
American Heart Association/American Stroke Association. Stroke 30. Tymianski M, Charlton MP, Carlen PL et al (1993) Secondary Ca
40(8):2945–2948 2+ overload indicates early neuronal injury which precedes stain-
7. Sandercock P, Wardlaw JM, Lindley RI et al (2012) The benefits ing with viability indicators. Brain Res 607(1):319–323
and harms of intravenous thrombolysis with recombinant tissue 31. Pizzi M, Fallacara C, Arrighi V et al (1993) Attenuation of excit-
plasminogen activator within 6 h of acute ischaemic stroke (the atory amino acid toxicity by metabotropic glutamate receptor
1180 Neurol Sci (2017) 38:1167–1186

agonists and aniracetam in primary cultures of cerebellar granule 51. Hou L, Klann E (2004) Activation of the phosphoinositide 3-
cells. J Neurochem 61(2):683–689 kinase-Akt-mammalian target of rapamycin signaling pathway is
32. Mosbacher J, Schöpfer R, Monyer H et al (1994) A molecular required for metabotropic glutamate receptor-dependent long-term
determinant for submillisecond desensitization in glutamate recep- depression. J Neurosci 24(28):6352–6361
tors. Science 266(5187):1059–1062 52. Bruno V, Battaglia G, Copani A et al (2001) Metabotropic gluta-
33. Moriyoshi K, Masu M, Ishii T et al (1991) Molecular cloning and mate receptor subtypes as targets for neuroprotective drugs. J
characterization of the rat NMD receptor. Nature 354:31–37 Cereb Blood Flow Metab 21(9):1013–1033
34. Berdichevsky E, Riveros N, Sánchez-Armáss S et al (1983) 53. Yang Z-B, Zhang Z, Li T-B et al (2014) Up-regulation of brain-
Kainate, N-methylaspartate and other excitatory amino acids in- enriched miR-107 promotes excitatory neurotoxicity through
crease calcium influx into rat brain cortex cells in vitro. Neurosci down-regulation of glutamate transporter-1 expression following
Lett 36(1):75–80 ischaemic stroke. Clin Sci 127(12):679–689
35. Liu B, Liao M, Mielke JG et al (2006) Ischemic insults direct 54. Fang Q, Hu W-W, Wang X-F et al (2014) Histamine up-regulates
glutamate receptor subunit 2-lacking AMPA receptors to synaptic astrocytic glutamate transporter 1 and protects neurons against
sites. J Neurosci 26(20):5309–5319 ischemic injury. Neuropharmacology 77:156–166
36. Peng PL, Zhong X, Tu W et al (2006) ADAR2-dependent RNA 55. Lee J-M, Zipfel GJ, Choi DW (1999) The changing landscape of
editing of AMPA receptor subunit GluR2 determines vulnerability ischaemic brain injury mechanisms. Nature 399:A7–A14
of neurons in forebrain ischemia. Neuron 49(5):719–733 56. Rothman SM, Olney JW (1995) Excitotoxicity and the NMDA
37. Hsu CY (1998) Ischemic stroke: from basic mechanisms to new receptor-still lethal after eight years. Trends Neurosci 18(2):57–58
drug development, vol 16. Karger Medical and Scientific 57. Traynelis SF, Wollmuth LP, McBain CJ et al (2010) Glutamate
Publishers, Basel receptor ion channels: structure, regulation, and function.
38. Boscia F, Gala R, Pignataro G et al (2006) Permanent focal brain Pharmacol Rev 62(3):405–496
ischemia induces isoform-dependent changes in the pattern of 58. Collingridge GL, Peineau S, Howland JG et al (2010) Long-term
Na&plus;/Ca2&plus; exchanger gene expression in the ischemic depression in the CNS. Nat Rev Neurosci 11(7):459–473
core, periinfarct area, and intact brain regions. J Cereb Blood Flow 59. Liu Y, Wong TP, Aarts M et al (2007) NMDA receptor subunits
Metab 26(4):502–517 have differential roles in mediating excitotoxic neuronal death
39. Molinaro P, Cantile M, Cuomo O et al (2013) Neurounina-1, a both in vitro and in vivo. J Neurosci 27(11):2846–2857
novel compound that increases Na+/Ca2+ exchanger activity, ef- 60. Chen M, Lu T-J, Chen X-J et al (2008) Differential roles of
fectively protects against stroke damage. Mol Pharmacol 83(1): NMDA receptor subtypes in ischemic neuronal cell death and
142–156 ischemic tolerance. Stroke 39(11):3042–3048
61. Ryan TJ, Emes RD, Grant SG et al (2008) Evolution of NMDA
40. Bano D, Young KW, Guerin CJ et al (2005) Cleavage of the
receptor cytoplasmic interaction domains: implications for organi-
plasma membrane Na+/Ca 2+ exchanger in excitotoxicity. Cell
sation of synaptic signalling complexes. BMC Neurosci 9(1):6
120(2):275–285
62. Zhou M, Baudry M (2006) Developmental changes in NMDA
41. Castilho RF, Hansson O, Ward MW et al (1998) Mitochondrial
neurotoxicity reflect developmental changes in subunit composi-
control of acute glutamate excitotoxicity in cultured cerebellar
tion of NMDA receptors. J Neurosci 26(11):2956–2963
granule cells. J Neurosci 18(24):10277–10286
63. DeRidder MN, Simon MJ, Siman R et al (2006) Traumatic me-
42. Abramov AY, Duchen MR (2008) Mechanisms underlying the chanical injury to the hippocampus in vitro causes regional
loss of mitochondrial membrane potential in glutamate caspase-3 and calpain activation that is influenced by NMDA
excitotoxicity. Biochimica et Biophysica Acta (BBA)- receptor subunit composition. Neurobiol Dis 22(1):165–176
Bioenergetics 1777(7):953–964
64. Terasaki Y, Sasaki T, Yagita Y et al (2010) Activation of NR2A
43. Ward MW, Rego AC, Frenguelli BG et al (2000) Mitochondrial receptors induces ischemic tolerance through CREB signaling. J
membrane potential and glutamate excitotoxicity in cultured cer- Cereb Blood Flow Metab 30(8):1441–1449
ebellar granule cells. J Neurosci 20(19):7208–7219 65. Harraz MM, Eacker SM, Wang X et al (2012) MicroRNA-223 is
44. Stout AK, Raphael HM, Kanterewicz BI et al (1998) Glutamate- neuroprotective by targeting glutamate receptors. Proc Natl Acad
induced neuron death requires mitochondrial calcium uptake. Nat Sci 109(46):18962–18967
Neurosci 1(5):366–373 66. Martin HG, Wang YT (2010) Blocking the deadly effects of the
45. White RJ, Reynolds IJ (1996) Mitochondrial depolarization in NMDA receptor in stroke. Cell 140(2):174–176
glutamate-stimulated neurons: an early signal specific to 67. Hardingham GE, Fukunaga Y, Bading H (2002) Extrasynaptic
excitotoxin exposure. J Neurosci 16(18):5688–5697 NMDARs oppose synaptic NMDARs by triggering CREB shut-
46. White RJ, Reynolds IJ (1997) Mitochondria accumulate Ca2+ off and cell death pathways. Nat Neurosci 5(5):405–414
following intense glutamate stimulation of cultured rat forebrain 68. Tu W, Xu X, Peng L et al (2010) DAPK1 interaction with NMDA
neurones. J Physiol 498(Pt 1):31 receptor NR2B subunits mediates brain damage in stroke. Cell
47. Tymianski M, Charlton MP, Carlen PL et al (1993) Source spec- 140(2):222–234
ificity of early calcium neurotoxicity in cultured embryonic spinal 69. Zhou L, Li F, Xu H-B et al (2010) Treatment of cerebral ischemia
neurons. J Neurosci 13(5):2085–2104 by disrupting ischemia-induced interaction of nNOS with PSD-
48. Baudry M, Greget R, Pernot F et al (2012) Roles of group I 95. Nat Med 16(12):1439–1443
metabotropic glutamate receptors under physiological conditions 70. Lai TW, Shyu W-C, Wang YT (2011) Stroke intervention path-
and in neurodegeneration. Wiley Interdisciplinary Reviews: ways: NMDA receptors and beyond. Trends Mol Med 17(5):266–
Membrane Transport and Signaling 1(4):523–532 275
49. Rong R, Ahn J-Y, Huang H et al (2003) PI3 kinase enhancer– 71. Aluclu MU, Arslan S, Acar A et al (2008) Evaluation of effects of
Homer complex couples mGluRI to PI3 kinase, preventing neu- memantine on cerebral ischemia in rats. Neurosciences (Riyadh)
ronal apoptosis. Nat Neurosci 6(11):1153–1161 13(2):113–116
50. Chong ZZ, Li F, Maiese K (2006) Group I metabotropic receptor 72. Okamoto S-I, Pouladi MA, Talantova M et al (2009) Balance
neuroprotection requires Akt and its substrates that govern between synaptic versus extrasynaptic NMDA receptor activity
FOXO3a, Bim, and β-catenin during oxidative stress. Curr influences inclusions and neurotoxicity of mutant huntingtin.
Neurovasc Res 3(2):107–117 Nat Med 15(12):1407–1413
Neurol Sci (2017) 38:1167–1186 1181

73. Aarts M, Liu Y, Liu L et al (2002) Treatment of ischemic brain 96. Nicholls DG (2008) Oxidative stress and energy crises in neuronal
damage by perturbing NMDA receptor-PSD-95 protein interac- dysfunction. Ann N Y Acad Sci 1147(1):53–60
tions. Science 298(5594):846–850 97. Abramov AY, Scorziello A, Duchen MR (2007) Three distinct
74. Lai TW, Wang YT (2010) Fashioning drugs for stroke. Nat Med mechanisms generate oxygen free radicals in neurons and contrib-
16(12):1376–1378 ute to cell death during anoxia and reoxygenation. J Neurosci
75. Petralia RS, Wang Y-X, Hua F et al (2010) Organization of 27(5):1129–1138
NMDA receptors at extrasynaptic locations. Neuroscience 98. Förstermann U (2010) Nitric oxide and oxidative stress in vascular
167(1):68–87 disease. Pflügers Archiv-European Journal of Physiology 459(6):
76. Koumura A, Nonaka Y, Hyakkoku K et al (2008) A novel calpain 923–939
inhibitor,((1S)-1 ((((1S)-1-benzyl-3-cyclopropylamino-2, 3-di- 99. Wei EP, Kontos HA, Beckman JS (1996) Mechanisms of cerebral
oxopropyl) amino) carbonyl)-3-methylbutyl) carbamic acid 5- vasodilation by superoxide, hydrogen peroxide, and peroxynitrite.
methoxy-3-oxapentyl ester, protects neuronal cells from cerebral Am J Phys Heart Circ Phys 271(3):H1262–H1266
ischemia-induced damage in mice. Neuroscience 157(2):309–318 100. Aarts MM, Tymianski M (2005) TRPMs and neuronal cell death.
77. López-Menéndez C, Gascón S, Sobrado M et al (2009) Pflugers Arch 451(1):243–249
Kidins220/ARMS downregulation by excitotoxic activation of 101. Pacher P, Beckman JS, Liaudet L (2007) Nitric oxide and
NMDARs reveals its involvement in neuronal survival and death peroxynitrite in health and disease. Physiol Rev 87(1):315–424
pathways. J Cell Sci 122(19):3554–3565 102. Nakamura T, Lipton SA (2009) According to GOSPEL: filling in
78. Xu J, Kurup P, Zhang Y et al (2009) Extrasynaptic NMDA recep- the GAP (DH) of NO-mediated neurotoxicity. Neuron 63(1):3–6
tors couple preferentially to excitotoxicity via calpain-mediated 103. Gu Z, Kaul M, Yan B et al (2002) S-nitrosylation of matrix me-
cleavage of STEP. J Neurosci 29(29):9330–9343 talloproteinases: signaling pathway to neuronal cell death. Science
79. Taghibiglou C, Martin HG, Lai TW et al (2009) Role of NMDA 297(5584):1186–1190
receptor—dependent activation of SREBP1 in excitotoxic and is-
104. Faraci FM (2006) Reactive oxygen species: influence on cerebral
chemic neuronal injuries. Nat Med 15(12):1399–1406
vascular tone. J Appl Physiol 100(2):739–743
80. Beckman KB, Ames BN (1998) Mitochondrial aging: open ques-
105. Lipton SA (2007) Pathologically activated therapeutics for neuro-
tions. Ann N Y Acad Sci 854(1):118–127
protection. Nat Rev Neurosci 8(10):803–808
81. Suh SW, Shin BS, Ma H et al (2008) Glucose and NADPH oxi-
106. Neumar RW (2000) Molecular mechanisms of ischemic neuronal
dase drive neuronal superoxide formation in stroke. Ann Neurol
injury. Ann Emerg Med 36(5):483–506
64(6):654–663
107. Gariballa S, Hutchin T, Sinclair A (2002) Antioxidant capacity
82. Allen C, Bayraktutan U (2009) Oxidative stress and its role in the
after acute ischaemic stroke. QJM 95(10):685–690
pathogenesis of ischaemic stroke. Int J Stroke 4(6):461–470
83. Love S (1999) Oxidative stress in brain ischemia. Brain Pathol 108. Spranger M, Krempien S, Schwab S et al (1997) Superoxide dis-
9(1):119–131 mutase activity in serum of patients with acute cerebral ischemic
injury correlation with clinical course and infarct size. Stroke
84. Chan PH (2001) Reactive oxygen radicals in signaling and dam-
28(12):2425–2428
age in the ischemic brain. J Cereb Blood Flow Metab 21(1):2–14
85. Kontos HA (2001) Oxygen radicals in cerebral ischemia the 2001 109. Alfieri A, Srivastava S, Siow R et al (2011) Targeting the Nrf2–
Willis lecture. Stroke 32(11):2712–2716 Keap1 antioxidant defence pathway for neurovascular protection
in stroke. J Physiol 589(17):4125–4136
86. Cherubini A, Ruggiero C, Polidori MC et al (2005) Potential
markers of oxidative stress in stroke. Free Radic Biol Med 110. Margaill I, Plotkine M, Lerouet D (2005) Antioxidant strategies in
39(7):841–852 the treatment of stroke. Free Radic Biol Med 39(4):429–443
87. Coyle JT, Puttfarcken P (1993) Oxidative stress, glutamate, and 111. Zhang C, Shu L, Kong A-N T (2015) MicroRNAs: new players in
neurodegenerative disorders. Science 262(5134):689–695 cancer prevention targeting Nrf2, oxidative stress and inflamma-
88. Cuzzocrea S, Riley DP, Caputi AP et al (2001) Antioxidant ther- tory pathways. Current pharmacology reports 1(1):21–30
apy: a new pharmacological approach in shock, inflammation, and 112. Johnson JA, Johnson DA, Kraft AD et al (2008) The Nrf2–ARE
ischemia/reperfusion injury. Pharmacol Rev 53(1):135–159 pathway. Ann N Y Acad Sci 1147(1):61–69
89. Lafon-Cazal M, Pietri S, Culcasi M et al (1993) NMDA- 113. Hardingham GE, Lipton SA (2011) Regulation of neuronal oxida-
dependent superoxide production and neurotoxicity. Nature tive and nitrosative stress by endogenous protective pathways and
364(6437):535–537 disease processes. Antioxid Redox Signal 14(8):1421–1424
90. Piantadosi CA, Zhang J (1996) Mitochondrial generation of reac- 114. Dang J, Brandenburg L-O, Rosen C et al (2012) Nrf2 expression
tive oxygen species after brain ischemia in the rat. Stroke 27(2): by neurons, astroglia, and microglia in the cerebral cortical pen-
327–332 umbra of ischemic rats. J Mol Neurosci 46(3):578–584
91. Sugawara T, Chan PH (2003) Reactive oxygen radicals and path- 115. Joshi GA, Johnson J (2012) The Nrf2-ARE pathway: a valuable
ogenesis of neuronal death after cerebral ischemia. Antioxid therapeutic target for the treatment of neurodegenerative diseases.
Redox Signal 5(5):597–607 Recent patents on CNS drug discovery 7(3):218–229
92. Saeed SA, Shad KF, Saleem T et al (2007) Some new prospects in 116. Jiang S, Deng C, Lv J et al (2016) Nrf2 weaves an elaborate
the understanding of the molecular basis of the pathogenesis of network of neuroprotection against stroke. Mol Neurobiol 1–16
stroke. Exp Brain Res 182(1):1–10 117. Zhang M, An C, Gao Y et al (2013) Emerging roles of Nrf2 and
93. Adibhatla RM, Hatcher JF (2010) Lipid oxidation and peroxida- phase II antioxidant enzymes in neuroprotection. Prog Neurobiol
tion in CNS health and disease: from molecular mechanisms to 100:30–47
therapeutic opportunities. Antioxid Redox Signal 12(1):125–169 118. Liou AK, Clark RS, Henshall DC et al (2003) To die or not to die
94. Girouard H, Wang G, Gallo EF et al (2009) NMDA receptor for neurons in ischemia, traumatic brain injury and epilepsy: a
activation increases free radical production through nitric oxide review on the stress-activated signaling pathways and apoptotic
and NOX2. J Neurosci 29(8):2545–2552 pathways. Prog Neurobiol 69(2):103–142
95. Brennan AM, Suh SW, Won SJ et al (2009) NADPH oxidase is the 119. Chamorro Á, Hallenbeck J (2006) The harms and benefits of in-
primary source of superoxide induced by NMDA receptor activa- flammatory and immune responses in vascular disease. Stroke
tion. Nat Neurosci 12(7):857–863 37(2):291–293
1182 Neurol Sci (2017) 38:1167–1186

120. McColl B, Allan S, Rothwell N (2009) Systemic infection, inflam- prerequisite for successful immune cell entry to the brain.
mation and acute ischemic stroke. Neuroscience 158(3):1049– Neuropathol Appl Neurobiol 37(1):24–39
1061 144. Yang MS, Min KJ, Joe E (2007) Multiple mechanisms that pre-
121. Pan J, Palmateer J, Schallert T et al (2014) Novel humanized vent excessive brain inflammation. J Neurosci Res 85(11):2298–
recombinant T cell receptor ligands protect the female brain after 2305
experimental stroke. Translational stroke research 5(5):577–585 145. Vincent V, Tilders F, Van Dam AM (1997) Inhibition of
122. Jeong H-K, Ji K, Min K et al (2013) Brain inflammation and endotoxin-induced nitric oxide synthase production in microglial
microglia: facts and misconceptions. Exp Neurobiol 22(2):59–67 cells by the presence of astroglial cells: a role for transforming
123. Amantea D, Nappi G, Bernardi G et al (2009) Post-ischemic brain growth factor β. Glia 19(3):190–198
damage: pathophysiology and role of inflammatory mediators. 146. Pyo H, Yang M-S, Jou I et al (2003) Wortmannin enhances
FEBS J 276(1):13–26 lipopolysaccharide-induced inducible nitric oxide synthase ex-
124. Kriz J (2006) Inflammation in ischemic brain injury: timing is pression in microglia in the presence of astrocytes in rats.
important. Critical reviews™ in Neurobiology 18 (1–2): Neurosci Lett 346(3):141–144
125. Stanimirovic DB, Wong J, Shapiro A et al (1997) Increase in 147. Min K-J, Yang M-S, Kim S-U et al (2006) Astrocytes induce
surface expression of ICAM-1, VCAM-1 and E-selectin in human hemeoxygenase-1 expression in microglia: a feasible mechanism
cerebromicrovascular endothelial cells subjected to ischemia-like for preventing excessive brain inflammation. J Neurosci 26(6):
insults. In: Brain Edema X. Springer, Berlin Heidelberg New 1880–1887
York, p 12–16 148. Kim JH, Min KJ, Seol W et al (2010) Astrocytes in injury states
126. Becker K (1998) Inflammation and acute stroke. Curr Opin Neurol rapidly produce anti-inflammatory factors and attenuate microglial
11(1):45–49 inflammatory responses. J Neurochem 115(5):1161–1171
127. Lakhan SE, Kirchgessner A, Hofer M (2009) Inflammatory mech- 149. Kim B, Jeong H-K, Kim J-H et al (2011) Uridine 5′-diphosphate
anisms in ischemic stroke: therapeutic approaches. J Transl Med induces chemokine expression in microglia and astrocytes through
7(1):1 activation of the P2Y6 receptor. J Immunol 186(6):3701–3709
128. Gan Y, Liu Q, Wu W et al (2014) Ischemic neurons recruit natural 150. Hoek RM, Ruuls SR, Murphy CA et al (2000) Down-regulation of
killer cells that accelerate brain infarction. Proc Natl Acad Sci the macrophage lineage through interaction with OX2 (CD200).
111(7):2704–2709 Science 290(5497):1768–1771
129. Rogove A, Lu W, Tsirka S (2002) Microglial activation and re- 151. Cardona AE, Pioro EP, Sasse ME et al (2006) Control of
cruitment, but not proliferation, suffice to mediate neurodegener- microglial neurotoxicity by the fractalkine receptor. Nat
ation. Cell Death Differ 9(8):801–806 Neurosci 9(7):917–924
130. Graeber MB, Streit WJ (2010) Microglia: biology and pathology. 152. Kim YS, Kim SS, Cho JJ et al (2005) Matrix metalloproteinase-3:
Acta Neuropathol 119(1):89–105 a novel signaling proteinase from apoptotic neuronal cells that
131. McKimmie CS, Roy D, Forster T et al (2006) Innate immune activates microglia. J Neurosci 25(14):3701–3711
response gene expression profiles of N9 microglia are pathogen-
153. Iadecola C, Anrather J (2011) The immunology of stroke: from
type specific. J Neuroimmunol 175(1):128–141
mechanisms to translation. Nat Med 17(7):796–808
132. Hoehn BD, Palmer TD, Steinberg GK (2005) Neurogenesis in rats
154. Swanson RA, Ying W, Kauppinen TM (2004) Astrocyte influ-
after focal cerebral ischemia is enhanced by indomethacin. Stroke
ences on ischemic neuronal death. Curr Mol Med 4(2):193–205
36(12):2718–2724
155. Ferrarese C, Mascarucci P, Zoia C et al (1999) Increased cytokine
133. Pena-Philippides JC, Yang Y, Bragina O et al (2014) Effect of
release from peripheral blood cells after acute stroke. J Cereb
pulsed electromagnetic field (PEMF) on infarct size and inflam-
Blood Flow Metab 19(9):1004–1009
mation after cerebral ischemia in mice. Translational stroke re-
search 5(4):491–500 156. Fu Y, Liu Q, Anrather J et al (2015) Immune interventions in
134. Yilmaz G, Granger DN (2008) Cell adhesion molecules and ische- stroke. Nat Rev Neurol 11(9):524–535
mic stroke. Neurol Res 30(8):783–793 157. Chamorro Á, Meisel A, Planas AM et al (2012) The immunology
135. Mrak RE, Griffin WST (2005) Glia and their cytokines in progres- of acute stroke. Nat Rev Neurol 8(7):401–410
sion of neurodegeneration. Neurobiol Aging 26(3):349–354 158. Gelderblom M, Leypoldt F, Steinbach K et al (2009) Temporal and
136. Eikelenboom P, Rozemuller AJ, Hoozemans JJ et al (2000) spatial dynamics of cerebral immune cell accumulation in stroke.
Neuroinflammation and Alzheimer disease: clinical and therapeu- Stroke 40(5):1849–1857
tic implications. Alzheimer Dis Assoc Disord 14(1):S54–S61 159. Hammond MD, Taylor RA, Mullen MT et al (2014) CCR2+
137. Lucas SM, Rothwell NJ, Gibson RM (2006) The role of inflam- Ly6Chi inflammatory monocyte recruitment exacerbates acute
mation in CNS injury and disease. Br J Pharmacol 147(S1):S232– disability following intracerebral hemorrhage. J Neurosci 34(11):
S240 3901–3909
138. Fernandes A, Miller-Fleming L, Pais TF (2014) Microglia and 160. Zhang R, Chopp M, Zhang Z et al (1998) The expression of P-and
inflammation: conspiracy, controversy or control? Cell Mol Life E-selectins in three models of middle cerebral artery occlusion.
Sci 71(20):3969–3985 Brain Res 785(2):207–214
139. Nathan C, Ding A (2010) Nonresolving inflammation. Cell 161. Huang J, Upadhyay UM, Tamargo RJ (2006) Inflammation in
140(6):871–882 stroke and focal cerebral ischemia. Surg Neurol 66(3):232–245
140. Widgerow AD (2012) Cellular resolution of inflammation— 162. Chou WH, Choi DS, Zhang H et al (2004) Neutrophil protein
catabasis. Wound Repair Regen 20(1):2–7 kinase Cdelta as a mediator of stroke-reperfusion injury. J Clin
141. Fleming JC, Norenberg MD, Ramsay DA et al (2006) The cellular Invest 114(1):49–56
inflammatory response in human spinal cords after injury. Brain 163. Liu T, Clark R, McDonnell P et al (1994) Tumor necrosis factor-
129(12):3249–3269 alpha expression in ischemic neurons. Stroke 25(7):1481–1488
142. Ross AM, Hurn P, Perrin N et al (2007) Evidence of the peripheral 164. Zhu Y, Yang G-Y, Ahlemeyer B et al (2002) Transforming growth
inflammatory response in patients with transient ischemic attack. J factor-β1 increases bad phosphorylation and protects neurons
Stroke Cerebrovasc Dis 16(5):203–207 against damage. J Neurosci 22(10):3898–3909
143. Greenwood J, Heasman S, Alvarez J et al (2011) Review: leuco- 165. Spera PA, Ellison JA, Feuerstein GZ et al (1998) IL-10 reduces rat
cyte–endothelial cell crosstalk at the blood–brain barrier: a brain injury following focal stroke. Neurosci Lett 251(3):189–192
Neurol Sci (2017) 38:1167–1186 1183

166. Vila N, Castillo J, Dávalos A et al (2003) Levels of anti- 185. Nawashiro H, Martin D, Hallenbeck JM (1997) Neuroprotective
inflammatory cytokines and neurological worsening in acute is- effects of TNF binding protein in focal cerebral ischemia. Brain
chemic stroke. Stroke 34(3):671–675 Res 778(2):265–271
167. Viviani B, Bartesaghi S, Gardoni F et al (2003) Interleukin-1β 186. Ooboshi H, Ibayashi S, Shichita T et al (2005) Postischemic gene
enhances NMDA receptor-mediated intracellular calcium increase transfer of interleukin-10 protects against both focal and global
through activation of the Src family of kinases. J Neurosci 23(25): brain ischemia. Circulation 111(7):913–919
8692–8700 187. Liesz A, Suri-Payer E, Veltkamp C et al (2009) Regulatory T cells
168. Bernardes-Silva M, Anthony DC, Issekutz AC et al (2001) are key cerebroprotective immunomodulators in acute experimen-
Recruitment of Neutrophils across the blood–brain barrier: the tal stroke. Nat Med 15(2):192–199
role of E-and P-selectins. J Cereb Blood Flow Metab 21(9): 188. Iadecola C, Anrather J (2011) Stroke research at a crossroad: ask-
1115–1124 ing the brain for directions. Nat Neurosci 14(11):1363–1368
169. Konsman JP, Vigues S, Mackerlova L et al (2004) Rat brain vas- 189. McNeill H, Williams C, Guan J et al (1994) Neuronal rescue with
cular distribution of interleukin-1 type-1 receptor immunoreactiv- transforming growth factor-[beta] 1 after hypoxic-ischaemic brain
ity: relationship to patterns of inducible cyclooxygenase expres- injury. Neuroreport 5(8):901–904
sion by peripheral inflammatory stimuli. J Comp Neurol 472(1):
190. Mori E, Del Zoppo GJ, Chambers JD et al (1992) Inhibition of
113–129
polymorphonuclear leukocyte adherence suppresses no-reflow af-
170. Mazzotta G, Sarchielli P, Caso V et al (2004) Different cytokine
ter focal cerebral ischemia in baboons. Stroke 23(5):712–718
levels in thrombolysis patients as predictors for clinical outcome.
191. Nurmi A, Lindsberg PJ, Koistinaho M et al (2004) Nuclear factor-
Eur J Neurol 11(6):377–381
κB contributes to infarction after permanent focal ischemia. Stroke
171. Bö L, Peterson JW, Mørk S et al (1996) Distribution of immuno-
35(4):987–991
globulin superfamily members ICAM-1,-2,-3, and the β2 integrin
LFA-1 in multiple sclerosis lesions. J Neuropathol Exp Neurol 192. Hayden MS, Ghosh S (2008) Shared principles in NF-κB signal-
55(10):1060–1072 ing. Cell 132(3):344–362
172. Huang FP, Wang ZQ, Wu DC et al (2003) Early NFκB activation 193. Ridder D, Schwaninger M (2009) NF-κB signaling in cerebral
is inhibited during focal cerebral ischemia in interleukin-1β- ischemia. Neuroscience 158(3):995–1006
converting enzyme deficient mice. J Neurosci Res 73(5):698–707 194. Han H, Yenari M (2003) Cellular targets of brain inflammation in
173. Ohtaki H, Takaki A, Yin L et al (2003) Suppression of oxidative stroke. Current opinion in investigational drugs (London,
stress after transient focal ischemia in interleukin-1 knock out England: 2000) 4(5):522–529
mice. In: Brain Edema XII. Springer, Berlin Heidelberg New 195. Baeuerle P, Henkel T (1994) Dunction and activation of NF-B in
York, p 191–194 the immune system. Annu Rev Immunol 12(141):79
174. Wang X, Yue T-L, Young PR et al (1995) Expression of interleu- 196. Ko HM, Koppula S, Kim B-W et al (2010) Inflexin attenuates
kin-6, c-fos, and zif268 mRNAs in rat ischemic cortex. J Cereb proinflammatory responses and nuclear factor-ΚB activation in
Blood Flow Metab 15(1):166–171 LPS-treated microglia. Eur J Pharmacol 633(1):98–106
175. Tarkowski E, Rosengren L, Blomstrand C et al (1995) Early in- 197. Jin H, Zhu ZG, Yu PJ et al (2012) Myrislignan attenuates
trathecal production of interleukin-6 predicts the size of brain le- lipopolysaccharide-induced inflammation reaction in murine
sion in stroke. Stroke 26(8):1393–1398 macrophage cells through inhibition of NF-κB signalling pathway
176. Erta M, Quintana A, Hidalgo J (2012) Interleukin-6, a major cy- activation. Phytother Res 26(9):1320–1326
tokine in the central nervous system. Int J Biol Sci 8(9):1254– 198. Wang X, Hu D, Zhang L et al (2014) Gomisin A inhibits
1266 lipopolysaccharide-induced inflammatory responses in N9 mi-
177. Hakkoum D, Stoppini L, Muller D (2007) Interleukin-6 promotes croglia via blocking the NF-κB/MAPKs pathway. Food Chem
sprouting and functional recovery in lesioned organotypic hippo- Toxicol 63:119–127
campal slice cultures. J Neurochem 100(3):747–757 199. Montaner J, Alvarez-Sabín J, Molina C et al (2001) Matrix metal-
178. Tancredi V, D'Antuono M, Cafè C et al (2000) The inhibitory loproteinase expression after human cardioembolic stroke tempo-
effects of interleukin-6 on synaptic plasticity in the rat hippocam- ral profile and relation to neurological impairment. Stroke 32(8):
pus are associated with an inhibition of mitogen-activated protein 1759–1766
kinase ERK. J Neurochem 75(2):634–643
200. Danton GH, Dietrich WD (2003) Inflammatory mechanisms after
179. Relton J, Martin D, Thompson R et al (1996) Peripheral adminis-
ischemia and stroke. J Neuropathol Exp Neurol 62(2):127–136
tration of interleukin-1 receptor antagonist inhibits brain damage
201. Jin R, Yang G, Li G (2010) Molecular insights and therapeutic
after focal cerebral ischemia in the rat. Exp Neurol 138(2):206–
targets for blood–brain barrier disruption in ischemic stroke: crit-
213
ical role of matrix metalloproteinases and tissue-type plasminogen
180. Azzimondi G, Bassein L, Nonino F et al (1995) Fever in acute
activator. Neurobiol Dis 38(3):376–385
stroke worsens prognosis: a prospective study. Stroke 26(11):
2040–2043 202. Asahi M, Wang X, Mori T et al (2001) Effects of matrix
181. Zaremba J, Skrobanski P, Losy J (2001) Tumour necrosis factor- metalloproteinase-9 gene knock-out on the proteolysis of blood–
alpha is increased in the cerebrospinal fluid and serum of ischae- brain barrier and white matter components after cerebral ischemia.
mic stroke patients and correlates with the volume of evolving J Neurosci 21(19):7724–7732
brain infarct. Biomed Pharmacother 55(5):258–263 203. Zhao B-Q, Wang S, Kim H-Y et al (2006) Role of matrix metal-
182. Dawson DA, Martin D, Hallenbeck JM (1996) Inhibition of tumor loproteinases in delayed cortical responses after stroke. Nat Med
necrosis factor-alpha reduces focal cerebral ischemic injury in the 12(4):441–445
spontaneously hypertensive rat. Neurosci Lett 218(1):41–44 204. Weinstein JR, Koerner IP, Möller T (2010) Microglia in ischemic
183. Lavine SD, Hofman FM, Zlokovic BV (1998) Circulating anti- brain injury. Future Neurol 5(2):227–246
body against tumor necrosis factor-alpha protects rat brain from 205. Kawai T, Akira S (2010) The role of pattern-recognition receptors
reperfusion injury. J Cereb Blood Flow Metab 18(1):52–58 in innate immunity: update on Toll-like receptors. Nat Immunol
184. Nawashiro H, Martin D, Hallenbeck JM (1997) Inhibition of tu- 11(5):373–384
mor necrosis factor and amelioration of brain infarction in mice. J 206. Karikó K, Ni H, Capodici J et al (2004) mRNA is an endogenous
Cereb Blood Flow Metab 17(2):229–232 ligand for Toll-like receptor 3. J Biol Chem 279(13):12542–12550
1184 Neurol Sci (2017) 38:1167–1186

207. Marsh BJ, Stenzel-Poore MP (2008) Toll-like receptors: novel 229. Justicia C, Panés J, Solé S et al (2003) Neutrophil infiltration
pharmacological targets for the treatment of neurological diseases. increases matrix metalloproteinase-9 in the ischemic brain after
Curr Opin Pharmacol 8(1):8–13 occlusion/reperfusion of the middle cerebral artery in rats. J
208. Brea D, Blanco M, Ramos-Cabrer P et al (2011) Toll-like recep- Cereb Blood Flow Metab 23(12):1430–1440
tors 2 and 4 in ischemic stroke: outcome and therapeutic values. J 230. Ajmo CT, Collier LA, Leonardo CC et al (2009) Blockade of
Cereb Blood Flow Metab 31(6):1424–1431 adrenoreceptors inhibits the splenic response to stroke. Exp
209. Caso JR, Pradillo JM, Hurtado O et al (2007) Toll-like receptor 4 is Neurol 218(1):47–55
involved in brain damage and inflammation after experimental 231. Felger JC, Abe T, Kaunzner UW et al (2010) Brain dendritic cells
stroke. Circulation 115(12):1599–1608 in ischemic stroke: time course, activation state, and origin. Brain
210. Yao L, Kan EM, Lu J et al (2013) Toll-like receptor 4 mediates Behav Immun 24(5):724–737
microglial activation and production of inflammatory mediators in 232. Kostulas N, Li H-L, Xiao B-G et al (2002) Dendritic cells are
neonatal rat brain following hypoxia: role of TLR4 in hypoxic present in ischemic brain after permanent middle cerebral artery
microglia. J Neuroinflammation 10(1):23 occlusion in the rat. Stroke 33(4):1129–1134
211. Hyakkoku K, Hamanaka J, Tsuruma K et al (2010) Toll-like re- 233. Yilmaz A, Fuchs T, Dietel B et al (2009) Transient decrease in
ceptor 4 (TLR4), but not TLR3 or TLR9, knock-out mice have circulating dendritic cell precursors after acute stroke: potential
neuroprotective effects against focal cerebral ischemia. recruitment into the brain. Clin Sci 118(2):147–157
Neuroscience 171(1):258–267 234. Saino O, Taguchi A, Nakagomi T et al (2010) Immunodeficiency
212. Lehnardt S, Lehmann S, Kaul D et al (2007) Toll-like receptor 2 reduces neural stem/progenitor cell apoptosis and enhances
mediates CNS injury in focal cerebral ischemia. J Neuroimmunol neurogenesis in the cerebral cortex after stroke. J Neurosci Res
190(1):28–33 88(11):2385–2397
213. Cao C-X, Yang Q-w, Lv F-L et al (2007) Reduced cerebral 235. Santana M, Rosenstein Y (2003) What it takes to become an
ischemia-reperfusion injury in Toll-like receptor 4 deficient mice. effector T cell: the process, the cells involved, and the mecha-
Biochem Biophys Res Commun 353(2):509–514 nisms. J Cell Physiol 195(3):392–401
214. Bohacek I, Cordeau P, Lalancette-Hébert M et al (2012) Toll-like 236. Collison LW, Workman CJ, Kuo TT et al (2007) The inhibitory
receptor 2 deficiency leads to delayed exacerbation of ischemic cytokine IL-35 contributes to regulatory T-cell function. Nature
injury. J Neuroinflammation 9(1):1 450(7169):566–569
215. Akira S (2006) TLR signaling, in from innate immunity to immu- 237. Niedbala W, Wei XQ, Cai B et al (2007) IL-35 is a novel cytokine
nological memory. Springer, Berlin Heidelberg New York, pp 1– with therapeutic effects against collagen-induced arthritis through
16 the expansion of regulatory T cells and suppression of Th17 cells.
216. Kono H, Rock KL (2008) How dying cells alert the immune Eur J Immunol 37(11):3021–3029
system to danger. Nat Rev Immunol 8(4):279–289 238. Shichita T, Sugiyama Y, Ooboshi H et al (2009) Pivotal role of
217. Petrovic-Djergovic D, Goonewardena SN, Pinsky DJ (2016) cerebral interleukin-17–producing γδT cells in the delayed phase
Inflammatory disequilibrium in stroke. Circ Res 119(1):142–158 of ischemic brain injury. Nat Med 15(8):946–950
218. Garcia-Bonilla L, Iadecola C (2012) Peroxiredoxin sets the brain 239. Liesz A, Karcher S, Veltkamp R (2013) Spectratype analysis of
on fire after stroke. Nat Med 18(6):858–859 clonal T cell expansion in murine experimental stroke. J
219. Benakis C, Garcia-Bonilla L, Iadecola C et al (2015) The role of Neuroimmunol 257(1):46–52
microglia and myeloid immune cells in acute cerebral ischemia. 240. Ren X, Akiyoshi K, Dziennis S et al (2011) Regulatory B cells
Front Cell Neurosci 8:461 limit CNS inflammation and neurologic deficits in murine exper-
220. Singh V, Roth S, Veltkamp R et al (2016) HMGB1 as a key imental stroke. J Neurosci 31(23):8556–8563
mediator of immune mechanisms in ischemic stroke. Antioxid 241. Benakis C, Brea D, Caballero S et al (2016) Commensal microbi-
Redox Signal 24(12):635–651 ota affects ischemic stroke outcome by regulating intestinal
221. Marsh BJ, Williams-Karnesky RL, Stenzel-Poore MP (2009) Toll- [gamma][delta] T cells. Nat Med 22:516–523
like receptor signaling in endogenous neuroprotection and stroke. 242. Singh V, Roth S, Llovera G et al (2016) Microbiota dysbiosis
Neuroscience 158(3):1007–1020 controls the neuroinflammatory response after stroke. J Neurosci
222. Anrather J, Iadecola C (2016) Inflammation and stroke: an over- 36(28):7428–7440
view. Neurotherapeutics 1–10 243. Karlsson FH, Fåk F, Nookaew I et al (2012) Symptomatic athero-
223. Schilling M, Strecker J-K, Ringelstein EB et al (2009) The role of sclerosis is associated with an altered gut metagenome. Nat
CC chemokine receptor 2 on microglia activation and blood-borne Commun 3:1245
cell recruitment after transient focal cerebral ischemia in mice. 244. Yin J, Liao SX, He Y et al (2015) Dysbiosis of gut microbiota with
Brain Res 1289:79–84 reduced trimethylamine-N-oxide level in patients with large-artery
224. Konsman JP, Drukarch B, Van Dam A-M (2007) (Peri) vascular atherosclerotic stroke or transient ischemic attack. J Am Heart
production and action of pro-inflammatory cytokines in brain pa- Assoc 4(11):e002699
thology. Clin Sci 112(1):1–25 245. Arpaia N, Campbell C, Fan X et al (2013) Metabolites produced
225. Mantovani A, Sica A, Locati M (2005) Macrophage polarization by commensal bacteria promote peripheral regulatory T-cell gen-
comes of age. Immunity 23(4):344–346 eration. Nature 504(7480):451–455
226. Connolly ES Jr, Winfree CJ, Springer TA et al (1996) Cerebral 246. Zhou W, Liesz A, Bauer H et al (2013) Postischemic brain infil-
protection in homozygous null ICAM-1 mice after middle cerebral tration of leukocyte subpopulations differs among murine perma-
artery occlusion. Role of neutrophil adhesion in the pathogenesis nent and transient focal cerebral ischemia models. Brain Pathol
of stroke. J Clin Invest 97(1):209 23(1):34–44
227. Connolly E, Winfree C, Prestigiacomo C et al (1997) Exacerbation 247. Vandenabeele P, Galluzzi L, Berghe TV et al (2010) Molecular
of cerebral injury in mice that express the P-selectin gene. Circ Res mechanisms of necroptosis: an ordered cellular explosion. Nat
81(3):304–310 Rev Mol Cell Biol 11(10):700–714
228. Soriano SG, Lipton SA, Wang YF et al (1996) Intercellular adhe- 248. Wei L, Ying D-J, Cui L et al (2004) Necrosis, apoptosis and hybrid
sion molecule-1-deficient mice are less susceptible to cerebral death in the cortex and thalamus after barrel cortex ischemia in
ischemia-reperfusion lnjury. Ann Neurol 39(5):618–624 rats. Brain Res 1022(1):54–61
Neurol Sci (2017) 38:1167–1186 1185

249. Ünal-Çevik I, Kılınç M, Can A et al (2004) Apoptotic and necrotic 273. Yin K-J, Deng Z, Huang H et al (2010) miR-497 regulates neuro-
death mechanisms are concomitantly activated in the same cell nal death in mouse brain after transient focal cerebral ischemia.
after cerebral ischemia. Stroke 35(9):2189–2194 Neurobiol Dis 38(1):17–26
250. Adams JM (2003) Ways of dying: multiple pathways to apoptosis. 274. Moon J-M, Xu L, Giffard RG (2013) Inhibition of microRNA-181
Genes Dev 17(20):2481–2495 reduces forebrain ischemia-induced neuronal loss. J Cereb Blood
251. Kroemer G, Galluzzi L, Brenner C (2007) Mitochondrial mem- Flow Metab 33(12):1976–1982
brane permeabilization in cell death. Physiol Rev 87(1):99–163 275. Huang W, Liu X, Cao J et al (2015) miR-134 regulates ischemia/
252. Nikoletopoulou V, Markaki M, Palikaras K et al (2013) Crosstalk reperfusion injury-induced neuronal cell death by regulating
between apoptosis, necrosis and autophagy. Biochimica et CREB signaling. J Mol Neurosci 55(4):821–829
Biophysica Acta (BBA)-Molecular Cell Research 1833(12): 276. Shinoura N, Satou R, Yoshida Yet al (2000) Adenovirus-mediated
3448–3459 transfer of Bcl-X L protects neuronal cells from Bax-induced ap-
253. Culmsee C, Zhu C, Landshamer S et al (2005) Apoptosis-inducing optosis. Exp Cell Res 254(2):221–231
factor triggered by poly (ADP-ribose) polymerase and Bid medi- 277. Zhao H, Yenari MA, Cheng D et al (2003) Bcl-2 overexpression
ates neuronal cell death after oxygen-glucose deprivation and fo- protects against neuron loss within the ischemic margin following
cal cerebral ischemia. J Neurosci 25(44):10262–10272 experimental stroke and inhibits cytochrome c translocation and
254. Broughton BR, Reutens DC, Sobey CG (2009) Apoptotic mech- caspase-3 activity. J Neurochem 85(4):1026–1036
anisms after cerebral ischemia. Stroke 40(5):e331–e339 278. Gonzalez R, Hirsch J, Koroshetz W et al (2007) Acute ischemic
255. Li H, Colbourne F, Sun P et al (2000) Caspase inhibitors reduce stroke: imaging and intervention. Am J Neuroradiol 28(8):1622
neuronal injury after focal but not global cerebral ischemia in rats. 279. Guan Q-H, Pei D-S, Liu X-M et al (2006) Neuroprotection against
Stroke 31(1):176–182 ischemic brain injury by SP600125 via suppressing the extrinsic
2 5 6 . H i c k e y E J , Yo u X , K a i m a k t c h i e v V e t a l ( 2 0 0 7 ) and intrinsic pathways of apoptosis. Brain Res 1092(1):36–46
Lipopolysaccharide preconditioning induces robust protection 280. Guan Q-H, Pei D-S, Zong Y-Y et al (2006) Neuroprotection
against brain injury resulting from deep hypothermic circulatory against ischemic brain injury by a small peptide inhibitor of c-
arrest. J Thorac Cardiovasc Surg 133(6):1588–1596 Jun N-terminal kinase (JNK) via nuclear and non-nuclear path-
257. Kroemer G, Reed JC (2000) Mitochondrial control of cell death. ways. Neuroscience 139(2):609–627
Nat Med 6(5) 281. Kim JS, Kim Y-J, Ahn S-H et al (2016) Location of cerebral
258. Adams JM, Cory S (2001) Life-or-death decisions by the Bcl-2 atherosclerosis: why is there a difference between East and
protein family. Trends Biochem Sci 26(1):61–66 West? Int J Stroke
259. Antonsson B, Montessuit S, Sanchez B et al (2001) Bax is present 282. Ritz K, Denswil NP, Stam OC et al (2014) Cause and mechanisms
as a high molecular weight oligomer/complex in the mitochondrial of intracranial atherosclerosis. Circulation 130(16):1407–1414
membrane of apoptotic cells. J Biol Chem 276(15):11615–11623 283. Suri MFK, Qiao Y, Ma X et al (2016) Prevalence of intracranial
260. Hengartner MO (2000) The biochemistry of apoptosis. Nature atherosclerotic stenosis using high-resolution magnetic resonance
407(6805):770–776 angiography in the general population. Stroke 47(5):1187–1193
261. Zoppo G, Ginis I, Hallenbeck JM et al (2000) Inflammation and 284. Hu X, De Silva TM, Chen J et al (2017) Cerebral vascular disease
stroke: putative role for cytokines, adhesion molecules and iNOS and neurovascular injury in ischemic stroke. Circ Res 120(3):449–
in brain response to ischemia. Brain Pathol 10(1):95–112 471
262. Zoppo GJ (1997) Microvascular responses to cerebral ischemia/ 285. Hollander W, Prusty S, Kemper T et al (1993) The effects of
inflammation. Ann N Y Acad Sci 823(1):132–147 hypertension on cerebral atherosclerosis in the cynomolgus mon-
263. Jin K, Graham SH, Mao X et al (2001) Fas (CD95) may mediate key. Stroke 24(8):1218–1226
delayed cell death in hippocampal CA1 sector after global cerebral 286. Arvanitakis Z, Capuano AW, Leurgans SE et al (2016) Relation of
ischemia. J Cereb Blood Flow Metab 21(12):1411–1421 cerebral vessel disease to Alzheimer’s disease dementia and cog-
264. Namura S, Zhu J, Fink K et al (1998) Activation and cleavage of nitive function in elderly people: a cross-sectional study. The
caspase-3 in apoptosis induced by experimental cerebral ischemia. Lancet Neurology 15(9):934–943
J Neurosci 18(10):3659–3668 287. Roher AE, Esh C, Kokjohn TA et al (2003) Circle of Willis ath-
265. Green DR (2005) Apoptotic pathways: ten minutes to dead. Cell erosclerosis is a risk factor for sporadic Alzheimer’s disease.
121(5):671–674 Arterioscler Thromb Vasc Biol 23(11):2055–2062
266. Ferrer I, Planas AM (2003) Signaling of cell death and cell sur- 288. Gupta A, Iadecola C (2015) Impaired Aβ clearance: a potential
vival following focal cerebral ischemia: life and death struggle in link between atherosclerosis and Alzheimer’s disease. Front
the penumbra. J Neuropathol Exp Neurol 62(4):329–339 Aging Neurosci 16(7):115
267. Seko Y, Kayagaki N, K-i S et al (2002) Role of Fas/FasL pathway 289. Ballinger SW, Patterson C, Knight-Lozano CA et al (2002)
in the activation of infiltrating cells in murine acute myocarditis Mitochondrial integrity and function in atherogenesis.
caused by Coxsackievirus B3. J Am Coll Cardiol 39(8):1399– Circulation 106(5):544–549
1403 290. Napoli C, Witztum JL, de Nigris F et al (1999) Intracranial arteries
268. Ma J, Endres M, Moskowitz MA (1998) Synergistic effects of of human fetuses are more resistant to hypercholesterolemia-
caspase inhibitors and MK-801 in brain injury after transient focal induced fatty streak formation than extracranial arteries.
cerebral ischaemia in mice. Br J Pharmacol 124(4):756–762 Circulation 99(15):2003–2010
269. Graham SH, Chen J (2001) Programmed cell death in cerebral 291. D’armiento FP, Bianchi A, de Nigris F et al (2001) Age-related
ischemia. J Cereb Blood Flow Metab 21(2):99–109 effects on atherogenesis and scavenger enzymes of intracranial
270. Wei N, Xiao L, Xue R et al (2015) MicroRNA-9 mediates the cell and extracranial arteries in men without classic risk factors for
apoptosis by targeting Bcl2l11 in ischemic stroke. Mol Neurobiol atherosclerosis. Stroke 32(11):2472–2480
1–9 292. Wang Z, Roberts AB, Buffa JA et al (2015) Non-lethal inhibition
271. Luo S, Rubinsztein DC (2013) BCL2L11/BIM: a novel molecular of gut microbial trimethylamine production for the treatment of
link between autophagy and apoptosis. Autophagy 9(1):104–105 atherosclerosis. Cell 163(7):1585–1595
272. Sionov RV, Vlahopoulos SA, Granot Z (2015) Regulation of Bim 293. Kinlay S, Michel T, Leopold JA (2016) The future of vascular
in health and disease. Oncotarget 6(27):23058 biology and medicine. Circulation 133(25):2603–2609
1186 Neurol Sci (2017) 38:1167–1186

294. Zhu W, Gregory JC, Org E et al (2016) Gut microbial metabolite 312. Cuccione E, Padovano G, Versace A et al (2016) Cerebral collat-
TMAO enhances platelet hyperreactivity and thrombosis risk. Cell eral circulation in experimental ischemic stroke. Exp Transl Stroke
165(1):111–124 Med 8(1):2
295. Shobha N, Buchan AM, Hill MD et al (2010) Thrombolysis at 3– 313. Jackman K, Iadecola C (2015) Neurovascular regulation in the
4.5 hours after acute ischemic stroke onset–evidence from the ischemic brain. Antioxid Redox Signal 22(2):149–160
Canadian Alteplase for Stroke Effectiveness Study (CASES) reg- 314. Bang OY, Saver JL, Buck BH et al (2008) Impact of collateral
istry. Cerebrovasc Dis 31(3):223–228 flow on tissue fate in acute ischaemic stroke. J Neurol Neurosurg
296. Parmar S, Moore-Langston S, Fredrickson V et al (2015) Psychiatry 79(6):625–629
Neuroprotective mechanisms of oxygen and ethanol: a potential 315. Lima FO, Furie KL, Silva GS et al (2010) The pattern of
combination therapy in stroke. Curr Med Chem 22(10):1194– leptomeningeal collaterals on CT angiography is a strong predic-
1204 tor of long-term functional outcome in stroke patients with large
297. Geng X, Fu P, Ji X et al (2013) Synergetic neuroprotection of vessel intracranial occlusion. Stroke 41(10):2316–2322
normobaric oxygenation and ethanol in ischemic stroke through 316. Shuaib A, Butcher K, Mohammad AA et al (2011) Collateral
improved oxidative mechanism. Stroke 44(5):1418–1425 blood vessels in acute ischaemic stroke: a potential therapeutic
298. Geng X, Parmar S, Li X et al (2013) Reduced apoptosis by com- target. The Lancet Neurology 10(10):909–921
bining normobaric oxygenation with ethanol in transient ischemic
317. Mergenthaler P, Dirnagl U (2011) Protective conditioning of the
stroke. Brain Res 1531:17–24
brain: expressway or roadblock? J Physiol 589(17):4147–4155
299. Geng X, Sy CA, Kwiecien TD et al (2015) Reduced cerebral
monocarboxylate transporters and lactate levels by ethanol and 318. Hess DC, Hoda MN, Bhatia K (2013) Remote limb perconditioning
normobaric oxygen therapy in severe transient and permanent and postconditioning. Stroke 44(4):1191–1197
ischemic stroke. Brain Res 1603:65–75 319. Stevens SL, Vartanian KB, Stenzel-Poore MP (2014) Reprogramming
300. Choi K-E, Hall CL, Sun J-M et al (2012) A novel stroke therapy of the response to stroke by preconditioning. Stroke 45(8):2527–2531
pharmacologically induced hypothermia after focal cerebral ische- 320. Pignataro G, Scorziello A, Di Renzo G et al (2009) Post-ischemic
mia in mice. FASEB J 26(7):2799–2810 brain damage: effect of ischemic preconditioning and
301. Katz LM, Young AS, Frank JE et al (2004) Regulated hypother- postconditioning and identification of potential candidates for
mia reduces brain oxidative stress after hypoxic-ischemia. Brain stroke therapy. FEBS J 276(1):46–57
Res 1017(1):85–91 321. Zhao H, Joo S, Xie W et al (2013) Using hormetic strategies to
302. Truettner JS, Suzuki T, Dietrich WD (2005) The effect of thera- improve ischemic preconditioning and postconditioning against
peutic hypothermia on the expression of inflammatory response stroke. Int J Physiol Pathophysiol Pharmacol 5(2):61–72
genes following moderate traumatic brain injury in the rat. Mol 322. Meng R, Asmaro K, Meng L et al (2012) Upper limb ischemic
Brain Res 138(2):124–134 preconditioning prevents recurrent stroke in intracranial arterial
303. Lee JH, Wei L, Gu X et al (2014) Therapeutic effects of pharma- stenosis. Neurology 79(18):1853–1861
cologically induced hypothermia against traumatic brain injury in 323. Stowe AM, Wacker BK, Cravens PD et al (2012) CCL2 upregu-
mice. J Neurotrauma 31(16):1417–1430 lation triggers hypoxic preconditioning-induced protection from
304. Polderman KH, Joe RTT, Peerdeman SM et al (2002) Effects of stroke. J Neuroinflammation 9(1):33
therapeutic hypothermia on intracranial pressure and outcome in 324. Ouk T, Laprais M, Bastide M et al (2009) Withdrawal of
patients with severe head injury. Intensive Care Med 28(11):1563– fenofibrate treatment partially abrogates preventive neuroprotec-
1573 tion in stroke via loss of vascular protection. Vasc Pharmacol
305. Lee JH, Wei ZZ, Cao W et al (2016) Regulation of therapeutic 51(5):323–330
hypothermia on inflammatory cytokines, microglia polarization, 325. Khan MB, Hoda MN, Vaibhav K et al (2015) Remote ischemic
migration and functional recovery after ischemic stroke in mice. postconditioning: harnessing endogenous protection in a murine
Neurobiol Dis 96:248–260 model of vascular cognitive impairment. Translational stroke re-
306. Zausinger S, Schöller K, Plesnila N et al (2003) Combination drug search 6(1):69–77
therapy and mild hypothermia after transient focal cerebral ische- 326. Stowe AM, Altay T, Freie AB et al (2011) Repetitive hypoxia
mia in rats. Stroke 34(9):2246–2251 extends endogenous neurovascular protection for stroke. Ann
307. Kollmar R, Henninger N, Bardutzky J et al (2004) Combination Neurol 69(6):975–985
therapy of moderate hypothermia and thrombolysis in experimen-
327. Bowen KK, Naylor M, Vemuganti R (2006) Prevention of inflam-
tal thromboembolic stroke—an MRI study. Exp Neurol 190(1):
mation is a mechanism of preconditioning-induced neuroprotec-
204–212
tion against focal cerebral ischemia. Neurochem Int 49(2):127–
308. Zhao H, Shimohata T, Wang JQ et al (2005) Akt contributes to
135
neuroprotection by hypothermia against cerebral ischemia in rats.
J Neurosci 25(42):9794–9806 328. Joo SP, Xie W, Xiong X et al (2013) Ischemic postconditioning
309. Zhao H, Yenari MA, Sapolsky RM et al (2004) Mild postischemic protects against focal cerebral ischemia by inhibiting brain inflam-
hypothermia prolongs the time window for gene therapy by mation while attenuating peripheral lymphopenia in mice.
inhibiting cytochrome C release. Stroke 35(2):572–577 Neuroscience 243:149–157
310. Faillace MP, Keller Sarmiento MI, Rosenstein RE (1996) Melatonin 329. Dai Y, Li W, Zhong M et al (2014) Preconditioning and post-
effect on [3H] glutamate uptake and release in the golden hamster treatment with cobalt chloride in rat model of perinatal hypoxic–
retina. J Neurochem 67(2):623–628 ischemic encephalopathy. Brain and Development 36(3):228–240
311. Qian Y, Tang X, Guan T et al (2016) Neuroprotection by combined 330. Pignataro G, Meller R, Inoue K et al (2008) In vivo and in vitro
administration with maslinic acid, a natural product from Olea characterization of a novel neuroprotective strategy for stroke:
europaea, and MK-801 in the cerebral ischemia model. ischemic postconditioning. J Cereb Blood Flow Metab 28(2):
Molecules 21(8):1093 232–241

View publication stats

You might also like