Abstract
Stroke is a frequent cause of long-term disability and death worldwide. Ischemic stroke is more commonly encountered compared to hemorrhagic stroke, and leads to tissue death by ischemia due to occlusion of a cerebral artery. Inflammation is known to result as a result of ischemic injury, long thought to be involved in initiating the recovery and repair process. However, work over the past few decades indicates that aspects of this inflammatory response may in fact be detrimental to stroke outcome.
Acutely, inflammation appears to have a detrimental effect, and anti-inflammatory treatments have been been studied as a potential therapeutic target. Chronically, reports suggest that post-ischemic inflammation is also essential for the tissue repairing and remodeling. The majority of the work in this area has centered around innate immune mechanisms, which will be the focus of this review. This review describes the different key players in neuroinflammation and their possible detrimental and protective effects in stroke. A better understanding of the roles of the different immune cells and their temporal profile of damage versus repair will help to clarify more effective modulation of inflammation post stroke. Introduction Stroke refers to conditions caused by occlusion and/or rupture of blood vessels in the brain, and is a leading cause of death and disability in the industrialized world.
Keywords: brain ischemia, inflammation, neuroprotection, stroke
Ischemic strokes represent more than 80% of all cases of stroke and are characterized by the occlusion of a brain arterial blood vessel due to a thrombus or embolus [1]. In spite of its high prevalence, effective therapies are few [2] [1]. Brain inflammation has been implicated as a secondary injury mechanism following ischemia and stroke [3] [4] [5]. Stroke triggers this inflammatory response as a result of several factors, such as necrotic cells and debris and reactive oxygen species (ROS) and many other factors which have yet to be precisely identified. These triggering factors lead to microglial activation, leading to more cytokine generation and induction of adhesion molecules within the cerebral vasculature, all within 24 hours of the ischemic insult [6] [7] [8]. Chemokine upregulation stimulates inflammatory cell chemotaxis into ischemic brain, especially around the penumbra, or the infarct’s border. Adhesion molecules on activated endothelia in turn leads to the adhesion of circulating leukocytes causing microvascular occlusion and infiltration of immune cells into the brain parenchyma [9] [10]. Activated inflammatory cells then elaborate a variety of cytotoxic molecules such as more pro-inflammatory cytokines, matrix metalloproteinases (MMPs), nitric oxide (NO) and more ROS. These molecules in turn potentiate brain cell damage, including disruption of the blood-brain barrier (BBB) and extracellular matrix [11].
A disrupted BBB can further exacerbate brain injury, contributing to secondary ischemic damage by permitting serum elements and blood to enter the brain. This often leads to brain edema and hemorrhagic transformation. Brain ischemia is also thought to influence immune cells in the circulation possibly through increased activation of the sympathetic nervous system (SNS) and the hypothalamic-pituitary-adrenal axis (HPA). This may lead to fewer circulating immune cells, and increase the risk of infectious complications [12]. Leukocyte plugging of the brain’s microvasculature has also been implicated in microvascular stasis leading to hypoperfusion.
Inhibiting variousinflammatory molecules has shown to reduce injury in experimental stroke models 13], although this has yet to be shown in human stroke patients.
In contrast, other immune responses may be beneficial to the ischemic brain by secreting neurotrophic factors and by scavenging necrotic debris allowing for the reorganization of a new environment for neural repair [14] [15] [16] [17]. These reparative responses are thought to be especially relevant during the chronic phases of stroke. Thus, complete inhibition of immune responses could be predicted to impede recovery. While both acutely damaging and chronically reparative properties of post stroke inflammation have been embraced for quite some time, the precise nature of these dual properties has not been well defined. A better understanding of this tight balance may help better tailor both anti- and pro- immune therapies for maximum effectiveness.
Over the past few years, there have been advances in inflammatory signaling molecules in brain ischemia. This review focuses on recent findings as they largely pertain to innate immune responses and mechanisms in brain ischemia, plus possible therapeutic targets.
Entry of immune cells into the ischemic brain
Inflammation is traditionally defined in histological terms by the presence of immune cells in the tissue of interest. Inflammatory cells have long been documented in autopsy specimens of patient who suffered stroke. Circulating immune cells gain access to the ischemic brain beginning with rapid upregulation of adhesion molecules [18]. Circulating immune cells can then gain access into the injured brain and elaborate immune molecules that exacerbate ischemic cell death. Ischemic brain cells also activate endogenous immune cells such as microglia. A few studies implicate astrocytes as another brain resident immune cell which also activate in response to ischemia.
The initial vascular response to ischemic includes activation of the endothelium within and around the ischemic brain, as well as activation of circulating leukocytes. Ischemia leads to the upregulation and activation of a family of adhesion molecules involved in acute inflammatory responses, and permit interactions between endothelial cells, platelets, leukocytes, and lymphocytes, and leads to the infiltration of immune cells into the brain parenchyma after stroke [19]. Migration of these cells into the brain is mediated by cell adhesion molecules: selectins, integrins, and immunoglobulins, the expression of which is regulated both intracellularly and by cytokine signaling [20]. Activated leukocytes, namely neutrophils in the acute stages, home to the site of ischemic injury through interactions between the various adhesion molecules including the selectins (P-selectin, E-selectin, and L-selectin), immunoglobulin superfamily (e.g., ICAM-1, or intercellular cell adhesion molecule-1) and integrins (CD11a, b and c) [21] [22].
Selectins are calcium-dependent, transmembrane glycoproteins that bind to carbohydrate residues (sialyl-Lewisx), and mediate rolling and adhesion to vascular endothelium. E-, P-, and L-selectin have all been shown to be involved in leukocyte trafficking in brain ischemia [23] [10] [24]. E-selectin [25] [26], or P-selectin [9] [27] have been shown to participate in initial leukocyte rolling and recruitment [9] [25] [26] [28], whereas L-selectin guides unstimulated leukocytes to areas of activated endothelium [29]. P- and E- selectin expression have been documented in various experimental stroke models and their upregulation appears to be involved in promoting both ischemic inflammatory responses and injury [9] [26] [30] [31] [32] [33]. In mice overexpressing P-selectin, larger infarcts have been documented. Similarly, P- and E-selectin inhibition or blocking was associated with improved neurological outcome [34] [30] [35]. However, how L-select participates in brain ischemia is less clear. L-selectin is involved in the immediate steps leading to leukocyte transmigration. When it encounters activated endothelium, it sheds from the leukocyte surface prior to entry into ischemic brain. Yet, inhibiting L-selectin shedding does not consistently influence stroke outcome. In a rabbit model of transient focal brain ischemia, treatment with an L-selectin antibody did not affect stroke outcome [36].
Once leukocytes have entered the ischemic brain, members of the immunoglobin superfamily permit binding of activated leukocytes to activated endothelium. These molecules contribute to the inflammatory response by attaching immune cells tightly to the endothelial wall, and facilitate and even stimulate diapedesis through the vessel wall to the site of injury [20] [37]. Among the immunoglobulin superfamily members, ICAM-1 and vascular cellular adhesion molecule-1 (VCAM-1) have been the most investigated in brain ischemia [7]. Expression of ICAM-1 increases soon after ischemia following activation by pro-inflammatory cytokines [7] [38] [11]. ICAM-1 is increased in ischemic brain within hours after stroke onset, and peaks at about 12–48 h [7] [38] [11] [39] [40]. Animals deficient in adhesion molecules, or treated with strategies that block ICAM-1 have decreased ischemic damage and less brain leukocyte infiltration in experimental stroke [41] [42] [43] [44] [45] [46] [47] [48] [7]. Further, mice lacking ICAM-1 had smaller infarcts compared to wild-type mice [49] [42].
VCAM-1’s contribution to stroke is less clear. Some groups have shown that VCAM-1 mRNA and protein both increase after cerebral ischemia [50] [51], whereas others found no change in expression [47]. ONO-1078, a leukotriene receptor antagonist, improved neurological deficits and reduced neuronal death in a model of forebrain ischemia, and this was correlated to decreased VCAM-1 upregulation in the hippocampus [52]. Another study showed that unfractionated heparin reduced infarct size in a stroke model, and this was associated with decreased VCAM-1 expression and suppression of the inflammatory response [53]. Liesz el al. [48] also found that knocking down VCAM-1 mRNA with siRNA led to reduced T lymphocyte infiltration in to the brain and decreased infarct volume after experimental stroke. However, there are conflicting results, as other studies did not show any beneficial outcome using anti-VCAM-1 antibodies [51] [48].
Integrins are adhesion molecules composed of heterodimeric combination of various α and β subunits. Prior to leukocytes binding, integrins are expressed on cell surfaces and will subunits will associate in order to recognize endothelial cell adhesion molecules [54]. Almost all leukocytes express CD11a/CD18 (leukocyte function–associated antigen-1 [LFA-1]) and CD11b/CD18, also known as Mac-1, which are integrins that contain a common β2 chain (CD18) and are thus known as β2 integrins. These integrins allow the cells to bind to endothelial ICAM-1 and migrate through the vessel. In addition to the β2 integrins, lymphocytes and monocytes express α4β1 (CD49d/CD29) and α4β7 (CD49d/CD103).
Several groups have shown that preventing neutrophil improves outcome from experimental stroke [55] [56] [57] [58] [59], but blocking integrins involved in lymphocyte and monocyte trafficking has also similar effects [60] [48]. Treatment of experimental stroke with anti-α 4 integrin antibody led to smaller infarct size and reduced neurological deficits [60] [61].
Blood brain barrier disruption: entry of immune cells and brain edema and hemorrhage
Disruption of the blood-brain barrier (BBB) after ischemic stroke permits infiltration of circulating immune cells into the brain, and also exacerbates edema and hemorrhage. This leads to functional impairment of the so-called ‘neurovascular unit’ (the functional unit including neurons, supporting glia and the cerebral vasculature as an integrated whole), which includes basement membrane tight junction proteins, transport proteins, endothelial cells, astrocytes and neurons. Molecules involved in these pathologies include the matrix metalloproteinases MMPs), cyclooxygenase (COX), nitric oxide (NO) and reactive oxygen species (ROS).
Endothelial cells are actively engaged in processes of microvascular stasis and are the first cells which face the impact of ischemia. When damaged by ischemic stimuli, endothelial cells swell or detachment from the underlying basement membrane, leading to compromises in barrier function. This leads to increased BBB permeability causing serum protein extravasation and interstitial edema as well as entry of immune molecules and cells [18] [62]. Endothelial cells form the BBB through tight junction (TJ) proteins. Among a number of TJ proteins (claudins, occludin, and zonula occludens proteins ZO-1, ZO-2, and ZO-3), major TJ proteins are occludin and claudins, closing to the blood and junctional adhesion molecules deeper in the endothelial cell clefts [63]. Stabilization of TJ involves a complex of claudins and occludins and ZO proteins linking the transmembrane to the actin cytoskeleton. A basal lamina is composed mainly of type IV collagen, fibronectin, heparan sulfate, and laminin enveloped the abluminal surface of the endothelial cell. These function as a charge and molecular weight barrier and interact in complex ways with integrins to regulate permeability and cellular transport across the BBB [64]. Pericytes, which embedded in the basal lamina, functions a hybrid cells with both macrophage and smooth muscle properties [65]. The disruption of the BBB facilitates the entry of free water and serum from the intravascular compartment, and increases brain edema. At its most extreme, BBB disruption can lead to hemorrhage. Experimental strategies to prevent BBB disruption have largely involved the inhibition of matrix metalloproteinases (MMPs) and endogenous tissue plasminogen activator (rt-PA), proteases known to cleave and disrupt the extracellular matrix. Treatment with MMP inhibitors [66] [67] or neuroserpin, an endogenous inhibitor of rt-PA [68] [69] [70], led to improved neurological outcome and decreased cerebral hemorrhage.
Leukocyte damage to the ischemic brain
Following ischemia, infiltrated leukocytes release proinflammaroty mediators into the area of ischemia.Neutrophils initially enter the ischemic brain having been observed at about 6–12 h in models of transient focal ischemia [71] [72] [24]. Leukocytes promote cerebral ischemic injury in several different ways. First, adhesion of leukocytes to the endothelium can impair the flow of erythrocytes through the microvasculature causing the cerebral no-reflow phenomenon and additional ischemic injury [62]. Second, activated leukocytes at the surface of the endothelium produce ROS, proteases, gelatinases, and collagenases, and damage potentially salvageable blood vessels and brain tissues. Third, phospholipase activation in leukocytes results in the production of biologically active substances like leukotrienes, eicosanoids, prostaglandins, and platelet-activating factor, which can cause vasoconstriction and increase platelet aggregation. Finally, infiltrated leukocytes elaborate proinflammatory cytokines and other immune molecules in around the penumbra surrounding the infarct core causing further neuronal injury [73] [74] [45].
While the data are conflicting, lymphocytes have largely been shown to negatively contribute to ischemic brain pathogenesis. Like neutrophils, lymphocytes are also sources of pro-inflammatory cytokines and cytotoxic substances, such as ROS. A few studies in stroke models have shown that lymphocytes are elevated in the ischemic brain later than neutrophils (3 to 6 days post stroke) [75] [76]. Blocking lymphocyte entry into ischemic brain decreased injury, and suggests that like neutrophils, lymphocytes also play a deleterious role [60]. T lymphocytes, but not B lymphocytes are now considered to be the central to the development of inflammation in stroke models [77] [78]. Several recent studies have evaluated the role of T lymphocyte deficient mice in the transient focal ischemia model, and have consistently reported a smaller infarct volume and improved functional outcome than in control groups [79] [78] [80] [81]. Protection observed in lymphocyte-deficient mice subjected to stroke appears to be due to the lack of T lymphocytes, and not B lymphocytes, as the reconstitution of B lymphocytes does not affect the protection observed. By contrast, when T lymphocytes are transplanted back in to Rag1−/− mice, this protection was lost [78] [80] [81]. However, Saino el al. [82] failed to see significant differences infarct size between immunodeficient mice (deficient in both T and B lymphocytes) and wildtype. The reason for these differences is unclear, although the latter study used a model of permanent rather than temporary focal cerebral ischemia.
However, not all T lymphocytes subtypes are detrimental to acute stroke outcome. A recent study showed that neither T lymphocytes nor natural killer (NK) cells contribute to stroke injury [78]. Furthermore, the role of regulatory T (Treg) lymphocytes is still in question. Liesz et al. [83] showed that infarct volume and neurological deficit were significantly increased in mice given an antibody to neutralize Treg lymphocytes compared to controls. They also suggested that IL-10 signaling may be essential for this immunomodulatory effect. However, Ren et al. [84] could not find any modulatory effect of Treg cells. In addition, there is now evidence that resident NK cell function in the liver is profoundly impaired due to augmented sympathetic neurotransmission following stroke, and that this loss of NK cell activity substantially contributes to the immunosuppression and susceptibility to infections that occur following stroke [85].
Clinical studies also support a damaging role for lymphocytes. One study showed that increases in circulating lymphocytes correlated to an increased risk of stroke recurrence and death [86]. However, in a model of ischemia-like necrotic injury to cultured primary hippocampal neurons, isolated neutrophils worsened neuronal injury due to excitotoxin exposure, whereas lymphocytes were not neurotoxic and actually increased astrocyte proliferation [87].
The precise mechanisms of lymphocyte-mediated brain injury are currently unclear. Classically, T lymphocytes kill bacteria- and virus-infected cells either by the release of cytokines, or cytotoxins [88], and similar actions probably occur at the site of ischemic injury. Alternatively, T lymphocytes may cause cell death via interaction with the Fas receptor [89], and a few groups showed that neutralization of T lymphocyte-derived cytokines (IL-17, IL-12, IL-23, interferon gamma) decreased infarct volume and improved neurological outcome in stroke models [90] [80] [83]. Moreover, stroke in perforin-deficient mice show significant neuroprotection, suggesting that perforin, released by T lymphocytes also contributes to ischemic damage [48]. In addition to these studies, the recent evidence showed that T lymphocytes may contribute to oxidative tissue injury following stroke, potentially via NADPH oxidase type 2 (Nox-2)-derived superoxide. T lymphocytes are known to contain a functional Nox-2 oxidase and, soon after the ischemic stroke, circulating T cells produce 7 to 15 fold greater amounts of Nox2-derived superoxide than from the control mice [91].
Microglia/Macrophages & Astrocytes
Microglia represent anywhere from 5–20 % of the total glial population and are key modulators of the immune response in the brain [92]. Microglia are often considered the brain’s resident immune cell [93]. Of myeloid origin, microglia can undergo morphologic transformation from a resting state referred to as “ramified” to an “amoeboid” state, where they become virtually indistinguishable from circulating macrophages [92] [94]. Therefore, activated microglia are often called brain macrophages. Through its phagocytic properties, microglia clear foreign organisms as well as injured brain cells [71] [95] [96] [97]. Cerebral ischemia also induces microglial activation, however, the precise mechanisms of its activation following ischemia are not completely understood. Activated microglia have been documented in the cerebral cortex of the ischemic hemisphere of animals exposed to transient focal cerebral ischemia [107] [96]. Accumulating data shows that CD14 receptors, followed by toll-like receptor 4 (TRL4) have been documented in activated microglia in the infarct brain and could be one of the mechanisms involved in its activation [98] [99] [100]. Activated macrophages can be detected as early as 2 hours after ischemia, whereas blood-derived macrophages do not enter the brain before 10 hours. By 22–46 hours after the insult, activated microglia and macrophages are distributed throughout the entire lesion and are detectable up to 1 week after the insult [39] [72] [101] [102] [103]. Once, activated, microglia are thought to release a variety of inflammatory and cytotoxic mediators contributing to cell damage and cell death [104] [95]. Treatment with edaravone, a free radical scavenger which acts as a mimic to glutathione peroxidase (GPx), was found to be neuroprotective against brain ischemia mice by decreasing microglial activation [105]. Multiple hyperbaric oxygen (HBO) treatments similarly reduced infarct volume by suppressing microglia activation [106]. Minocycline, a tetracycline family antibiotic, has now been shown by a few grops to provide significant protection against brain ischemia through its ability to inhibit microglial activation [108] [109]. More direct evidence of a damaging role of microglia/macrophages was demonstrated when their direct application potentiated neuron cell death [110] [100] [96] [111]. However, microglia are also a major producer of the growth factor TGF-β1, which is generally believed to be neuroprotective [112] [95]. When microglial proliferation was inhibited in transgenic mice, infarct size was increased following ischemia, and suggests that proliferating microglia cells exert a beneficial role [113]. There are some possible mechanisms underlying these observations. First, microglia produce neurotrophic factors which stimulate neurogenesis and plasticity. Secondly, phagocytosis of neutrophils by activated microglia may prevent the release of toxic mediators [114] [115]. Finally, resident macrophages scavenge and remove necrotic debris and other potentiall harmful substances [115].
Astrocytes exert many active roles in brain homeostasis, including the regulation of immune reactions. In addition to traditional immune cells, astrocytes have also been documented to express various inflammatory mediators [116] [117]. Following brain ischemia, astrocytes are capable of activation which leads to upregulation of glial fibrillary acidic protein (GFAP). Astrocytes also contribute reactive gliosis and glial scar formation [118]. Astrocytic gliosis can be destructive after injury [119] [71]. A massive astroglial response starts in the core of the lesion from 4 hours to 1 day after the insult, and reaches a peak around 4 days and is observed up to 28 days after stroke onset [120] [121]. This glial scar has both neurotoxic and neurotrophic properties. The scar can function as a barrier which prevents axonal ingrowth and reinnervation, thus impending recovery. However, while this scar also isolates damaged tissue from viable tissue, and prevents additional damage to the surrounding brain [120]. Astrocytes are also capable of expressing various immune molecules, such as cytokines, chemokines and indicible nitric oxide synthase (iNOS) and can develop a Th2 (anti-inflammatory) immune response, although this has yet to be demonstrated in brain ischemia [122]. In a model of global cerebral ischemia, iNOS was detected in reactive hippocampal astrocytes [123]. Furthermore, astrocyte generated iNOS has been shown to exacerbate ischemia-like injury to neurons [32]. The inflammatory role of astrocytes was demonstrated in a study of tumor necrosis factor like weak inducer of apoptosis (TWEAK), a member of the tumor necrosis factor superfamily. TWEAK has been detected on neurons, astrocytes and endothelial cells, and, through interaction with the astrocytic Fn14 receptor, stimulates proinflammatory molecule production [124] [125] [126]. TWEAK and Fn14 expression have been documented in rodent stroke models, and Fn14 blockade led to reduced ischemic injury [125]. These data indicate that while astrocytes have long been viewed to play scaffolding and supportive roles for neurons, activated astrocytes can play a similarly detrimental role as traditional immune cells.
Mediators of Inflammation
Activated immune cells elaborate numerous substances which mediate inflammation. They include a variety of bioactive molecules including ROS, NO, cytokines, chemokines, prostaglandins, leukotrienes, and platelet-activating factor (PAF) [127] [40] [7] [128]. These molecules all appear to act directly or indirectly to lead to worsened brain cell death. However, other molecules, such as families of trophic factors, appear important in recovery and repair, and several anti-inflammatory cytokines have been documented to limit the pro-inflammatory response and improve outcome from stroke.
Cytokines
Increased cytokine production has been documented in the brain following various acute insults including stroke. Microglia and infiltrating leukocytes are known to elaborate cytokines, but some reports have shown that cytokines can also be produced by resident brain cells following brain ischemia, including glia and neurons, [129] [79] and in humans [130]. Interleukin-1 (IL-1), TNF-α, interleukin-6 (IL-6), interleukin-10 (IL-10) and transforming growth factor-β (TGF-β) have been the most extentively studied cytokines in brain ischemia [13]. Of these cytokines, IL-1 has been shown to mediate ischemic, excitotoxic and traumatic brain injury, probably through multiple actions on glia, neurons and the vasculature, while TNF-α might contribute to both neuronal injury and protection. IL-10 and TGF-β may be neuroprotective [131]. Reports of the role of IL-6 appear to be conflicting [132] [133] [54] [134].
IL-1 has been strongly implicated in the pathogenesis of ischemic brain as a neurotoxic mediator. There are two IL-1 isoforms, IL-1α and IL-1β, plus an endogenous inhibitor, IL-1 receptor antagonist (IL-1ra) which have been studied in brain ischemia models [135] [136]. IL-1 has been shown to increase after ischemia [137] [138], and IL-1β was found to have a biphasic expression pattern, increasing initially during early reperfusion (1 h), then peaked again at a later timepoint (6–24 h) [139] [140]. IL-1β, rather than IL-1α is considered to be more engaged in the ischemic pathogenesis [141]. Administering IL-1β to rats exposed to focal cerebral ischemia led to worsened outcome, indicating a damaging role for the protein [142]. Similarly, mice deficient in IL-1 had smaller infarcts compared to wildtype [141]. Reperfusion in diabetic rats also demonstrated higher protein level of IL-1 compared to wild type and may explain the worsening of ischemic damage by diabetes [143]. IL-1β generation results from conversion from its pro-form by IL-1β converting enzyme (ICE-1, a member of the caspase family) to its active form [144]. IL-1 has two receptors, IL-1R1, which is involved in signal transduction and IL-1R2, which has no known function but may serve as a decoy receptor [138]. In a model of brain hypoxia-ischemia, IL-1R1 deficiency or inhibition decreased brain damage and improved neurological function [145]. Overexpression or treatment with the IL-1ra, IL-1’s endogenous inhibitor, also led to neuroprotection [146] [147]. Similarly, mice deficient in ILL-1ra had worsened ischemic damage after experimental stroke [135]. Further, excitotoxic injury due to NMDA or AMPA was higher in neuron-astrocyte co-cultures derived from IL-1ra deficient mice [135]. TNF-α has also been shown to have similar expression patters and IL-1 β following brain ischemia [140], with initial upregulation 1–3 h post ischemia, and a second and peak at 12–24h [38][148] [149]. TNF-α expression has been observed in neurons [129], astrocytes [150] in addition to the peripheral immune system [149] in stroke models.
Although TNF-α and IL-1β often work synergistically, TNF-α seems to have both neurotoxic and neuroprotective effects, while IL-1β seems generally neurotoxic [151] [152].
TNF-α inhibition has been shown to be neuroprotective against ischemic brain injury [153], while treatment with recombinant TNF-α protein post ischemia onset appears to exacerbate brain damage [154]. However, other work indicates that TNF-α may also be neuroprotective under certain circumstances. TNF-α has been linked to the phenomenon of ischemic tolerance, whereby preischemic treatment leads to improved outcome [155]. Consistent with a beneficial role of TNF, mice lacking TNF receptors have larger infarcts [156]. TNF-α released in the striatum leads to neurodegeneration, while release in the hippocampus may promote neuroprotection [157]. TNF-α stimulates apoptosis of endothelial cells and contributes to vasogenic edema and infiltration of circulatory immune cells are stimulated by this BBB breakdown. On the other hand, TNF-α activates repair processes of the cerebral microvasculature and mediates neuronal plasticity [157]. The reasons for this disparity are still unknown, but a few hypotheses have been proposed. First, TNF-α ‘s actions may depend on the timing. It appears to contribute to detrimental effects in the early phase of the inflammatory response, but may have more beneficial effects at a later stage [158], although this does not explain why TNF-α seems to underlie the phenomenon of tolerance. Another hypothesis relates to the receptors to which TNF-α binds. Soluble TNF-α which binds to TNF receptor 1 primarily leads to detrimental effects, whereas membrane bound TNF-α which binds to TNF receptor 2 leads to neuroprotection [95]. Other studies suggest that TNF receptor 1 signal pathways are also neuroprotective [159].
The precise role of IL-6 in the ischemic stroke has not been clearly identified. IL-6 has been shown to increase its expression continuously up to 24 hours after ischemia onset [38]. However, ischemic brain damage was not attenuated in IL-6 deficient mice or in IL-6 receptor antagonist treated mice compared to wildtype, and suggests that it probably does not contribute significantly to ischemic pathogenesis [132] [134]. Yet, Herrmann et al. [133] reported a beneficial effect of IL-6, while Smith et al. [160] showed detrimental effects. There are also reports showing strong correlation between serum IL-6 levels and in-hospital mortality rates in stroke patients [160] [161]. There has recently been a new focus on brain derived IL-6 where it appears to contribute to neoangiogenesis and neuronal survival through STAT3 activation and manganese-superoxide dismutase [162] [163].
IL-10 and IL-4 are anti-inflammatory cytokines. IL-10 acts by inhibiting proinflammatory cytokines such as IL-1 and TNF-α. IL-10 also by suppresses cytokine receptor expression and downstream signalling, and is upregulated in microglia and astrocytes following experimental stroke [164]. Both exogenous administration [165] and overexpression by gene transfer [166] of IL-10 in stroke models appears to be neuroprotective. IL-4 acts to differentiate T lymphocytes towards a Th2, or anti-inflammatory phenotype. IL-4 deficient mice were observed to have worsened outcome after experimental stroke with exacerbated pro-inflammatory responses [167].
TGF-β1 has been observed in microglia and astrocytes, with low levels in neurons [168]. TGF-β1 overexpression using an adenoviral vector improved outcome from experimental stroke, and this correlated to a reduced inflammatory response [169]. In some model systems, microglia were observed to protect cultured neurons from ischemia-like insults by secreting TGF-β1 [170].
Chemokines
Chemokines are chemotactic cytokines and, together with their receptors expressed on leukocytes, they play a crucial role in the extravasation and migration of leukocytes under inflammatory conditions. Chemokines are expressed by injured neurons, astrocytes, microglia, and endothelial cells, as well as circulating immune cells [117] [171] [172] [79]. Different classes of chemokines are differentiated by their structures, the main classes being CXC or CC, C, CX3C. The “Cs” refer to the two N-terminal cysteine residues, and the classes are divided depending on whether there is an amino acid between them (CXC), or whether they are adjacent (CC). Like cytokines, chemokines act through both unique and overlapping receptors, and these receptors are a part of a superfamily of G-protein-coupled receptors [173] [174]. The CXC subfamily can be further split into ELR+ or ELR− groups based on whether the glutamate–leucine–arginine motif is present between the N-terminus and the first cysteine [174] [175]. Members of the chemokine superfamily tend to bind to several receptors, and a chemokine receptor possibly binding multiple ligands [176]. However, the ELR+ CXC chemokine subfamily are thought to be mainly neutrophil chemoattractants, whereas the CC chemokines more typically attract monocytes and T lymphocytes [177] [79]. Several chemokines in CXC group have been shown to participate in stroke pathogenesis [178] [77] by mediating leukocyte infiltration. Because of this, chemokines have also been implicated in the worsening of stroke outcome [22]. Thus, chemokine ligands and receptors are potential therapeutic targets. Brait el al. [77] showed large increases in expression (by 10- to 300-fold) of key members of the ELR+ CXC chemokine subfamily, the neutrophil receptor CXCR2, and its ligands CXCL1 and CXCL2 which are mostly reached maximum at 24 to 72 hours. As predicted, pharmacological inhibition of CXCR2 following transient focal ischemia prevented the increases in the expression of these genes as well as neutrophil infiltration into the brain; however, this treatment had no effect on functional outcome, infarct or edema volume at 72 h after stroke.
In stroke models, CC chemokines such as monocyte chemoattractant protein-1 (MCP-1,CCL2), macrophage inflammatory protein-1α (MIP-1α, CCL-3), regulated on activation, normal T-cell expressed and secreted (RANTES, CCL5), and macrophage inflammatory protein-3 alpha (MIP-3α) have been documented to increase in expression [38] [117] [179] [180]. At baseline, MCP-1 mRNA expression was almost absent, but ischemia led to a significant increase in MCP-1 mRNA expression in the ischemic cortex after either permanent or temporary MCAO around 12 h to 2 days and remained elevated up to 5 days [171] [172]. Inhibition or deficiency of these chemokines leads to reduced injury [181]. Similarly, overexpression of MCP-1 worsened ischemic brain injury, and this was associated with increased infiltration of inflammatory cells [182].
Fractalkine (CX3CL1) is a neuronally expressed chemokine, acts through its receptor CX3C. Its expression has been localized to viable neurons, and in brain ischemia model, it is largely localized to neurons of the infarct periphery. Its receptor, CX3CR1 was observed exclusively on microglia/macrophages, and suggests that fractalkine may be involved in neuron-microglial signaling [183]. Fractalkine deficient mice subjected to focal cerebral ischemia also have smaller infarct sizes and better outcomes compared to wildtype mice, and indicate that fractalkine contributes negatively to ischemic pathology [184]. In addition to their chemotactic properties, chemokines were discovered to directly affect the BBB. For example, when MCP-1 was added to cocultures of endothelial cells and astrocytes, this was associated loss of tight junction (TJ) proteins, suggesting that MCP-1 may be involved in BBB permeability [185]. Chemokines may also play a role in cell based therapies for stroke and related conditions. For example, they appear to be involved in honing stem cells to regions of injury [186] [188] [189] [190], and MCP-1 and SDF-1 and their corresponding receptors have been documented in the vicinity of ischemic brain tissue and transplanted cells [187]. Optimization of these signals may improve the successful application of such therapies.
Arachidonic acid metabolites
Following immune cell activation, the arachidonic acid (AA) cascade is initiated as a result of the release of phospholipase A2 (PLA2) [127]. Upstream of PLA2 release is the increased intracellular calcium accumulation due to energy failure and loss of ion concentration gradients. High intracellular calcium activates PLA2, which in turn hydrolyses glycerophospholipids to release AA. Increased PLA2 activity has been documented in experimental stroke models [191]. AA metabolites are signialing molecules which contribute to many post ischemic immune responses [192]. In line with a detrimental role in brain ischemia, PLA2 deficient mice had smaller infarcts and more favorable neurological outcome than wild type controls [193].
AA is further metabolized through the cyclooxygenase (COX) and lipoxygenase (LOX) pathways. Once released from brain phospholipids, AA is converted to prostaglandin H2 (PGH2) by COX, of which there are two isoforms. COX-1 is constitutively expressed, whereas COX-2 is inducible. In brain ischemia, COX-1 has been described in many cells types, including microglia and leukocytes [194]. COX-1 deficient mice have worsened outcome against brain ischemia, consistent a protective role possibly through a favorable effect on cerebral blood flow [195]. However, in a model of global cerebral ischemia, pharmacologic inhibition of COX-1 enhanced hippocampal neuron survival suggesting a damaing role [196]. The reasons for these differences are unclear, but could point to slight differences between focal and global cerebral ischemia.
COX-2, the inducible COX isoform, is essential for prostanoid synthesis. It is upregulated within ischemic borderzone areas in focal cerebral ischemia models [197]. Autopsy specimens from stroke patients have also documented the presence of COX-2 in ischemic brain regions [198] [199]. There are many functions of COX and its metabolites, but the collective literature suggests that most of these molecules are deleterious in stroke. Several studies have now shown that COX-2 inhibition improves neurological outcome in brain ischemia [197] [201]. In addition, COX-2 deficient mice are protected from injury due to N-methyl-D-aspartate (NMDA) exposure [202], whereas COX-2 over expression worsens brain injury [203]. Further, COX-2 appears to act through PGE2 rather than ROS, even though COX-2 generates both [204].
Compared to the COX pathway, less is known about the LOX pathway in brain ischemia. AA is converted to 5-hydroperoxyeicosatetraenoic acid (5-HPETE) by 5-lipoxygenase (5-LOX). 5-LOX is then metabolized to leukotriene A4 (LTA4), a precursor of cysteinyl leukotriene (cysLTs). LTA4 acts as a chemoattractant implicated in BBB dysfunction and neuronal death in ischemia. Like cytokines and other immune moleculars, biphasic AA and LTC4 expression patterns have been documented and also seem to correlate to the biphasic patterns of BBB opening [205]. 5-LOX has also been observed in post mortem ischemic human brains, typically localizing to perivascular monocytes [206]. In a brain ischemia model, treatment with AA861, a 5-LOX inhibitor, led to decreases in LTC4 levels and amelioration of ischemic brain injury [207]. In a model of in vitro ischemia (OGD), the 5-LOX inhibitor caffeic acid attenuated PC12 cell death [208]. However, the role of LOX in brain ischemia is not entirely clear since no protection in 5-LOX deficient mice could be observed in various experimental stroke models [209]. There are no obvious explanations for these conflicting observations, but more work in this area is clearly needed.
Nitric oxide/nitric oxide synthase
Oxidative stress can damage the organism if the physiological balance between oxidants and anti-oxidants is disrupted in favor of the former. Nitric oxide (NO) has been implicated in a variety of functions following brain ischemia. It has been documented to be involved in neuronal synapses, host defense, regulation of vascular tone, and as an inhibitor of platelet aggregation and leukocyte adhesion. Nitric oxide is generated from L-arginine through nitric oxide synthases (NOS). To date, three NOS have been studied in brain injury models. Endothelial NOS (eNOS, NOS-3), neuronal NOS (nNOS, NOS-1), and inducible NOS (iNOS, NOS-2). Of these isoforms, iNOS is perhaps the most relevant to inflammation. iNOS expression is limited almost exclusively to immune cells such as leukocytes and microglia, but has been observed in astrocytes as well [210] [211] [212]. In addition to its signaling properties, NO may also react with superoxide to form peroxynitrite, and even more reactive specia that may cause DNA damage [213] [30]. Several studies have now shown that iNOS inhibitors are neuroprotective [211], and iNOS deficient mice have better outcomes from stroke [214].
Furthermore, therapeutic hypothermia and neuroprotection by estrogen and progesterone is associated with reduced iNOS generation, indicating that NO/iNOS play a damaging role [215][216] [217].
Reactive oxygen species
Reactive oxygen species (ROS) production by inflammatory cells occurs via several enzyme systems. Superoxide is generated via COX, xanthine dehydrogenase, xanthine oxidase and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. Hypochlorous acid and H2O2 are generated through myeloperoxidase (MPO) and monoamine oxidase (MAO) [218]. ROS are an important underlying factor in delayed neuronal death induced by cerebral ischemia–reperfusion. During reperfusion, robust oxidants are generated and are directly involved in the damage to cellular macromolecules, such as lipids, proteins, and nucleic acids, eventually leading to cell death [219].
Superoxide can be produced in phagosomes, which contain ingested bacteria and fungi, or it can be produced outside of the cell. In a phagosome, superoxide can spontaneously form hydrogen peroxide that will undergo further reactions to generate ROS. Vascular ROS are produced in endothelial, adventitial, and vascular smooth muscle cells and derived primarily from NADPH oxidase (NOX), a multisubunit enzyme catalyzing a O2•–production by the 1 electron reduction of oxygen using NADPH as the electron donor: 2O2 + NADPH → 2O2− + NADP + H+ [220]. NOX was originally identified in immune cells as playing an important microbicidal role. NOX consists of cytoplasmic subunits (p45phox, p67phox, and p40phox and Rac2) and upon phosphorylation, these subunits can form a complex and translocate to the plasma membrane to dock with the plasma membrane subunits (p91phox, p22phox) [221]. Catalysis of NOX occurs in the p91phox subunit (Nox2) and is initiated by transferring of electrons from molecular oxygen through redox coupling with NADPH, FAD and heme to produce superoxide anion [222] (Fig. 1).
Figure 1.
The structure of the active NADPH oxidase complex in stroke. The NADPH oxidase comprises a cytosolic subunits (p47phox, p67phox, p40phox and Rac) and membrane subunits (gp91phox and p22phox) which associate with this complex in the activated enzyme. The NADPH-binding domain is predicted to be on one side of the membrane, whereas O2•–generation is predicted to occur on the other in stroke.
Immune cell generated NOX (NOX2) also appears important in the maintenance of vascular integrity. The addition of microglia to endothelial cell and astrocyte cocultures worsens ischemia-like injury, and inhibiting superoxide production preserved these BBB constituents in an in vitro model [223], and reduced brain edema formation, matrix metalloproteinase-9 (MMP-9) expression [224], BBB disruption, hemorrhagic transformation [225] and immune cell responses in in vivo stroke models [226]. Thus, NOX contributes to BBB disruption.
NOX is also expressed in the central nervous system. In vitro studies have shown NOX expression in neurons, astrocytes, and in microglia [222]. Immunohistochemical studies have shown that NOX subunits are widely distributed in the cortex, the hippocampus, and in the cerebellum in vivo [227] [228] [229] [230]. NOX has been documented to increase in the brain after experimental stroke [231] and we have shown that NOX derived from circulating cells contributes significantly to stroke pathogenesis compare to the brain resident cells [232]. Walder el al. [233] showed that NOX2 deficient mice are protected from experimental stroke, and work from our lab has shown that microglia derived NOX2 leads to BBB damage [223]. Further, NOX appears to significantly contribute to reperfusion injury, as reperfusion permits the restoration of glucose to the ischemic brain. The restoration of glucose (rather than oxygen, which is traditionally thought to be a source of ROS in this setting) appears to ‘fuel’ NOX by serving as an electron donor to produce damaging levels of superoxide [234]. Interestingly, reperfusion in the presence of glucose appears to increase neuronal NOX activity and NOX deficiency or inhibition prevents this. NOX also appears to be a primary source of ROS generated by NMDA receptor activation [235].
A few studies have examined the therapeutic potential of treatment with the NOX inhibitor apocynin. Apocynin, the active compound found in Picrorhiza kurroa, a botanical plant used as an herbal medicine for treatment of a number of inflammatory diseases. From our own lab, we found that a dose of 2.5 mg/kg given parenterally just prior to reperfusion, or 1.5 h after ischemia onset, resulted in reduced infarct volume and improved neurological outcome [225]. We also found that O2•–is largely generated in neurons and some microglia/monocytes, with no generation in brain vascular endothelial cells. Apocynin markedly reduced O2•–in the brain. However, apocynin at higher doses (3.75 and 5 mg/kg) failed to show any benefit, and actually increased the severity of brain hemorrhage. Thus, this rather narrow therapeutic dose range may limit its translation to the clinical level. However, other groups have shown salutary effects of apocynin at doses as high as 50 mg/kg [236] [237]. In global cerebral ischemia, 5 mg/kg apocynin attenuated hippocampal injury when given prior to ischemia onset [238].
Myeloperoxidase (MPO), present in leukocytes including neutrophils and monocytes, mediates bactericidal killing through H2O2 and hypochlorous acid. MPO has been documented within infiltrating neutrophils following both permanent and transient MCAO [181]. However, MPO deficient mice had worsened outcome following experimental stroke, suggesting a protective role rather than a damaging one [239]. There were also more products of nitrosylation within the ischemic brain of MPO deficient mice, suggesting that MPO’s protective effect may be due to its ability to scavenge nitrotyrosine (a by product of peroxynitrite reactions) [239]. Thus, MPO may decrease ROS induced ischemic injury, rather than potentiate it.
Matrix metalloproteinases
Matrix metalloproteinases (MMPs) are a family of essential proteases that break down components of extracellular matrix. Physiologically, these proteases participate in tissue development, wound healing, cone growth, ovulation and angiogenesis, however, after brain ischemia, MMPs are upregulated, activated, and involved in both neuroinflammatory response and extracellular matrix remodeling. Under physiologic conditions, many MMPs exist as a pro- or inactivated protein, but undergo activation after cleavage by other proteases such as plasmin or other MMPs [240]. Tissue plasminogen activator (tPA) has been shown to disrupt the BBB due to MMP-9 resulting in hemorrhagic transformation [241] [242]. Microglia are a major source of the MMPs in brain ischemia. The y also stimulate astrocytes to generate active MMPs [243]. In models of focal ischemia, MMP-9 activity increases at early time points (15–48 h) and returns to baseline around 15 days, followed by MMP-2 (peaks at 5 days and return to baseline around 15 days) [244] [245]. MMPs are also involved in receptor mediated (extrinsic pathway) apoptotic neuronal cell death by processing TNF-α and FasL. In stroke models, MMP inhibition is beneficial and not only decreases infarct size, but ameliorates brain edema and hemorrhage a well [246]. Similarly, mice lacking MMP-9 had smaller infarcts compared to wildtype mice [247]. However, no such effect was observed in MMP-2 deficient mice [248], suggesting that MMP-9 may be primarily involved with edema, while MMP-2 may correlate more with neovascularization [249]. Further, bone marrow chimera models showed that circulating immune cells, rather than brain derived MMP-9 may contribute significantly to ischemic brain injury. Mice transplanted with MMP-8 deficient bone marrow suffered less injury and BBB disruption than mice transplanted with marrow containing intact MMP-9 [250]. MMPs may be involved in neuron migration. Animals given the broad spectrum MMP inhibitor GM6001 were found to have less migration of newly formed neurons in the migratory stream after transient focal cerebral ischemia in mice [251].
MMPs also seem to play a differential roles depending on the phase of ischemic injury. During the later phases, they appear to participate in plasticity and recovery. MMPs were found to associate with factors involved in angiogenesis, including vascular endothelial growth factor (VEGF). Treatment with the MMP inhibitor FN-439 suppressed neurovascular remodeling in a stroke model, and impaired functional recovery while reducing VEGF signaling [252].
Transcriptional regulation of inflammation
Cerebral ischemia is well known to upregulate gene expression. Transcription factor activation has been studied in several brain ischemia models, and some of these factors are involved in the inflammatory response (Fig. 2).
Figure 2.
Several transcription factors have been documented in the inflammatory response, Ischemic injury activates IKK and MAPK cascade to stimulate IkB, SAPK/JNK, p38 MAPKs and ERK phosphorylation, leading to transcription factors; NF-kB (subunit p50 and p65) and AP-1 (phosphorylation of c-Jun and upregulation of c-Fos) activation and then enhances pro-inflammatory genes expression.
Nuclear factor κB (NF-κ B)
NF-κB is a classic transcription factor involved in the activation of inflammatory responses [253]. It is a heteromeric transcription factor consisting of various combinations of subunits from the Rel family: Rel (cRel), RelA (p65), RelB, NF-κB1(p50 and its precursor p105) and NF-κB. The most common form is composed of Rel A (p65) and p50. NF-κB is normally sequestered in the cytoplasm bound to its inhibitor protein, IκB. Phosphorylation of IκB by IκB kinase (IKK) leads to IκB phosphorylation, ubiquitination and degradation. This liberates NF-κB to translocate to the nucleus, and bind to concensus κB sites, promoter domains present in many pro-inflammatory genes, such as tumor necrosis factor-α (TNF-α), intercellular adhesion molecule-1 (ICAM-1), cyclooxygenase-2 (COX-2), iNOS and interleukin-6 (IL-6). Therapeutic hypothermia in a stroke model was correlated to IKK and NF-κB inactivation [13]. However, NF-κB also regulates genes involved in neuroprotection, and NF-κB’s role in stroke is controversial [254]. Mice deficient in the p50 subunit of NF-κB or treated with a NF-κB inhibitor are protected from brain ischemia [255][33] [257]. Similarly, inhibition of IKK reduced infarct size [256], and activation of IKK enlarged the infarct size [256]. However, NF-κB inhibition with diethyldithiocarbamate (DDTC) increased infarct size, suggesting a beneficial role [258]. The reasons for these differences are not entirely clear, but have been postulated to be dependent on the cell type in which NF-κB is activated. It is also possible that these differences are due to the experimental model studied, or off target effects of the pharmacological inhibitors.
Mitogen-activated protein kinase (MAPK)
MAPKs are known to transduce stress-related signals through a cascade of interlinked signaling pathways which lead to inflammatory induction [3] [259]. In brain ischemia models, the stress-activated protein kinases/c-Jun N-terminal kinases (SAPK/JNK), the p38 MAPKs and extracellular signal-regulated kinases (ERKs) have been described [259] [260] [261]. p38 MAPK is involved in the stabilization and translation of several proinflammatory mRNAs [262], and activation of this pathway was observed to occur 30 min and 3 days following brain ischemia [263]. Phosphorylated p38 MAPK was detected in neurons [261] and microglia [265] of ischemic brain tissue, and suggests its role in the inflammatory response. Interruption of this pathway at its apex by knockdown of POSH (Plenty of SH3s) attenuated downstream signaling and significantly increased neuron survival. In cultured astrocytes exposed to in vitro ischemia, the addition of bone marrow stromal cells activated MAPK, and subsequently led to cytoprotection [264]. Treatment with CDP-choline, a neuronal membrane lipid precursor, led to improved recovery after ischemic stroke with reduction in phosphorylation of MAPK family members, ERK1/2 and MEK1/2, and Elk-1 transcription factor. Similar studies of p38 MAPK inhibitors have shown reduction in brain injury and improvement in neurological deficits against focal cerebral ischemia along with reduction in ischemic-induced cytokine production [3].
Initiation of innate immune responses
The initiation of the immune response following stroke is still not fully clear, but recent studies have focused on various pro-inflammatory factors elaborated by the ischemic brain that might act on receptors involved in innate immune responses. The two main groups of innate immune receptors studied in brain ischemia are found on microglia and circulating immune cells, and include the Toll-like receptors (TLRs) and purinergic receptors. The ischemic brain is thought to generate extracellular nucleic acid following cell lysis. These and other ligands are often referred to as danger associated molecular pattern molecules (DAMPs). When bound to their respective ligands, an inflammasome is formed consisting of nucleic acids such ATP, UTP, adenosine and other pro-inflammatory molecules such as caspase 1, leading to the maturation and elaboration of pro-inflammatory cytokines and a full blown inflammatory response [14] [266]. Toll-like receptors (TRLs) and purinergic receptors are widely expressed on microglia. [14] [267].
Toll-like receptors (TLRs) are a large of pattern recognition receptors that recognize exogenous pathogen-associated molecular patterns (PAMPs) and endogenous DAMPs. They have been the focus of recent investigation, and are considered to play critical roles in the initiation of the immune response in stroke and related injuries [268] [269]. TLRs have traditionally been found on immune cells, but they have also been described in various cell types of the central nervous system (CNS), including microglia, astrocytes, neurons, and cerebral vascular cells [270] [271]. Reports in the brain ischemia literature indicate that TLRs are most likely activated by DAMPs, such as heat shock proteins, high mobility group box 1 protein (HMGB1) [272], extracellular peroxiredoxin [273] and nucleic acids [14].
Activation of microglial cells in response to cerebral ischemia is associated with signaling through several TLRs, especially TLR2 (TNF-α, IL-6, IL-10), TLR3 (TNF-α, IL-6, IL-10, IL-12, CXCL-10, IFN-β), and TLR4 (TNF-α, IL-6, IL-10,CXCL-10, IFN-β), yet astrocytes initiate only minor IL-6 responses to all but TLR3 stimulation [274]. The TLRs signal through intracellular pathways leading to transcription factor and activation and the generation of cytokines and chemokines [275].
Among TLR family, TLR2 and 4 has been shown to a key player in cerebral ischemic damage. Brain TLR2 expression is increased in transient and permanent focal ischemia models as well as in vitro ischemia models [276] [277] [278]. TLR2-deficient mice have less CNS injury compared with controls in a model of focal cerebral ischemia [237]. TLR2 was expressed mainly in microglia in post-ischemic brain tissue, but also in selected endothelial cells, neurons, and astrocytes; TLR2-related genes with pro-inflammatory and pro-apoptotic, such as NF-κB, Cyclooxygenase-2 (COX2), IL-1β, IL-17, IL-23, were also induced after ischemia [276] [278] [279] [280]. In a model of transient focal cerebral ischemia, infarct volume in TLR2-deficient mice was significantly smaller compared to wild-type mice. Therefore, TLR2 upregulation and signaling are important events in focal cerebral ischemia and contribute to neuronal damage [276]. A recent study demonstrated that inflammatory signaling of the TLR2 heterodimer TLR2/1 in the post-ischemic brain requires the scavenger receptor CD36 [281]. The link between CD36 and TLR2/1 was specific for brain inflammation because CD36 is required for TLR2/6 (another TLR2 heterodimer) signaling. Another study demonstrated that TLR2 mediates leukocyte and microglial infiltration and neuronal death, which can be attenuated by TLR2 inhibition [282]. TLR 2 knockout mice also experienced higher mortality and increased infarct size compared to wildtype mice [283].
Brain expression of TLR4 in ischemic brain is also up regulated [284] [285] and plays a crucial role in the innate immunity of the CNS [286]. Numerous studies demonstrate that TLR4 participates in ischemic injury. Several studies confirm that cerebral ischemia results in the upregulation of TLR4 mRNA in neurons as early as one hour after initiation of cerebral ischemia model [237] [287]. TLR4-deficient mice exhibit reduced infarct size compared with wild-type mice after cerebral ischemic injury [288] [289] [285] [290]. Following MCAO and forebrain ischemia, these mice exhibited improved neurological behavior and reduced edema, as well as reduced secretion of proinflammatory cytokines such as TNF-α and IL-6. In addition, TLR4 knock-out mice have reduced expression of inducible nitric oxide synthase (iNOS), cyclooxygenase 2 (COX2), and IFN-γ[285] [289]. Moreover, after MCAO, loss of TLR4 function is associated with reduced expression of p38 and Erk1/2 in damaged neurons, implicating TLR4 in MCAO injury. Taken together, these studies indicate that TLR4 signaling contributes to the severity of ischemia-induced neuronal damage. Thus, targeting TLR signaling may be a novel therapeutic strategy of inflammatory for cerebral ischemic injury.
The purinergic receptors are found in numerous cell types, both in brain and peripherally, and mediate a variety of cellular functions. The P2 purinoreceptors consist of two families: the ionotropic receptors (P2X) contain channels that permit ion flow, whereas the metabotropic receptors (P2Y) are G-protein coupled second messenger systems. In immune cells, they are involved in pro-inflammatory responses, migration, and phagocytosis [291]. The family of purinergic receptors on microglia have recently become of interest because they bind to nucleotides that may be released by injured cells, and may initiate proinflammatory signaling. The role of the purinergic receptors in the microglial inflammatory response has largely focused on P2X7, where it has been shown to modulate microglial activation following experimental brain ischemia and stroke, and its pharmacologic blockade led to decreased ischemic damage [292] [293] [294] [295]. However, little has been studied on the Gi coupled ATP receptor, P2Y12 [296] [291]. P2Y12 is present on microglia and is expressed on the membrane surface in the resting state and activated by ATP, ADP, or neighboring neurotoxicity. It promotes microglial migration toward the source of these nucleotides and involves in the phosphorylation of Akt [297]. Because P2Y12 is also the target of a widely used antiplatelet agent, clopidogrel, it is an attractive target for modulating the microglial inflammatory cascade. We have recently shown that P2Y12 participates in ischemia related inflammation by mediating microglial migration and potentiation of neurotoxicity using P2Y12 knockout mice [298].
Inflammatory responses to the other organs following ischemic stroke
Recent studies indicate that inflammatory responses following stroke affect the entire body, and not simply the brain. For example, splenectomy has been shown to confer neuroprotection against experimental stroke. The spleen is an important lymphatic organ, and sequesters red and white blood cells. It also synthesizes antibodies in its white pulp and removes antibody coated blood cells from blood and lymph node circulation. Ajmo et al. [299] have shown that removal of the spleen significantly reduced neurodegeneration after brain ischemia. Rats splenectomized 2 weeks before permanent middle cerebral artery occlusion had a >80% decrease in infarction volume and splenectomy also resulted in decreased numbers of activated microglia, macrophages, and neutrophils present in the brain tissue. They concluded that these results demonstrate that the peripheral immune response as mediated by the spleen is a major contributor to the inflammation that enhances ischemic damage after stroke.
Yet, evidence suggests that stroke renders the body in a state of immunodepression which could be detrimental. Soon after stroke, circulating immune cells are quickly reduced, thus increasing the risk of developing infections. This systemic immunodepression occurs as early as 12 hours after ischemic stroke, and may continue out to several weeks [12] [83] [300]. This phenomenon involves reduced numbers of T cells and other immune cells present in the spleen, thymus, liver and lymph node [300] [83] [301] [12] [149], and is considered to be mediated by hyperactivity of the sympathetic nervous system (SNS) and the hypothalamic-pituitary-adrenal axis (HPA) [12] [302]. This leads to increased apoptosis of immune cells in these organs and as a result, these secondary lymphatic organs undergo atrophy [83] [149] [12]. However, it has yet to be shown whether the infiltration of these cells into the brain is what contributes to the lower circulating cell numbers.
As a consequence of this phenomenon, infectious complications often arise after stroke, predominantly lung and urinary tract infections in animal models [83] [149] [12] which lead to worsened outcome [303] [304] [305]. Hyperactivity of the sympathetic nervous system (SNS) and the hypothalamic pituitary axis (HPA) is thought to underlie this phenomenon, although the precise signals and mechanisms that trigger this immunodepression remain unclear. Blocking the SNS and HPA significantly reversed the percentage of apoptotic splenocytes to control levels and prevented the decrease in circulating lymphocytes, bacterial infections and mortality following stroke in the animal model [302] [12]. Consistent with experimental reports, several recent clinical studies have found evidence that SNS-mediated stroke-induced immunodepression and subsequent susceptibility to post stroke infections also occurs in patients. Chamorro et al. [4] found that acute ischemic stroke is associated with an early activation of the sympathetic adrenomedullar pathway that lowers the threshold of infection and increases the mortality. Urra et al. [306] reported increased apoptosis and a reduction in the circulating levels of T and B lymphocytes following stroke. They also found a correlation between SNS and HPA activation, lower level of T lymphocytes and infection. In addition to this, another pathway of communication between the nervous and immune systems, known as the vagal cholinergic anti-inflammatory pathway has been indentified. When the vagus nerve is activated by pro-inflammatory cytokines, it releases acetylcholine, which results in inhibition of the release of more pro-inflammatory mediators by macrophages [307] [308]. Experimental studies have shown that vagal nerve signaling inhibits the release of pro-inflammatory cytokines and improves outcomes following different models of ischemic stroke [308]. This vagal cholinergic anti-inflammatory pathway is another potential mediator of immunodepression.
Clinical approaches
Following several promising preclinical studies, a few clinical trials were carried out to determine whether anti-inflammatory strategies were beneficial (Table 1). However, some of these trials did not meet with success either due to unanticipated effects of the agents tested or inappropriate study design [309] [310] [311]. Yet, with increased knowledge of the complexities of inflammation in stroke, a few treatments may be on the horizon. In order to limit the leukocyte adhesion and migration into the infarct brain, clinical studies have studied anti-integrin therapies in stroke patients. In the HALT study, a humanized antibody against the integrin CD11/CD18 was given to patients who presented within 12 h of symptom onset [310]. There was also a phase IIb study of recombinant neutrophil inhibiting factor (rNIF), a nonantibody peptide, in stroke patients (Acute Stroke Therapy by Inhibition of Neutrophils or ASTIN) who presented within 6 h of symptom onset [312]. Neither study showed any beneficial effect, as determine by study endpoints, and were terminated prematurely, although both compounds showed benefit in animal models [313] [58], The reasons for an absence of a beneficial effect in humans may multifold, stemming from incomplete preclinical testing, to the heterogeneity of clinical stroke. Another possibility is that neutrophil integrins are different in acute ischemic stroke patients compared to rodents. While CD11b is thought it increase in animal stroke models [315], it is actually decreased in human stroke [314]. Therefore, this approach would not be expected to work in humans. Nevertheless, more work in this area including improved trial design and guidelines for preclinical development are needed.
Table 1.
Agent | Representative Name | Design | Phase | n | Status | Results | ref |
---|---|---|---|---|---|---|---|
Anti-integrin | The HALT Stroke Study | Trial of Hu23F2G anti-adhesion to limit cytotoxic injury in acute ischemic stroke | III | - | Complete Aborted | Stopped early for futility: no benefit would occur if the study was completed | 301 |
ASTIN | Trial of recombinant neutrophil inhibitory factor | II | 966 | Complete Aborted | Stopped early for futility: no benefit would occur if the study was completed | 303 | |
Anti-ICAM1 | Enlimomab Acute Stroke Trial | Efficacy and safety of enlimomab versus placebo | III | 625 | Complete | Not effective, may worsen outcome. | 300 |
Minocycline | Trial of 200 mg oral minocycline to evaluate its efficacy | Open-Label | 152 | Complete | Treated patients showed better NIHSS, mRS and B.I. | 312 | |
MINOS | Dose-Finding study of minocycline | nonrandomized, dose-escalation trial | 60 | Complete | Showed safe up to dose of 10 mg/kg i.v. | 313 | |
MINOS (Sub-analysis) | Determine the impact of i.v. minocycline of MMP-9 expresion | nonrandomized, dose-escalation trial | 60 | Complete | Showed lower MMP-9 expression with the minocyline administration | 314 | |
NeuMAST | Determine the efficacy of minocyline in long term recovery | IV | - | Complete Aborted | Stopped early for futility: no benefit would occur if the study was completed | NCT00930020 |
Abbreviations; NIHSS, NIH Stroke Scale; mRS, modified Rankin Scale; B.I., Barthel Index
An antibody against ICAM-1 (enlimomab) was studied at the phase III level in stroke patients. However, it, too, was not an effective treatment for ischemic stroke [309] [311]. Not only did this approach not benefit patients, but those who received treatment had worsened stroke outcome. Reasons for this worsened outcome have been attributed to the fact that the Enlimomab antibody is a murine antibody, and possibly not suited for use in humans. The thought being that the murine antibody itself could lead to unwanted neutrophil and complement activation. It also could be said of these trials that the therapies interfered with endogenous immunoregulatory defenses to promote the development of clinically significant infections and fever, thereby negating any potential cerebroprotective effects. This deleterious immunomodulation is not entirely unexpected, because ICAM-1 and integrin are critical to numerous host defenses such as leukocyte adhesion, diapedesis, oxidative burst, and selectin expression [21]. Second, proinflammatory microvascular failure leading to “no-reflow” might be important in rodent stroke, yet of limited relevance to primate stroke, due to differences in cerebrovascular collateralization [316].
Therapeutic Hypothermia
The role of the hypothermia in brain ischemia is well described in our recent review [317]. It has largely been embraced at the clinical level, as it has been shown to improve neurological outcome following cardiac arrest. Because therapeutic hypothermia affects pathways leading to excitotoxicity [318], apoptosis [319], inflammation and free radical production, as well as blood flow, intracranial pressure [320], metabolism [320] and blood-brain barrier integrity in acute, subacute and chronic stages of ischemia, it is likely that no single factor can explain the neuroprotection provided by hypothermia, however, from the concept of this review, here we will discuss the effect of hypothermia from the inflammation aspect.
Hypothermia affects many aspects of immune response. It lowers numbers of neutrophils and activated microglia in the ischemic area [321] [322] and reduces levels of many inflammatory mediators including ROS [323] and reactive nitrogen species [215], adhesion molecules [321] [322], proimflammatory cytokines (such as IL-1), TNF-α, and IL-6, IL-10) [324] and the Chemokine such as macrophage inflammatory protein-3α (MIP3) and its only receptor C-C chemokine receptor 6 (CCR6) [180]. Hypothermia also suppresses the activation of NF-κB [13] [325], one of the most important transcription factors playing a pivotal role in activating many inflammation-related genes. However, NF-κB also regulates genes involved in cell survival and growth; thus, the net effect of hypothermia-induced suppression of NF-κB activity is difficult to predict. Hypothermia also affects the mitogen-activated protein kinase (MAPK) pathway, another important enzyme system that regulates inflammation [326] [327].
However, anti-inflammatory cytokines such as IL-10 and TGF-β are also reduced by hypothermia as well, indicating that hypothermia does not have a purely anti-inflammatory effect [328] [329]. Regardless, hypothermia has a largely suppressive effect on inflammation, and this anti-inflammatory property might serve as a major protective mechanism under ischemic conditions.
Minocycline
Minocycline is a member of the tetracycline family of antibiotics with recently recognized anti-apoptotic, anti-inflammatory properties, reduction of microglial activation, MMP reduction, and NO production. The anti-apoptotic properties of Minocycline appear to be due to tis ability to inhibit caspase-3 [330], whereas its anti-inflammatory effect appears to be due to a mechanism inhibiting MAPK activation in microglia [331]. As such, Minocycline has been shown to protect the brain against ischemic insults and improve functional impairment [332] [109] [333] [334]. According to these results, Lampl et al. [335] have conducted a clinical trial using oral Minocycline administration to the ischemic patients and found that both NIHSS and mRS were significantly lower and BI scores were significantly higher in minocycline-treated patients. This pattern was already apparent on day 7 and day 30 of follow-up. Deaths, myocardial infarctions, recurrent strokes, and hemorrhagic transformations during follow-up did not differ by treatment group. However, this study was an open-labeled, evaluator-blinded study and the total number of the patients were relatively small. A second study established safety, dose ranging and feasibility in combination with rt-PA [336]. Clinical studies to determine minocycline’s efficacy in stroke and long term recovery (Neuroprotection with minocycline therapy for acute stroke recovery trial, NeuMAST), but was recently terminated due to futility. However, large prospective randomized trials are still needed.
Fingolimod (FTY 720)
FTY720 is a novel immunomodulatory agent, which in its phosphorylated form acts as a high affinity agonist of Sphingosine-1-phosphate (S1P) receptors [337] [338]. It became the first oral drug to be FDA-approved for clinical use in the treatment of multiple sclerosis. FTY720 readily crosses the blood-brain barrier and exerts a number of direct effects in the central nervous system. FTY720 is phosphorylated by Sphingosine kinase (SphK), mainly by SphK2 [339] [340], into the active compound phospho-FTY720, which then acts on 4 of the 5 known S1P receptor subtypes (S1P1, S1P3, S1P4, S1P5), and shows neuroprotective effect against many central nervous system disease including cerebral ischemia [341] [342] [343] [344]. Mechanisms include regulation of myelination and microglial activation following injury, proliferation and migration of neural precursor cells toward injury sites, and potentiation of growth-factor regulated neuronal differentiation, survival, and process extension, and also antiapoptotic and anti-inflammatory pathways [344] [345] [346] [347] [348] [349] [343]. FTY720 also exerts immunomodulatory actions by affecting lymphocyte production, trafficking, and apoptosis through S1P receptors which induces a depletion of circulating lymphocytes by preventing the egress of lymphocytes from the lymph nodes. Mechanistically, this is due to a down regulation of the S1P type 1 receptor (S1P1). Expression levels of endothelial adhesion molecules such as E-selectin, P-selectin, ICAM-1 or VCAM-1 were shown to be induced by FTY720 treatment, and therefore might contribute to the prevention of early infiltration of neurotrophils and activation of microglia/macrophages. These findings suggest that anti-inflammatory mechanisms, and possibly vasculoprotection, rather than direct effects on neurons, underlie the beneficial effects of fingolimod after stroke. Most of the past reports have shown beneficial effect of S1P in the field of ischemia, but by contrast, Liesz et al. [48] showed opposite results. These authors found that S1P treatment did show a reduction of lymphocyte brain invasion but could not achieve a significant reduction of infarct volumes and behavior dysfunction [48]. Liu et al. [350] recently published a systematic meta-analysis of the efficacy of FTY720 in animal model of stroke. In this study, they concluded that FTY720 reduced infarct volume and improve functional outcome. However, the authors also indicated that more experimental studies should be performed to evaluate the safety of FTY720 in the future. Thus, taken this recent scientific highlights together, it is obvious that S1P receptor pathways and sphingolipids regulating enzymes are a highly promising target in stroke treatment.
Conclusions
Inflammation following ischemic stroke is increasingly recognized as a key element in its progression. The role of inflammation in stroke has become an increasingly popular area of investigation, given its pleotropic roles in both acute damage and long term recovery. In the early phases, accumulated research indicates that it appears to play a mostly detrimental role. However, inflammatory processes are also important to recovery and repair, processes that occur weeks to months later. Thus, immune modulating interventions should be tailored to the specific phase of stroke, as most certainly, inhibiting the same processes at the wrong time could prove damaging. However, what these time windows are, and how to best intervene are largely unknown, but areas ripe for future investigation.
Acknowledgments
This work was supported by grants from the National Institutes of Health (NS40516, to MY), the Veteran’s Merit Award (MY), the American Heart Association (to JYK), and the Uehara Foundation (2013 Research Fellowship, to MK). Grants to MY and JYK were administered by the Northern California Institute for Research and Education, and supported by resources of the Veterans Affairs Medical Center, San Francisco, California.
References
- 1.Durukan A, Tatlisumak T. Acute ischemic stroke: overview of major experimental rodent models, pathophysiology, and therapy of focal cerebral ischemia. Pharmacol Biochem Behav. 2007;87(1):179–197. doi: 10.1016/j.pbb.2007.04.015. [DOI] [PubMed] [Google Scholar]
- 2.NINDS, Tissue plasminogen activator for acute ischemic stroke. The National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group. N Engl J Med. 1995;333(24):1581–1587. doi: 10.1056/NEJM199512143332401. [DOI] [PubMed] [Google Scholar]
- 3.Barone FC, Feuerstein GZ. Inflammatory mediators and stroke: new opportunities for novel therapeutics. J Cereb Blood Flow Metab. 1999;19(8):819–834. doi: 10.1097/00004647-199908000-00001. [DOI] [PubMed] [Google Scholar]
- 4.Chamorro A, Hallenbeck J. The harms and benefits of inflammatory and immune responses in vascular disease. Stroke. 2006;37(2):291–293. doi: 10.1161/01.STR.0000200561.69611.f8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Davies CA, Loddick SA, Stroemer RP, Hunt J, Rothwell NJ. An integrated analysis of the progression of cell responses induced by permanent focal middle cerebral artery occlusion in the rat. Exp Neurol. 1998;154(1):199–212. doi: 10.1006/exnr.1998.6891. [DOI] [PubMed] [Google Scholar]
- 6.Becker KJ. Inflammation and acute stroke. Curr Opin Neurol. 1998;11(1):45–49. doi: 10.1097/00019052-199802000-00008. [DOI] [PubMed] [Google Scholar]
- 7.Stanimirovic DB, Wong J, Shapiro A, Durkin JP. Increase in surface expression of ICAM-1, VCAM-1 and E-selectin in human cerebromicrovascular endothelial cells subjected to ischemia-like insults. Acta Neurochir Suppl. 1997;70:12–16. doi: 10.1007/978-3-7091-6837-0_4. [DOI] [PubMed] [Google Scholar]
- 8.Morioka T, Kalehua AN, Streit WJ. Characterization of microglial reaction after middle cerebral artery occlusion in rat brain. J Comp Neurol. 1993;327(1):123–132. doi: 10.1002/cne.903270110. [DOI] [PubMed] [Google Scholar]
- 9.Suzuki H, Abe K, Tojo S, Morooka S, Kimura K, Mizugaki M, Itoyama Y. Postischemic expression of P-selectin immunoreactivity in rat brain. Neurosci Lett. 1997;228(3):151–154. doi: 10.1016/s0304-3940(97)00385-6. [DOI] [PubMed] [Google Scholar]
- 10.Hallenbeck JM. Significance of the inflammatory response in brain ischemia. Acta Neurochir Suppl. 1996;66:27–31. doi: 10.1007/978-3-7091-9465-2_5. [DOI] [PubMed] [Google Scholar]
- 11.Danton GH, Dietrich WD. Inflammatory mechanisms after ischemia and stroke. J Neuropathol Exp Neurol. 2003;62(2):127–136. doi: 10.1093/jnen/62.2.127. [DOI] [PubMed] [Google Scholar]
- 12.Prass K, Meisel C, Hoflich C, Braun J, Halle E, Wolf T, Ruscher K, Victorov IV, Priller J, Dirnagl U, Volk HD, Meisel A. Stroke-induced immunodeficiency promotes spontaneous bacterial infections and is mediated by sympathetic activation reversal by poststroke T helper cell type 1-like immunostimulation. J Exp Med. 2003;198(5):725–736. doi: 10.1084/jem.20021098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Han HS, Yenari MA. Cellular targets of brain inflammation in stroke. Curr Opin Investig Drugs. 2003;4(5):522–529. [PubMed] [Google Scholar]
- 14.Iadecola C, Anrather J. The immunology of stroke: from mechanisms to translation. Nat Med. 2011;17(7):796–808. doi: 10.1038/nm.2399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Spite M, Serhan CN. Novel lipid mediators promote resolution of acute inflammation: impact of aspirin and statins. Circ Res. 2010;107(10):1170–1184. doi: 10.1161/CIRCRESAHA.110.223883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Schilling M, Besselmann M, Muller M, Strecker JK, Ringelstein EB, Kiefer R. Predominant phagocytic activity of resident microglia over hematogenous macrophages following transient focal cerebral ischemia: an investigation using green fluorescent protein transgenic bone marrow chimeric mice. Exp Neurol. 2005;196(2):290–297. doi: 10.1016/j.expneurol.2005.08.004. [DOI] [PubMed] [Google Scholar]
- 17.Denes A, Vidyasagar R, Feng J, Narvainen J, McColl BW, Kauppinen RA, Allan SM. Proliferating resident microglia after focal cerebral ischaemia in mice. J Cereb Blood Flow Metab. 2007;27(12):1941–1953. doi: 10.1038/sj.jcbfm.9600495. [DOI] [PubMed] [Google Scholar]
- 18.Ishikawa M, Zhang JH, Nanda A, Granger DN. Inflammatory responses to ischemia and reperfusion in the cerebral microcirculation. Front Biosci. 2004;9:1339–1347. doi: 10.2741/1330. [DOI] [PubMed] [Google Scholar]
- 19.Sughrue ME, Mehra A, Connolly ES, Jr, D’Ambrosio AL. Anti-adhesion molecule strategies as potential neuroprotective agents in cerebral ischemia: a critical review of the literature. Inflamm Res. 2004;53(10):497–508. doi: 10.1007/s00011-004-1282-0. [DOI] [PubMed] [Google Scholar]
- 20.Huang J, Upadhyay UM, Tamargo RJ. Inflammation in stroke and focal cerebral ischemia. Surg Neurol. 2006;66(3):232–245. doi: 10.1016/j.surneu.2005.12.028. [DOI] [PubMed] [Google Scholar]
- 21.DeGraba TJ. The role of inflammation after acute stroke: utility of pursuing anti-adhesion molecule therapy. Neurology. 1998;51(3 Suppl 3):S62–68. doi: 10.1212/wnl.51.3_suppl_3.s62. [DOI] [PubMed] [Google Scholar]
- 22.Emsley HC, Tyrrell PJ. Inflammation and infection in clinical stroke. J Cereb Blood Flow Metab. 2002;22(12):1399–1419. doi: 10.1097/01.WCB.0000037880.62590.28. [DOI] [PubMed] [Google Scholar]
- 23.Carlos TM, Harlan JM. Leukocyte-endothelial adhesion molecules. Blood. 1994;84(7):2068–2101. [PubMed] [Google Scholar]
- 24.Kim JS. Cytokines and adhesion molecules in stroke and related diseases. J Neurol Sci. 1996;137(2):69–78. doi: 10.1016/0022-510x(95)00338-3. [DOI] [PubMed] [Google Scholar]
- 25.Zhang RL, Chopp M, Zhang ZG, Phillips ML, Rosenbloom CL, Cruz R, Manning A. E-selectin in focal cerebral ischemia and reperfusion in the rat. J Cereb Blood Flow Metab. 1996;16(6):1126–1136. doi: 10.1097/00004647-199611000-00006. [DOI] [PubMed] [Google Scholar]
- 26.Haring HP, Berg EL, Tsurushita N, Tagaya M, del Zoppo GJ. E-selectin appears in nonischemic tissue during experimental focal cerebral ischemia. Stroke. 1996;27(8):1386–1391. doi: 10.1161/01.str.27.8.1386. discussion 1391–1382. [DOI] [PubMed] [Google Scholar]
- 27.Barkalow FJ, Goodman MJ, Gerritsen ME, Mayadas TN. Brain endothelium lack one of two pathways of P-selectin-mediated neutrophil adhesion. Blood. 1996;88(12):4585–4593. [PubMed] [Google Scholar]
- 28.Zhang R, Chopp M, Zhang Z, Jiang N, Powers C. The expression of P-and E-selectins in three models of middle cerebral artery occlusion. Brain Res. 1998;785(2):207–214. doi: 10.1016/s0006-8993(97)01343-7. [DOI] [PubMed] [Google Scholar]
- 29.Bargatze RF, Kurk S, Butcher EC, Jutila MA. Neutrophils roll on adherent neutrophils bound to cytokine-induced endothelial cells via L-selectin on the rolling cells. J Exp Med. 1994;180(5):1785–1792. doi: 10.1084/jem.180.5.1785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Huang J, Choudhri TF, Winfree CJ, McTaggart RA, Kiss S, Mocco J, Kim LJ, Protopsaltis TS, Zhang Y, Pinsky DJ, Connolly ES., Jr Postischemic cerebrovascular E-selectin expression mediates tissue injury in murine stroke. Stroke. 2000;31(12):3047–3053. [PubMed] [Google Scholar]
- 31.Huang J, Kim LJ, Mealey R, Marsh HC, Jr, Zhang Y, Tenner AJ, Connolly ES, Jr, Pinsky DJ. Neuronal protection in stroke by an sLex-glycosylated complement inhibitory protein. Science. 1999;285(5427):595–599. doi: 10.1126/science.285.5427.595. [DOI] [PubMed] [Google Scholar]
- 32.Hewett SJ, Muir JK, Lobner D, Symons A, Choi DW. Potentiation of oxygen-glucose deprivation-induced neuronal death after induction of iNOS. Stroke. 1996;27(9):1586–1591. doi: 10.1161/01.str.27.9.1586. [DOI] [PubMed] [Google Scholar]
- 33.Khan M, Jatana M, Elango C, Singh Paintlia A, Singh AK, Singh I. Cerebrovascular protection by various nitric oxide donors in rats after experimental stroke. Nitric Oxide. 2006 doi: 10.1016/j.niox.2006.01.008. [DOI] [PubMed] [Google Scholar]
- 34.Goussev AV, Zhang Z, Anderson DC, Chopp M. P-selectin antibody reduces hemorrhage and infarct volume resulting from MCA occlusion in the rat. J Neurol Sci. 1998;161(1):16–22. doi: 10.1016/s0022-510x(98)00262-7. [DOI] [PubMed] [Google Scholar]
- 35.Mocco J, Choudhri T, Huang J, Harfeldt E, Efros L, Klingbeil C, Vexler V, Hall W, Zhang Y, Mack W, Popilskis S, Pinsky DJ, Connolly ES., Jr HuEP5C7 as a humanized monoclonal anti-E/P-selectin neurovascular protective strategy in a blinded placebo-controlled trial of nonhuman primate stroke. Circ Res. 2002;91(10):907–914. doi: 10.1161/01.res.0000042063.15901.20. [DOI] [PubMed] [Google Scholar]
- 36.Yenari MA, Sun GH, Kunis DM, Onley D, Vexler V. L-selectin inhibition does not reduce injury in a rabbit model of transient focal cerebral ischemia. Neurol Res. 2001;23(1):72–78. doi: 10.1179/016164101101198154. [DOI] [PubMed] [Google Scholar]
- 37.Kalinowska A, Losy J. PECAM-1, a key player in neuroinflammation. Eur J Neurol. 2006;13(12):1284–1290. doi: 10.1111/j.1468-1331.2006.01640.x. [DOI] [PubMed] [Google Scholar]
- 38.Yoshimoto T, Houkin K, Tada M, Abe H. Induction of cytokines, chemokines and adhesion molecule mRNA in a rat forebrain reperfusion model. Acta Neuropathol. 1997;93(2):154–158. doi: 10.1007/s004010050596. [DOI] [PubMed] [Google Scholar]
- 39.Zhang RL, Chopp M, Zaloga C, Zhang ZG, Jiang N, Gautam SC, Tang WX, Tsang W, Anderson DC, Manning AM. The temporal profiles of ICAM-1 protein and mRNA expression after transient MCA occlusion in the rat. Brain Res. 1995;682(1–2):182–188. doi: 10.1016/0006-8993(95)00346-r. [DOI] [PubMed] [Google Scholar]
- 40.Wang X, Siren AL, Liu Y, Yue TL, Barone FC, Feuerstein GZ. Upregulation of intercellular adhesion molecule 1 (ICAM-1) on brain microvascular endothelial cells in rat ischemic cortex. Brain Res Mol Brain Res. 1994;26(1–2):61–68. doi: 10.1016/0169-328x(94)90074-4. [DOI] [PubMed] [Google Scholar]
- 41.Connolly ES, Jr, Winfree CJ, Prestigiacomo CJ, Kim SC, Choudhri TF, Hoh BL, Naka Y, Solomon RA, Pinsky DJ. Exacerbation of cerebral injury in mice that express the P-selectin gene: identification of P-selectin blockade as a new target for the treatment of stroke. Circ Res. 1997;81(3):304–310. doi: 10.1161/01.res.81.3.304. [DOI] [PubMed] [Google Scholar]
- 42.Kitagawa K, Matsumoto M, Mabuchi T, Yagita Y, Ohtsuki T, Hori M, Yanagihara T. Deficiency of intercellular adhesion molecule 1 attenuates microcirculatory disturbance and infarction size in focal cerebral ischemia. J Cereb Blood Flow Metab. 1998;18(12):1336–1345. doi: 10.1097/00004647-199812000-00008. [DOI] [PubMed] [Google Scholar]
- 43.Soriano SG, Coxon A, Wang YF, Frosch MP, Lipton SA, Hickey PR, Mayadas TN. Mice deficient in Mac-1 (CD11b/CD18) are less susceptible to cerebral ischemia/reperfusion injury. Stroke. 1999;30(1):134–139. doi: 10.1161/01.str.30.1.134. [DOI] [PubMed] [Google Scholar]
- 44.Bowes MP, Zivin JA, Rothlein R. Monoclonal antibody to the ICAM-1 adhesion site reduces neurological damage in a rabbit cerebral embolism stroke model. Exp Neurol. 1993;119(2):215–219. doi: 10.1006/exnr.1993.1023. [DOI] [PubMed] [Google Scholar]
- 45.Chopp M, Li Y, Jiang N, Zhang RL, Prostak J. Antibodies against adhesion molecules reduce apoptosis after transient middle cerebral artery occlusion in rat brain. J Cereb Blood Flow Metab. 1996;16(4):578–584. doi: 10.1097/00004647-199607000-00007. [DOI] [PubMed] [Google Scholar]
- 46.Kanemoto Y, Nakase H, Akita N, Sakaki T. Effects of anti-intercellular adhesion molecule-1 antibody on reperfusion injury induced by late reperfusion in the rat middle cerebral artery occlusion model. Neurosurgery. 2002;51(4):1034–1041. doi: 10.1097/00006123-200210000-00033. discussion 1041–1032. [DOI] [PubMed] [Google Scholar]
- 47.Vemuganti R, Dempsey RJ, Bowen KK. Inhibition of intercellular adhesion molecule-1 protein expression by antisense oligonucleotides is neuroprotective after transient middle cerebral artery occlusion in rat. Stroke. 2004;35(1):179–184. doi: 10.1161/01.STR.0000106479.53235.3E. [DOI] [PubMed] [Google Scholar]
- 48.Liesz A, Zhou W, Mracsko E, Karcher S, Bauer H, Schwarting S, Sun L, Bruder D, Stegemann S, Cerwenka A, Sommer C, Dalpke AH, Veltkamp R. Inhibition of lymphocyte trafficking shields the brain against deleterious neuroinflammation after stroke. Brain. 2011;134(Pt 3):704–720. doi: 10.1093/brain/awr008. [DOI] [PubMed] [Google Scholar]
- 49.Connolly ES, Jr, Winfree CJ, Springer TA, Naka Y, Liao H, Yan SD, Stern DM, Solomon RA, Gutierrez-Ramos JC, Pinsky DJ. Cerebral protection in homozygous null ICAM-1 mice after middle cerebral artery occlusion. Role of neutrophil adhesion in the pathogenesis of stroke. J Clin Invest. 1996;97(1):209–216. doi: 10.1172/JCI118392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Blann A, Kumar P, Krupinski J, McCollum C, Beevers DG, Lip GY. Soluble intercelluar adhesion molecule-1, E-selectin, vascular cell adhesion molecule-1 and von Willebrand factor in stroke. Blood Coagul Fibrinolysis. 1999;10(5):277–284. doi: 10.1097/00001721-199907000-00009. [DOI] [PubMed] [Google Scholar]
- 51.Justicia C, Martin A, Rojas S, Gironella M, Cervera A, Panes J, Chamorro A, Planas AM. Anti-VCAM-1 antibodies did not protect against ischemic damage either in rats or in mice. J Cereb Blood Flow Metab. 2006;26(3):421–432. doi: 10.1038/sj.jcbfm.9600198. [DOI] [PubMed] [Google Scholar]
- 52.Zhang LH, Wei EQ. Neuroprotective effect of ONO-1078, a leukotriene receptor antagonist, on transient global cerebral ischemia in rats. Acta Pharmacol Sin. 2003;24(12):1241–1247. [PubMed] [Google Scholar]
- 53.Cervera A, Justicia C, Reverter JC, Planas AM, Chamorro A. Steady plasma concentration of unfractionated heparin reduces infarct volume and prevents inflammatory damage after transient focal cerebral ischemia in the rat. J Neurosci Res. 2004;77(4):565–572. doi: 10.1002/jnr.20186. [DOI] [PubMed] [Google Scholar]
- 54.Smith CW. Leukocyte-endothelial cell interactions. Semin Hematol. 1993;30(4 Suppl 4):45–53. discussion 54–45. [PubMed] [Google Scholar]
- 55.Bowes MP, Rothlein R, Fagan SC, Zivin JA. Monoclonal antibodies preventing leukocyte activation reduce experimental neurologic injury and enhance efficacy of thrombolytic therapy. Neurology. 1995;45(4):815–819. doi: 10.1212/wnl.45.4.815. [DOI] [PubMed] [Google Scholar]
- 56.Clark WM, Lessov N, Lauten JD, Hazel K. Doxycycline treatment reduces ischemic brain damage in transient middle cerebral artery occlusion in the rat. J Mol Neurosci. 1997;9(2):103–108. doi: 10.1007/BF02736854. [DOI] [PubMed] [Google Scholar]
- 57.Prestigiacomo CJ, Kim SC, Connolly ES, Jr, Liao H, Yan SF, Pinsky DJ. CD18-mediated neutrophil recruitment contributes to the pathogenesis of reperfused but not nonreperfused stroke. Stroke. 1999;30(5):1110–1117. doi: 10.1161/01.str.30.5.1110. [DOI] [PubMed] [Google Scholar]
- 58.Yenari MA, Kunis D, Sun GH, Onley D, Watson L, Turner S, Whitaker S, Steinberg GK. Hu23F2G, an antibody recognizing the leukocyte CD11/CD18 integrin, reduces injury in a rabbit model of transient focal cerebral ischemia. Exp Neurol. 1998;153(2):223–233. doi: 10.1006/exnr.1998.6876. [DOI] [PubMed] [Google Scholar]
- 59.Zhang RL, Zhang ZG, Chopp M. Increased therapeutic efficacy with rt-PA and anti-CD18 antibody treatment of stroke in the rat. Neurology. 1999;52(2):273–279. doi: 10.1212/wnl.52.2.273. [DOI] [PubMed] [Google Scholar]
- 60.Becker K, Kindrick D, Relton J, Harlan J, Winn R. Antibody to the alpha4 integrin decreases infarct size in transient focal cerebral ischemia in rats. Stroke. 2001;32(1):206–211. doi: 10.1161/01.str.32.1.206. [DOI] [PubMed] [Google Scholar]
- 61.Relton JK, Sloan KE, Frew EM, Whalley ET, Adams SP, Lobb RR. Inhibition of alpha4 integrin protects against transient focal cerebral ischemia in normotensive and hypertensive rats. Stroke. 2001;32(1):199–205. doi: 10.1161/01.str.32.1.199. [DOI] [PubMed] [Google Scholar]
- 62.Yilmaz G, Granger DN. Cell adhesion molecules and ischemic stroke. Neurol Res. 2008;30(8):783–793. doi: 10.1179/174313208X341085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Hawkins BT, Davis TP. The blood-brain barrier/neurovascular unit in health and disease. Pharmacol Rev. 2005;57(2):173–185. doi: 10.1124/pr.57.2.4. [DOI] [PubMed] [Google Scholar]
- 64.Milner R, Hung S, Wang X, Berg GI, Spatz M, del Zoppo GJ. Responses of endothelial cell and astrocyte matrix-integrin receptors to ischemia mimic those observed in the neurovascular unit. Stroke. 2008;39(1):191–197. doi: 10.1161/STROKEAHA.107.486134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Dore-Duffy P. Pericytes: pluripotent cells of the blood brain barrier. Curr Pharm Des. 2008;14(16):1581–1593. doi: 10.2174/138161208784705469. [DOI] [PubMed] [Google Scholar]
- 66.Lapchak PA, Chapman DF, Zivin JA. Metalloproteinase inhibition reduces thrombolytic (tissue plasminogen activator)-induced hemorrhage after thromboembolic stroke. Stroke. 2000;31(12):3034–3040. doi: 10.1161/01.str.31.12.3034. [DOI] [PubMed] [Google Scholar]
- 67.Yang Y, Estrada EY, Thompson JF, Liu W, Rosenberg GA. Matrix metalloproteinase-mediated disruption of tight junction proteins in cerebral vessels is reversed by synthetic matrix metalloproteinase inhibitor in focal ischemia in rat. J Cereb Blood Flow Metab. 2007;27(4):697–709. doi: 10.1038/sj.jcbfm.9600375. [DOI] [PubMed] [Google Scholar]
- 68.Jin R, Yang G, Li G. Molecular insights and therapeutic targets for blood-brain barrier disruption in ischemic stroke: critical role of matrix metalloproteinases and tissue-type plasminogen activator. Neurobiol Dis. 2010;38(3):376–385. doi: 10.1016/j.nbd.2010.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Cinelli P, Madani R, Tsuzuki N, Vallet P, Arras M, Zhao CN, Osterwalder T, Rulicke T, Sonderegger P. Neuroserpin, a neuroprotective factor in focal ischemic stroke. Mol Cell Neurosci. 2001;18(5):443–457. doi: 10.1006/mcne.2001.1028. [DOI] [PubMed] [Google Scholar]
- 70.Yepes M, Sandkvist M, Wong MK, Coleman TA, Smith E, Cohan SL, Lawrence DA. Neuroserpin reduces cerebral infarct volume and protects neurons from ischemia-induced apoptosis. Blood. 2000;96(2):569–576. [PubMed] [Google Scholar]
- 71.Wang Q, Tang XN, Yenari MA. The inflammatory response in stroke. J Neuroimmunol. 2007;184(1–2):53–68. doi: 10.1016/j.jneuroim.2006.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Nilupul Perera M, Ma HK, Arakawa S, Howells DW, Markus R, Rowe CC, Donnan GA. Inflammation following stroke. J Clin Neurosci. 2006;13(1):1–8. doi: 10.1016/j.jocn.2005.07.005. [DOI] [PubMed] [Google Scholar]
- 73.Arvin B, Neville LF, Barone FC, Feuerstein GZ. The role of inflammation and cytokines in brain injury. Neurosci Biobehav Rev. 1996;20(3):445–452. doi: 10.1016/0149-7634(95)00026-7. [DOI] [PubMed] [Google Scholar]
- 74.Hartl R, Schurer L, Schmid-Schonbein GW, del Zoppo GJ. Experimental antileukocyte interventions in cerebral ischemia. J Cereb Blood Flow Metab. 1996;16(6):1108–1119. doi: 10.1097/00004647-199611000-00004. [DOI] [PubMed] [Google Scholar]
- 75.Li GZ, Zhong D, Yang LM, Sun B, Zhong ZH, Yin YH, Cheng J, Yan BB, Li HL. Expression of interleukin-17 in ischemic brain tissue. Scand J Immunol. 2005;62(5):481–486. doi: 10.1111/j.1365-3083.2005.01683.x. [DOI] [PubMed] [Google Scholar]
- 76.Stevens SL, Bao J, Hollis J, Lessov NS, Clark WM, Stenzel-Poore MP. The use of flow cytometry to evaluate temporal changes in inflammatory cells following focal cerebral ischemia in mice. Brain Res. 2002;932(1–2):110–119. doi: 10.1016/s0006-8993(02)02292-8. [DOI] [PubMed] [Google Scholar]
- 77.Brait VH, Rivera J, Broughton BR, Lee S, Drummond GR, Sobey CG. Chemokine-related gene expression in the brain following ischemic stroke: no role for CXCR2 in outcome. Brain Res. 2011;1372:169–179. doi: 10.1016/j.brainres.2010.11.087. [DOI] [PubMed] [Google Scholar]
- 78.Kleinschnitz C, Schwab N, Kraft P, Hagedorn I, Dreykluft A, Schwarz T, Austinat M, Nieswandt B, Wiendl H, Stoll G. Early detrimental T-cell effects in experimental cerebral ischemia are neither related to adaptive immunity nor thrombus formation. Blood. 2010;115(18):3835–3842. doi: 10.1182/blood-2009-10-249078. [DOI] [PubMed] [Google Scholar]
- 79.Hurn PD, Subramanian S, Parker SM, Afentoulis ME, Kaler LJ, Vandenbark AA, Offner H. T- and B-cell-deficient mice with experimental stroke have reduced lesion size and inflammation. J Cereb Blood Flow Metab. 2007;27(11):1798–1805. doi: 10.1038/sj.jcbfm.9600482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Shichita T, Sugiyama Y, Ooboshi H, Sugimori H, Nakagawa R, Takada I, Iwaki T, Okada Y, Iida M, Cua DJ, Iwakura Y, Yoshimura A. Pivotal role of cerebral interleukin-17-producing gammadeltaT cells in the delayed phase of ischemic brain injury. Nat Med. 2009;15(8):946–950. doi: 10.1038/nm.1999. [DOI] [PubMed] [Google Scholar]
- 81.Yilmaz G, Arumugam TV, Stokes KY, Granger DN. Role of T lymphocytes and interferon-gamma in ischemic stroke. Circulation. 2006;113(17):2105–2112. doi: 10.1161/CIRCULATIONAHA.105.593046. [DOI] [PubMed] [Google Scholar]
- 82.Saino O, Taguchi A, Nakagomi T, Nakano-Doi A, Kashiwamura S, Doe N, Nakagomi N, Soma T, Yoshikawa H, Stern DM, Okamura H, Matsuyama T. Immunodeficiency reduces neural stem/progenitor cell apoptosis and enhances neurogenesis in the cerebral cortex after stroke. J Neurosci Res. 2010;88(11):2385–2397. doi: 10.1002/jnr.22410. [DOI] [PubMed] [Google Scholar]
- 83.Liesz A, Suri-Payer E, Veltkamp C, Doerr H, Sommer C, Rivest S, Giese T, Veltkamp R. Regulatory T cells are key cerebroprotective immunomodulators in acute experimental stroke. Nat Med. 2009;15(2):192–199. doi: 10.1038/nm.1927. [DOI] [PubMed] [Google Scholar]
- 84.Ren X, Akiyoshi K, Grafe MR, Vandenbark AA, Hurn PD, Herson PS, Offner H. Myelin specific cells infiltrate MCAO lesions and exacerbate stroke severity. Metab Brain Dis. 2012;27(1):7–15. doi: 10.1007/s11011-011-9267-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Wong CH, Jenne CN, Lee WY, Leger C, Kubes P. Functional innervation of hepatic iNKT cells is immunosuppressive following stroke. Science. 2011;334(6052):101–105. doi: 10.1126/science.1210301. [DOI] [PubMed] [Google Scholar]
- 86.Nadareishvili ZG, Li H, Wright V, Maric D, Warach S, Hallenbeck JM, Dambrosia J, Barker JL, Baird AE. Elevated pro-inflammatory CD4+CD28- lymphocytes and stroke recurrence and death. Neurology. 2004;63(8):1446–1451. doi: 10.1212/01.wnl.0000142260.61443.7c. [DOI] [PubMed] [Google Scholar]
- 87.Dinkel K, Dhabhar FS, Sapolsky RM. Neurotoxic effects of polymorphonuclear granulocytes on hippocampal primary cultures. Proc Natl Acad Sci U S A. 2004;101(1):331–336. doi: 10.1073/pnas.0303510101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Harrison DG, Guzik TJ, Goronzy J, Weyand C. Is hypertension an immunologic disease? Curr Cardiol Rep. 2008;10(6):464–469. doi: 10.1007/s11886-008-0073-6. [DOI] [PubMed] [Google Scholar]
- 89.Barry M, Bleackley RC. Cytotoxic T lymphocytes: all roads lead to death. Nat Rev Immunol. 2002;2(6):401–409. doi: 10.1038/nri819. [DOI] [PubMed] [Google Scholar]
- 90.Konoeda F, Shichita T, Yoshida H, Sugiyama Y, Muto G, Hasegawa E, Morita R, Suzuki N, Yoshimura A. Therapeutic effect of IL-12/23 and their signaling pathway blockade on brain ischemia model. Biochem Biophys Res Commun. 2010;402(3):500–506. doi: 10.1016/j.bbrc.2010.10.058. [DOI] [PubMed] [Google Scholar]
- 91.Brait VH, Jackman KA, Walduck AK, Selemidis S, Diep H, Mast AE, Guida E, Broughton BR, Drummond GR, Sobey CG. Mechanisms contributing to cerebral infarct size after stroke: gender, reperfusion, T lymphocytes, and Nox2-derived superoxide. J Cereb Blood Flow Metab. 2010;30(7):1306–1317. doi: 10.1038/jcbfm.2010.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Kreutzberg GW. Microglia: a sensor for pathological events in the CNS. Trends Neurosci. 1996;19(8):312–318. doi: 10.1016/0166-2236(96)10049-7. [DOI] [PubMed] [Google Scholar]
- 93.El Khoury J, Hickman SE, Thomas CA, Loike JD, Silverstein SC. Microglia, scavenger receptors, and the pathogenesis of Alzheimer’s disease. Neurobiol Aging. 1998;19(1 Suppl):S81–84. doi: 10.1016/s0197-4580(98)00036-0. [DOI] [PubMed] [Google Scholar]
- 94.Thomas WE. Brain macrophages: evaluation of microglia and their functions. Brain Res Brain Res Rev. 1992;17(1):61–74. doi: 10.1016/0165-0173(92)90007-9. [DOI] [PubMed] [Google Scholar]
- 95.Lai AY, Todd KG. Microglia in cerebral ischemia: molecular actions and interactions. Can J Physiol Pharmacol. 2006;84(1):49–59. doi: 10.1139/Y05-143. [DOI] [PubMed] [Google Scholar]
- 96.Zhang Z, Chopp M, Powers C. Temporal profile of microglial response following transient (2 h) middle cerebral artery occlusion. Brain Res. 1997;744(2):189–198. doi: 10.1016/S0006-8993(96)01085-2. [DOI] [PubMed] [Google Scholar]
- 97.Schubert P, Morino T, Miyazaki H, Ogata T, Nakamura Y, Marchini C, Ferroni S. Cascading glia reactions: a common pathomechanism and its differentiated control by cyclic nucleotide signaling. Ann N Y Acad Sci. 2000;903:24–33. doi: 10.1111/j.1749-6632.2000.tb06346.x. [DOI] [PubMed] [Google Scholar]
- 98.Saito S, Matsuura M, Tominaga K, Kirikae T, Nakano M. Important role of membrane-associated CD14 in the induction of IFN-beta and subsequent nitric oxide production by murine macrophages in response to bacterial lipopolysaccharide. Eur J Biochem. 2000;267(1):37–45. doi: 10.1046/j.1432-1327.2000.00956.x. [DOI] [PubMed] [Google Scholar]
- 99.Beschorner R, Schluesener HJ, Gozalan F, Meyermann R, Schwab JM. Infiltrating CD14+ monocytes and expression of CD14 by activated parenchymal microglia/macrophages contribute to the pool of CD14+ cells in ischemic brain lesions. J Neuroimmunol. 2002;126(1–2):107–115. doi: 10.1016/s0165-5728(02)00046-2. [DOI] [PubMed] [Google Scholar]
- 100.Lehnardt S, Massillon L, Follett P, Jensen FE, Ratan R, Rosenberg PA, Volpe JJ, Vartanian T. Activation of innate immunity in the CNS triggers neurodegeneration through a Toll-like receptor 4-dependent pathway. Proc Natl Acad Sci U S A. 2003;100(14):8514–8519. doi: 10.1073/pnas.1432609100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Stoll G, Jander S, Schroeter M. Inflammation and glial responses in ischemic brain lesions. Prog Neurobiol. 1998;56(2):149–171. doi: 10.1016/s0301-0082(98)00034-3. [DOI] [PubMed] [Google Scholar]
- 102.Dirnagl U, Iadecola C, Moskowitz MA. Pathobiology of ischaemic stroke: an integrated view. Trends Neurosci. 1999;22(9):391–397. doi: 10.1016/s0166-2236(99)01401-0. [DOI] [PubMed] [Google Scholar]
- 103.Clausen BE, Burkhardt C, Reith W, Renkawitz R, Forster I. Conditional gene targeting in macrophages and granulocytes using LysMcre mice. Transgenic Res. 1999;8(4):265–277. doi: 10.1023/a:1008942828960. [DOI] [PubMed] [Google Scholar]
- 104.Wood PL. Microglia as a unique cellular target in the treatment of stroke: potential neurotoxic mediators produced by activated microglia. Neurol Res. 1995;17(4):242–248. doi: 10.1080/01616412.1995.11740321. [DOI] [PubMed] [Google Scholar]
- 105.Zhang N, Komine-Kobayashi M, Tanaka R, Liu M, Mizuno Y, Urabe T. Edaravone reduces early accumulation of oxidative products and sequential inflammatory responses after transient focal ischemia in mice brain. Stroke. 2005;36(10):2220–2225. doi: 10.1161/01.STR.0000182241.07096.06. [DOI] [PubMed] [Google Scholar]
- 106.Gunther A, Kuppers-Tiedt L, Schneider PM, Kunert I, Berrouschot J, Schneider D, Rossner S. Reduced infarct volume and differential effects on glial cell activation after hyperbaric oxygen treatment in rat permanent focal cerebral ischaemia. Eur J Neurosci. 2005;21(11):3189–3194. doi: 10.1111/j.1460-9568.2005.04151.x. [DOI] [PubMed] [Google Scholar]
- 107.Yu YM, Kim JB, Lee KW, Kim SY, Han PL, Lee JK. Inhibition of the cerebral ischemic injury by ethyl pyruvate with a wide therapeutic window. Stroke. 2005;36(10):2238–2243. doi: 10.1161/01.STR.0000181779.83472.35. [DOI] [PubMed] [Google Scholar]
- 108.Yrjanheikki J, Keinanen R, Pellikka M, Hokfelt T, Koistinaho J. Tetracyclines inhibit microglial activation and are neuroprotective in global brain ischemia. Proc Natl Acad Sci U S A. 1998;95(26):15769–15774. doi: 10.1073/pnas.95.26.15769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Yrjanheikki J, Tikka T, Keinanen R, Goldsteins G, Chan PH, Koistinaho J. A tetracycline derivative, minocycline, reduces inflammation and protects against focal cerebral ischemia with a wide therapeutic window. Proc Natl Acad Sci U S A. 1999;96(23):13496–13500. doi: 10.1073/pnas.96.23.13496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Giulian D, Corpuz M, Chapman S, Mansouri M, Robertson C. Reactive mononuclear phagocytes release neurotoxins after ischemic and traumatic injury to the central nervous system. J Neurosci Res. 1993;36(6):681–693. doi: 10.1002/jnr.490360609. [DOI] [PubMed] [Google Scholar]
- 111.Huang WC, Qiao Y, Xu L, Kacimi R, Sun X, Giffard RG, Yenari MA. Direct protection of cultured neurons from ischemia-like injury by minocycline. Anat Cell Biol. 2010;43(4):325–331. doi: 10.5115/acb.2010.43.4.325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Watanabe H, Abe H, Takeuchi S, Tanaka R. Protective effect of microglial conditioning medium on neuronal damage induced by glutamate. Neurosci Lett. 2000;289(1):53–56. doi: 10.1016/s0304-3940(00)01252-0. [DOI] [PubMed] [Google Scholar]
- 113.Lalancette-Hebert M, Gowing G, Simard A, Weng YC, Kriz J. Selective ablation of proliferating microglial cells exacerbates ischemic injury in the brain. J Neurosci. 2007;27(10):2596–2605. doi: 10.1523/JNEUROSCI.5360-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Weston RM, Jones NM, Jarrott B, Callaway JK. Inflammatory cell infiltration after endothelin-1-induced cerebral ischemia: histochemical and myeloperoxidase correlation with temporal changes in brain injury. J Cereb Blood Flow Metab. 2007;27(1):100–114. doi: 10.1038/sj.jcbfm.9600324. [DOI] [PubMed] [Google Scholar]
- 115.Frank-Cannon TC, Alto LT, McAlpine FE, Tansey MG. Does neuroinflammation fan the flame in neurodegenerative diseases? Mol Neurodegener. 2009;4:47. doi: 10.1186/1750-1326-4-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Benveniste EN. Cytokine actions in the central nervous system. Cytokine Growth Factor Rev. 1998;9(3–4):259–275. doi: 10.1016/s1359-6101(98)00015-x. [DOI] [PubMed] [Google Scholar]
- 117.Che X, Ye W, Panga L, Wu DC, Yang GY. Monocyte chemoattractant protein-1 expressed in neurons and astrocytes during focal ischemia in mice. Brain Res. 2001;902(2):171–177. doi: 10.1016/s0006-8993(01)02328-9. [DOI] [PubMed] [Google Scholar]
- 118.Pekny M, Nilsson M. Astrocyte activation and reactive gliosis. Glia. 2005;50(4):427–434. doi: 10.1002/glia.20207. [DOI] [PubMed] [Google Scholar]
- 119.Kadhim HJ, Duchateau J, Sebire G. Cytokines and brain injury: invited review. J Intensive Care Med. 2008;23(4):236–249. doi: 10.1177/0885066608318458. [DOI] [PubMed] [Google Scholar]
- 120.Nowicka D, Rogozinska K, Aleksy M, Witte OW, Skangiel-Kramska J. Spatiotemporal dynamics of astroglial and microglial responses after photothrombotic stroke in the rat brain. Acta Neurobiol Exp (Wars) 2008;68(2):155–168. doi: 10.55782/ane-2008-1685. [DOI] [PubMed] [Google Scholar]
- 121.Zhu Y, Roth-Eichhorn S, Braun N, Culmsee C, Rami A, Krieglstein J. The expression of transforming growth factor-beta1 (TGF-beta1) in hippocampal neurons: a temporary upregulated protein level after transient forebrain ischemia in the rat. Brain Res. 2000;866(1–2):286–298. doi: 10.1016/s0006-8993(00)02240-x. [DOI] [PubMed] [Google Scholar]
- 122.Dong Y, Benveniste EN. Immune function of astrocytes. Glia. 2001;36(2):180–190. doi: 10.1002/glia.1107. [DOI] [PubMed] [Google Scholar]
- 123.Endoh M, Maiese K, Wagner J. Expression of the inducible form of nitric oxide synthase by reactive astrocytes after transient global ischemia. Brain Res. 1994;651(1–2):92–100. doi: 10.1016/0006-8993(94)90683-1. [DOI] [PubMed] [Google Scholar]
- 124.Donohue PJ, Richards CM, Brown SA, Hanscom HN, Buschman J, Thangada S, Hla T, Williams MS, Winkles JA. TWEAK is an endothelial cell growth and chemotactic factor that also potentiates FGF-2 and VEGF-A mitogenic activity. Arterioscler Thromb Vasc Biol. 2003;23(4):594–600. doi: 10.1161/01.ATV.0000062883.93715.37. [DOI] [PubMed] [Google Scholar]
- 125.Yepes M, Brown SA, Moore EG, Smith EP, Lawrence DA, Winkles JA. A soluble Fn14-Fc decoy receptor reduces infarct volume in a murine model of cerebral ischemia. Am J Pathol. 2005;166(2):511–520. doi: 10.1016/S0002-9440(10)62273-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Saas P, Boucraut J, Walker PR, Quiquerez AL, Billot M, Desplat-Jego S, Chicheportiche Y, Dietrich PY. TWEAK stimulation of astrocytes and the proinflammatory consequences. Glia. 2000;32(1):102–107. [PubMed] [Google Scholar]
- 127.Stanimirovic D, Satoh K. Inflammatory mediators of cerebral endothelium: a role in ischemic brain inflammation. Brain Pathol. 2000;10(1):113–126. doi: 10.1111/j.1750-3639.2000.tb00248.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Stanimirovic D, Shapiro A, Wong J, Hutchison J, Durkin J. The induction of ICAM-1 in human cerebromicrovascular endothelial cells (HCEC) by ischemia-like conditions promotes enhanced neutrophil/HCEC adhesion. J Neuroimmunol. 1997;76(1–2):193–205. doi: 10.1016/s0165-5728(97)00057-x. [DOI] [PubMed] [Google Scholar]
- 129.Liu T, Clark RK, McDonnell PC, Young PR, White RF, Barone FC, Feuerstein GZ. Tumor necrosis factor-alpha expression in ischemic neurons. Stroke. 1994;25(7):1481–1488. doi: 10.1161/01.str.25.7.1481. [DOI] [PubMed] [Google Scholar]
- 130.Sairanen T, Carpen O, Karjalainen-Lindsberg ML, Paetau A, Turpeinen U, Kaste M, Lindsberg PJ. Evolution of cerebral tumor necrosis factor-alpha production during human ischemic stroke. Stroke. 2001;32(8):1750–1758. doi: 10.1161/01.str.32.8.1750. [DOI] [PubMed] [Google Scholar]
- 131.Allan SM, Rothwell NJ. Cytokines and acute neurodegeneration. Nat Rev Neurosci. 2001 Oct;:734–744. doi: 10.1038/35094583. [DOI] [PubMed] [Google Scholar]
- 132.Clark WM, Rinker LG, Lessov NS, Hazel K, Hill JK, Stenzel-Poore M, Eckenstein F. Lack of interleukin-6 expression is not protective against focal central nervous system ischemia. Stroke. 2000;31(7):1715–1720. doi: 10.1161/01.str.31.7.1715. [DOI] [PubMed] [Google Scholar]
- 133.Herrmann O, Tarabin V, Suzuki S, Attigah N, Coserea I, Schneider A, Vogel J, Prinz S, Schwab S, Monyer H, Brombacher F, Schwaninger M. Regulation of body temperature and neuroprotection by endogenous interleukin-6 in cerebral ischemia. J Cereb Blood Flow Metab. 2003;23(4):406–415. doi: 10.1097/01.WCB.0000055177.50448.FA. [DOI] [PubMed] [Google Scholar]
- 134.Yamashita T, Sawamoto K, Suzuki S, Suzuki N, Adachi K, Kawase T, Mihara M, Ohsugi Y, Abe K, Okano H. Blockade of interleukin-6 signaling aggravates ischemic cerebral damage in mice: possible involvement of Stat3 activation in the protection of neurons. J Neurochem. 2005;94(2):459–468. doi: 10.1111/j.1471-4159.2005.03227.x. [DOI] [PubMed] [Google Scholar]
- 135.Pinteaux E, Rothwell NJ, Boutin H. Neuroprotective actions of endogenous interleukin-1 receptor antagonist (IL-1ra) are mediated by glia. Glia. 2006;53(5):551–556. doi: 10.1002/glia.20308. [DOI] [PubMed] [Google Scholar]
- 136.Pradillo JM, Denes A, Greenhalgh AD, Boutin H, Drake C, McColl BW, Barton E, Proctor SD, Russell JC, Rothwell NJ, Allan SM. Delayed administration of interleukin-1 receptor antagonist reduces ischemic brain damage and inflammation in comorbid rats. J Cereb Blood Flow Metab. 2012;32(9):1810–1819. doi: 10.1038/jcbfm.2012.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Allan SM, Tyrrell PJ, Rothwell NJ. Interleukin-1 and neuronal injury. Nat Rev Immunol. 2005;5(8):629–640. doi: 10.1038/nri1664. [DOI] [PubMed] [Google Scholar]
- 138.Rothwell NJ, Luheshi GN. Interleukin 1 in the brain: biology, pathology and therapeutic target. Trends Neurosci. 2000;23(12):618–625. doi: 10.1016/s0166-2236(00)01661-1. [DOI] [PubMed] [Google Scholar]
- 139.Haqqani AS, Nesic M, Preston E, Baumann E, Kelly J, Stanimirovic D. Characterization of vascular protein expression patterns in cerebral ischemia/reperfusion using laser capture microdissection and ICAT-nanoLC-MS/MS. Faseb J. 2005;19(13):1809–1821. doi: 10.1096/fj.05-3793com. [DOI] [PubMed] [Google Scholar]
- 140.Minami M, Katayama T, Satoh M. Brain cytokines and chemokines: roles in ischemic injury and pain. J Pharmacol Sci. 2006;100(5):461–470. doi: 10.1254/jphs.crj06005x. [DOI] [PubMed] [Google Scholar]
- 141.Boutin H, LeFeuvre RA, Horai R, Asano M, Iwakura Y, Rothwell NJ. Role of IL-1alpha and IL-1beta in ischemic brain damage. J Neurosci. 2001;21(15):5528–5534. doi: 10.1523/JNEUROSCI.21-15-05528.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Yamasaki Y, Matsuura N, Shozuhara H, Onodera H, Itoyama Y, Kogure K. Interleukin-1 as a pathogenetic mediator of ischemic brain damage in rats. Stroke. 1995;26(4):676–680. doi: 10.1161/01.str.26.4.676. discussion 681. [DOI] [PubMed] [Google Scholar]
- 143.Ding C, He Q, Li PA. Diabetes increases expression of ICAM after a brief period of cerebral ischemia. J Neuroimmunol. 2005;161(1–2):61–67. doi: 10.1016/j.jneuroim.2004.12.011. [DOI] [PubMed] [Google Scholar]
- 144.Hara H, Friedlander RM, Gagliardini V, Ayata C, Fink K, Huang Z, Shimizu-Sasamata M, Yuan J, Moskowitz MA. Inhibition of interleukin 1beta converting enzyme family proteases reduces ischemic and excitotoxic neuronal damage. Proc Natl Acad Sci U S A. 1997;94(5):2007–2012. doi: 10.1073/pnas.94.5.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Basu A, Lazovic J, Krady JK, Mauger DT, Rothstein RP, Smith MB, Levison SW. Interleukin-1 and the interleukin-1 type 1 receptor are essential for the progressive neurodegeneration that ensues subsequent to a mild hypoxic/ischemic injury. 2005 Jan; doi: 10.1038/sj.jcbfm.9600002. [DOI] [PubMed] [Google Scholar]
- 146.Mulcahy NJ, Ross J, Rothwell NJ, Loddick SA. Delayed administration of interleukin-1 receptor antagonist protects against transient cerebral ischaemia in the rat. Br J Pharmacol. 2003;140(3):471–476. doi: 10.1038/sj.bjp.0705462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Yang GY, Zhao YJ, Davidson BL, Betz AL. Overexpression of interleukin-1 receptor antagonist in the mouse brain reduces ischemic brain injury. Brain Res. 1997;751(2):181–188. doi: 10.1016/s0006-8993(96)01277-2. [DOI] [PubMed] [Google Scholar]
- 148.Murakami Y, Saito K, Hara A, Zhu Y, Sudo K, Niwa M, Fujii H, Wada H, Ishiguro H, Mori H, Seishima M. Increases in tumor necrosis factor-alpha following transient global cerebral ischemia do not contribute to neuron death in mouse hippocampus. J Neurochem. 2005;93(6):1616–1622. doi: 10.1111/j.1471-4159.2005.03163.x. [DOI] [PubMed] [Google Scholar]
- 149.Offner H, Subramanian S, Parker SM, Afentoulis ME, Vandenbark AA, Hurn PD. Experimental stroke induces massive, rapid activation of the peripheral immune system. J Cereb Blood Flow Metab. 2006;26(5):654–665. doi: 10.1038/sj.jcbfm.9600217. [DOI] [PubMed] [Google Scholar]
- 150.Uno H, Matsuyama T, Akita H, Nishimura H, Sugita M. Induction of tumor necrosis factor-alpha in the mouse hippocampus following transient forebrain ischemia. J Cereb Blood Flow Metab. 1997;17(5):491–499. doi: 10.1097/00004647-199705000-00002. [DOI] [PubMed] [Google Scholar]
- 151.Hallenbeck JM. The many faces of tumor necrosis factor in stroke. Nat Med. 2002;8(12):1363–1368. doi: 10.1038/nm1202-1363. [DOI] [PubMed] [Google Scholar]
- 152.Pan W, Kastin AJ. Tumor necrosis factor and stroke: role of the blood-brain barrier. Prog Neurobiol. 2007;83(6):363–374. doi: 10.1016/j.pneurobio.2007.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Yang GY, Gong C, Qin Z, Ye W, Mao Y, Bertz AL. Inhibition of TNFalpha attenuates infarct volume and ICAM-1 expression in ischemic mouse brain. Neuroreport. 1998;9(9):2131–2134. doi: 10.1097/00001756-199806220-00041. [DOI] [PubMed] [Google Scholar]
- 154.Barone FC, Arvin B, White RF, Miller A, Webb CL, Willette RN, Lysko PG, Feuerstein GZ. Tumor necrosis factor-alpha. A mediator of focal ischemic brain injury. Stroke. 1997;28(6):1233–1244. doi: 10.1161/01.str.28.6.1233. [DOI] [PubMed] [Google Scholar]
- 155.Ginis I, Jaiswal R, Klimanis D, Liu J, Greenspon J, Hallenbeck JM. TNF-alpha-induced tolerance to ischemic injury involves differential control of NF-kappaB transactivation: the role of NF-kappaB association with p300 adaptor. J Cereb Blood Flow Metab. 2002;22(2):142–152. doi: 10.1097/00004647-200202000-00002. [DOI] [PubMed] [Google Scholar]
- 156.Bruce AJ, Boling W, Kindy MS, Peschon J, Kraemer PJ, Carpenter MK, Holtsberg FW, Mattson MP. Altered neuronal and microglial responses to excitotoxic and ischemic brain injury in mice lacking TNF receptors. Nat Med. 1996;2(7):788–794. doi: 10.1038/nm0796-788. [DOI] [PubMed] [Google Scholar]
- 157.Sriram K, O’Callaghan JP. Divergent roles for tumor necrosis factor-alpha in the brain. J Neuroimmune Pharmacol. 2007;2(2):140–153. doi: 10.1007/s11481-007-9070-6. [DOI] [PubMed] [Google Scholar]
- 158.Amantea D, Nappi G, Bernardi G, Bagetta G, Corasaniti MT. Post-ischemic brain damage: pathophysiology and role of inflammatory mediators. Febs J. 2009;276(1):13–26. doi: 10.1111/j.1742-4658.2008.06766.x. [DOI] [PubMed] [Google Scholar]
- 159.Pradillo JM, Romera C, Hurtado O, Cardenas A, Moro MA, Leza JC, Davalos A, Castillo J, Lorenzo P, Lizasoain I. TNFR1 upregulation mediates tolerance after brain ischemic preconditioning. J Cereb Blood Flow Metab. 2005;25(2):193–203. doi: 10.1038/sj.jcbfm.9600019. [DOI] [PubMed] [Google Scholar]
- 160.Smith CJ, Emsley HC, Gavin CM, Georgiou RF, Vail A, Barberan EM, del Zoppo GJ, Hallenbeck JM, Rothwell NJ, Hopkins SJ, Tyrrell PJ. Peak plasma interleukin-6 and other peripheral markers of inflammation in the first week of ischaemic stroke correlate with brain infarct volume, stroke severity and long-term outcome. BMC Neurol. 2004;4:2. doi: 10.1186/1471-2377-4-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Rallidis LS, Vikelis M, Panagiotakos DB, Rizos I, Zolindaki MG, Kaliva K, Kremastinos DT. Inflammatory markers and in-hospital mortality in acute ischaemic stroke. Atherosclerosis. 2006;189(1):193–197. doi: 10.1016/j.atherosclerosis.2005.11.032. [DOI] [PubMed] [Google Scholar]
- 162.Jung JE, Kim GS, Chan PH. Neuroprotection by interleukin-6 is mediated by signal transducer and activator of transcription 3 and antioxidative signaling in ischemic stroke. Stroke. 2011;42(12):3574–3579. doi: 10.1161/STROKEAHA.111.626648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Gertz K, Kronenberg G, Kalin RE, Baldinger T, Werner C, Balkaya M, Eom GD, Hellmann-Regen J, Krober J, Miller KR, Lindauer U, Laufs U, Dirnagl U, Heppner FL, Endres M. Essential role of interleukin-6 in post-stroke angiogenesis. Brain. 2012;135(Pt 6):1964–1980. doi: 10.1093/brain/aws075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Strle K, Zhou JH, Shen WH, Broussard SR, Johnson RW, Freund GG, Dantzer R, Kelley KW. Interleukin-10 in the brain. Crit Rev Immunol. 2001;21(5):427–449. [PubMed] [Google Scholar]
- 165.Spera PA, Ellison JA, Feuerstein GZ, Barone FC. IL-10 reduces rat brain injury following focal stroke. Neurosci Lett. 1998;251(3):189–192. doi: 10.1016/s0304-3940(98)00537-0. [DOI] [PubMed] [Google Scholar]
- 166.Ooboshi H, Ibayashi S, Shichita T, Kumai Y, Takada J, Ago T, Arakawa S, Sugimori H, Kamouchi M, Kitazono T, Iida M. Postischemic gene transfer of interleukin-10 protects against both focal and global brain ischemia. Circulation. 2005;111(7):913–919. doi: 10.1161/01.CIR.0000155622.68580.DC. [DOI] [PubMed] [Google Scholar]
- 167.Xiong X, Barreto GE, Xu L, Ouyang YB, Xie X, Giffard RG. Increased brain injury and worsened neurological outcome in interleukin-4 knockout mice after transient focal cerebral ischemia. Stroke. 2011;42(7):2026–2032. doi: 10.1161/STROKEAHA.110.593772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Flanders KC, Ren RF, Lippa CF. Transforming growth factor-betas in neurodegenerative disease. Prog Neurobiol. 1998;54(1):71–85. doi: 10.1016/s0301-0082(97)00066-x. [DOI] [PubMed] [Google Scholar]
- 169.Pang L, Ye W, Che XM, Roessler BJ, Betz AL, Yang GY. Reduction of inflammatory response in the mouse brain with adenoviral-mediated transforming growth factor-ss1 expression. Stroke. 2001;32(2):544–552. doi: 10.1161/01.str.32.2.544. [DOI] [PubMed] [Google Scholar]
- 170.Lu YZ, Lin CH, Cheng FC, Hsueh CM. Molecular mechanisms responsible for microglia-derived protection of Sprague-Dawley rat brain cells during in vitro ischemia. Neurosci Lett. 2005;373(2):159–164. doi: 10.1016/j.neulet.2004.10.004. [DOI] [PubMed] [Google Scholar]
- 171.Kim JS, Gautam SC, Chopp M, Zaloga C, Jones ML, Ward PA, Welch KM. Expression of monocyte chemoattractant protein-1 and macrophage inflammatory protein-1 after focal cerebral ischemia in the rat. J Neuroimmunol. 1995;56(2):127–134. doi: 10.1016/0165-5728(94)00138-e. [DOI] [PubMed] [Google Scholar]
- 172.Wang X, Feuerstein GZ. Induced expression of adhesion molecules following focal brain ischemia. J Neurotrauma. 1995;12(5):825–832. doi: 10.1089/neu.1995.12.825. [DOI] [PubMed] [Google Scholar]
- 173.Bajetto A, Bonavia R, Barbero S, Florio T, Schettini G. Chemokines and their receptors in the central nervous system. 2001 Jul;:147–184. doi: 10.1006/frne.2001.0214. [DOI] [PubMed] [Google Scholar]
- 174.Rossi D, Zlotnik A. The biology of chemokines and their receptors. Annu Rev Immunol. 2000;18:217–242. doi: 10.1146/annurev.immunol.18.1.217. [DOI] [PubMed] [Google Scholar]
- 175.Bizzarri C, Beccari AR, Bertini R, Cavicchia MR, Giorgini S, Allegretti M. ELR+ CXC chemokines and their receptors (CXC chemokine receptor 1 and CXC chemokine receptor 2) as new therapeutic targets. Pharmacol Ther. 2006;112(1):139–149. doi: 10.1016/j.pharmthera.2006.04.002. [DOI] [PubMed] [Google Scholar]
- 176.Gerard C, Rollins BJ. Chemokines and disease. Nat Immunol. 2001;2(2):108–115. doi: 10.1038/84209. [DOI] [PubMed] [Google Scholar]
- 177.Rollins BJ. Chemokines. Blood. 1997;90(3):909–928. [PubMed] [Google Scholar]
- 178.Semple BD, Kossmann T, Morganti-Kossmann MC. Role of chemokines in CNS health and pathology: a focus on the CCL2/CCR2 and CXCL8/CXCR2 networks. J Cereb Blood Flow Metab. 2010;30(3):459–473. doi: 10.1038/jcbfm.2009.240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Terao S, Yilmaz G, Stokes KY, Russell J, Ishikawa M, Kawase T, Granger DN. Blood cell-derived RANTES mediates cerebral microvascular dysfunction, inflammation, and tissue injury after focal ischemia-reperfusion. Stroke. 2008;39(9):2560–2570. doi: 10.1161/STROKEAHA.107.513150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Terao Y, Ohta H, Oda A, Nakagaito Y, Kiyota Y, Shintani Y. Macrophage inflammatory protein-3alpha plays a key role in the inflammatory cascade in rat focal cerebral ischemia. Neurosci Res. 2009;64(1):75–82. doi: 10.1016/j.neures.2009.01.017. [DOI] [PubMed] [Google Scholar]
- 181.Garau A, Bertini R, Colotta F, Casilli F, Bigini P, Cagnotto A, Mennini T, Ghezzi P, Villa P. Neuroprotection with the CXCL8 inhibitor repertaxin in transient brain ischemia. Cytokine. 2005;30(3):125–131. doi: 10.1016/j.cyto.2004.12.014. [DOI] [PubMed] [Google Scholar]
- 182.Chen Y, Hallenbeck JM, Ruetzler C, Bol D, Thomas K, Berman NE, Vogel SN. Overexpression of monocyte chemoattractant protein 1 in the brain exacerbates ischemic brain injury and is associated with recruitment of inflammatory cells. J Cereb Blood Flow Metab. 2003;23(6):748–755. doi: 10.1097/01.WCB.0000071885.63724.20. [DOI] [PubMed] [Google Scholar]
- 183.Tarozzo G, Campanella M, Ghiani M, Bulfone A, Beltramo M. Expression of fractalkine and its receptor, CX3CR1, in response to ischaemia-reperfusion brain injury in the rat. Eur J Neurosci. 2002;15(10):1663–1668. doi: 10.1046/j.1460-9568.2002.02007.x. [DOI] [PubMed] [Google Scholar]
- 184.Soriano SG, Amaravadi LS, Wang YF, Zhou H, Yu GX, Tonra JR, Fairchild-Huntress V, Fang Q, Dunmore JH, Huszar D, Pan Y. Mice deficient in fractalkine are less susceptible to cerebral ischemia-reperfusion injury. J Neuroimmunol. 2002;125(1–2):59–65. doi: 10.1016/s0165-5728(02)00033-4. [DOI] [PubMed] [Google Scholar]
- 185.Stamatovic SM, Shakui P, Keep RF, Moore BB, Kunkel SL, Van Rooijen N, Andjelkovic AV. Monocyte chemoattractant protein-1 regulation of blood-brain barrier permeability. J Cereb Blood Flow Metab. 2005;25(5):593–606. doi: 10.1038/sj.jcbfm.9600055. [DOI] [PubMed] [Google Scholar]
- 186.Newman MB, Willing AE, Manresa JJ, Davis-Sanberg C, Sanberg PR. Stroke-induced migration of human umbilical cord blood cells: time course and cytokines. Stem Cells Dev. 2005;14(5):576–586. doi: 10.1089/scd.2005.14.576. [DOI] [PubMed] [Google Scholar]
- 187.Kelly S, Bliss TM, Shah AK, Sun GH, Ma M, Foo WC, Masel J, Yenari MA, Weissman IL, Uchida N, Palmer T, Steinberg GK. Transplanted human fetal neural stem cells survive, migrate, and differentiate in ischemic rat cerebral cortex. Proc Natl Acad Sci U S A. 2004;101(32):11839–11844. doi: 10.1073/pnas.0404474101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Wang L, Li Y, Chen J, Gautam SC, Zhang Z, Lu M, Chopp M. Ischemic cerebral tissue and MCP-1 enhance rat bone marrow stromal cell migration in interface culture. Exp Hematol. 2002;30(7):831–836. doi: 10.1016/s0301-472x(02)00829-9. [DOI] [PubMed] [Google Scholar]
- 189.Wang L, Li Y, Chen X, Chen J, Gautam SC, Xu Y, Chopp M. MCP-1, MIP-1, IL-8 and ischemic cerebral tissue enhance human bone marrow stromal cell migration in interface culture. Hematology. 2002;7(2):113–117. doi: 10.1080/10245330290028588. [DOI] [PubMed] [Google Scholar]
- 190.Shichinohe H, Kuroda S, Yano S, Hida K, Iwasaki Y. Role of SDF-1/CXCR4 system in survival and migration of bone marrow stromal cells after transplantation into mice cerebral infarct. Brain Res. 2007;1183:138–147. doi: 10.1016/j.brainres.2007.08.091. [DOI] [PubMed] [Google Scholar]
- 191.Adibhatla RM, Hatcher JF, Larsen EC, Chen X, Sun D, Tsao FH. CDP-choline significantly restores phosphatidylcholine levels by differentially affecting phospholipase A2 and CTP: phosphocholine cytidylyltransferase after stroke. 2006 doi: 10.1074/jbc.M512112200. [DOI] [PubMed] [Google Scholar]
- 192.Sanchez-Moreno C, Dashe JF, Scott T, Thaler D, Folstein MF, Martin A. Decreased levels of plasma vitamin C and increased concentrations of inflammatory and oxidative stress markers after stroke. Stroke. 2004;35(1):163–168. doi: 10.1161/01.STR.0000105391.62306.2E. [DOI] [PubMed] [Google Scholar]
- 193.Bonventre JV, Huang Z, Taheri MR, O’Leary E, Li E, Moskowitz MA, Sapirstein A. Reduced fertility and postischaemic brain injury in mice deficient in cytosolic phospholipase A2. Nature. 1997;390(6660):622–625. doi: 10.1038/37635. [DOI] [PubMed] [Google Scholar]
- 194.Schwab JM, Beschorner R, Meyermann R, Gozalan F, Schluesener HJ. Persistent accumulation of cyclooxygenase-1-expressing microglial cells and macrophages and transient upregulation by endothelium in human brain injury. J Neurosurg. 2002;96(5):892–899. doi: 10.3171/jns.2002.96.5.0892. [DOI] [PubMed] [Google Scholar]
- 195.Iadecola C, Sugimoto K, Niwa K, Kazama K, Ross ME. Increased susceptibility to ischemic brain injury in cyclooxygenase-1-deficient mice. J Cereb Blood Flow Metab. 2001;21(12):1436–1441. doi: 10.1097/00004647-200112000-00008. [DOI] [PubMed] [Google Scholar]
- 196.Candelario-Jalil E, Gonzalez-Falcon A, Garcia-Cabrera M, Alvarez D, Al-Dalain S, Martinez G, Leon OS, Springer JE. Assessment of the relative contribution of COX-1 and COX-2 isoforms to ischemia-induced oxidative damage and neurodegeneration following transient global cerebral ischemia. J Neurochem. 2003;86(3):545–555. doi: 10.1046/j.1471-4159.2003.01812.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Nogawa S, Zhang F, Ross ME, Iadecola C. Cyclo-oxygenase-2 gene expression in neurons contributes to ischemic brain damage. J Neurosci. 1997;17(8):2746–2755. doi: 10.1523/JNEUROSCI.17-08-02746.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Iadecola C, Forster C, Nogawa S, Clark HB, Ross ME. Cyclooxygenase-2 immunoreactivity in the human brain following cerebral ischemia. Acta Neuropathol (Berl) 1999;98(1):9–14. doi: 10.1007/s004010051045. [DOI] [PubMed] [Google Scholar]
- 199.Sairanen T, Ristimaki A, Karjalainen-Lindsberg ML, Paetau A, Kaste M, Lindsberg PJ. Cyclooxygenase-2 is induced globally in infarcted human brain. Ann Neurol. 1998;43(6):738–747. doi: 10.1002/ana.410430608. [DOI] [PubMed] [Google Scholar]
- 200.Kawano T, Anrather J, Zhou P, Park L, Wang G, Frys KA, Kunz A, Cho S, Orio M, Iadecola C. Prostaglandin E2 EP1 receptors: downstream effectors of COX-2 neurotoxicity. Nat Med. 2006;12(2):225–229. doi: 10.1038/nm1362. [DOI] [PubMed] [Google Scholar]
- 201.Sugimoto K, Iadecola C. Delayed effect of administration of COX-2 inhibitor in mice with acute cerebral ischemia. Brain Res. 2003;960(1–2):273–276. doi: 10.1016/s0006-8993(02)03805-2. [DOI] [PubMed] [Google Scholar]
- 202.Iadecola C, Niwa K, Nogawa S, Zhao X, Nagayama M, Araki E, Morham S, Ross ME. Reduced susceptibility to ischemic brain injury and N-methyl-D-aspartate-mediated neurotoxicity in cyclooxygenase-2-deficient mice. Proc Natl Acad Sci U S A. 2001;98(3):1294–1299. doi: 10.1073/pnas.98.3.1294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Dore S, Otsuka T, Mito T, Sugo N, Hand T, Wu L, Hurn PD, Traystman RJ, Andreasson K. Neuronal overexpression of cyclooxygenase-2 increases cerebral infarction. Ann Neurol. 2003;54(2):155–162. doi: 10.1002/ana.10612. [DOI] [PubMed] [Google Scholar]
- 204.Manabe Y, Anrather J, Kawano T, Niwa K, Zhou P, Ross ME, Iadecola C. Prostanoids, not reactive oxygen species, mediate COX-2-dependent neurotoxicity. Ann Neurol. 2004;55(5):668–675. doi: 10.1002/ana.20078. [DOI] [PubMed] [Google Scholar]
- 205.Rao AM, Hatcher JF, Kindy MS, Dempsey RJ. Arachidonic acid and leukotriene C4: role in transient cerebral ischemia of gerbils. Neurochem Res. 1999;24(10):1225–1232. doi: 10.1023/a:1020916905312. [DOI] [PubMed] [Google Scholar]
- 206.Tomimoto H, Shibata M, Ihara M, Akiguchi I, Ohtani R, Budka H. A comparative study on the expression of cyclooxygenase and 5-lipoxygenase during cerebral ischemia in humans. Acta Neuropathol (Berl) 2002;104(6):601–607. doi: 10.1007/s00401-002-0590-0. [DOI] [PubMed] [Google Scholar]
- 207.Baskaya MK, Hu Y, Donaldson D, Maley M, Rao AM, Prasad MR, Dempsey RJ. Protective effect of the 5-lipoxygenase inhibitor AA-861 on cerebral edema after transient ischemia. J Neurosurg. 1996;85(1):112–116. doi: 10.3171/jns.1996.85.1.0112. [DOI] [PubMed] [Google Scholar]
- 208.Song Y, Wei EQ, Zhang WP, Zhang L, Liu JR, Chen Z. Minocycline protects PC12 cells from ischemic-like injury and inhibits 5-lipoxygenase activation. Neuroreport. 2004;15(14):2181–2184. doi: 10.1097/00001756-200410050-00007. [DOI] [PubMed] [Google Scholar]
- 209.Kitagawa K, Matsumoto M, Hori M. Cerebral ischemia in 5-lipoxygenase knockout mice. Brain Res. 2004;1004(1–2):198–202. doi: 10.1016/j.brainres.2004.01.018. [DOI] [PubMed] [Google Scholar]
- 210.Iadecola C, Zhang F, Xu S, Casey R, Ross ME. Inducible nitric oxide synthase gene expression in brain following cerebral ischemia. J Cereb Blood Flow Metab. 1995;15(3):378–384. doi: 10.1038/jcbfm.1995.47. [DOI] [PubMed] [Google Scholar]
- 211.Iadecola C, Zhang F, Xu X. Inhibition of inducible nitric oxide synthase ameliorates cerebral ischemic damage. Am J Physiol. 1995;268(1 Pt 2):R286–292. doi: 10.1152/ajpregu.1995.268.1.R286. [DOI] [PubMed] [Google Scholar]
- 212.Lakhan SE, Kirchgessner A, Hofer M. Inflammatory mechanisms in ischemic stroke: therapeutic approaches. J Transl Med. 2009;7:97. doi: 10.1186/1479-5876-7-97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Cui J, Holmes EH, Liu PK. Oxidative damage to the c-fos gene and reduction of its transcription after focal cerebral ischemia. J Neurochem. 1999;73(3):1164–1174. doi: 10.1046/j.1471-4159.1999.0731164.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Zhao X, Haensel C, Araki E, Ross ME, Iadecola C. Gene-dosing effect and persistence of reduction in ischemic brain injury in mice lacking inducible nitric oxide synthase. Brain Res. 2000;872(1–2):215–218. doi: 10.1016/s0006-8993(00)02459-8. [DOI] [PubMed] [Google Scholar]
- 215.Han HS, Qiao Y, Karabiyikoglu M, Giffard RG, Yenari MA. Influence of mild hypothermia on inducible nitric oxide synthase expression and reactive nitrogen production in experimental stroke and inflammation. J Neurosci. 2002;22(10):3921–3928. doi: 10.1523/JNEUROSCI.22-10-03921.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Coughlan T, Gibson C, Murphy S. Modulatory effects of progesterone on inducible nitric oxide synthase expression in vivo and in vitro. J Neurochem. 2005;93(4):932–942. doi: 10.1111/j.1471-4159.2005.03068.x. [DOI] [PubMed] [Google Scholar]
- 217.Park EM, Cho S, Frys KA, Glickstein SB, Zhou P, Anrather J, Ross ME, Iadecola C. Inducible nitric oxide synthase contributes to gender differences in ischemic brain injury. J Cereb Blood Flow Metab. 2006;26(3):392–401. doi: 10.1038/sj.jcbfm.9600194. [DOI] [PubMed] [Google Scholar]
- 218.Chan PH. Reactive oxygen radicals in signaling and damage in the ischemic brain. J Cereb Blood Flow Metab. 2001;21(1):2–14. doi: 10.1097/00004647-200101000-00002. [DOI] [PubMed] [Google Scholar]
- 219.Chan PH. Mitochondria and neuronal death/survival signaling pathways in cerebral ischemia. Neurochem Res. 2004;29(11):1943–1949. doi: 10.1007/s11064-004-6869-x. [DOI] [PubMed] [Google Scholar]
- 220.Lassegue B, Clempus RE. Vascular NAD(P)H oxidases: specific features, expression, and regulation. Am J Physiol Regul Integr Comp Physiol. 2003;285(2):R277–297. doi: 10.1152/ajpregu.00758.2002. [DOI] [PubMed] [Google Scholar]
- 221.Groemping Y, Rittinger K. Activation and assembly of the NADPH oxidase: a structural perspective. Biochem J. 2005;386(Pt 3):401–416. doi: 10.1042/BJ20041835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev. 2007;87(1):245–313. doi: 10.1152/physrev.00044.2005. [DOI] [PubMed] [Google Scholar]
- 223.Yenari MA, Xu L, Tang XN, Qiao Y, Giffard RG. Microglia potentiate damage to blood-brain barrier constituents: improvement by minocycline in vivo and in vitro. Stroke. 2006;37(4):1087–1093. doi: 10.1161/01.STR.0000206281.77178.ac. [DOI] [PubMed] [Google Scholar]
- 224.Liu W, Sood R, Chen Q, Sakoglu U, Hendren J, Cetin O, Miyake M, Liu KJ. Normobaric hyperoxia inhibits NADPH oxidase-mediated matrix metalloproteinase-9 induction in cerebral microvessels in experimental stroke. J Neurochem. 2008;107(5):1196–1205. doi: 10.1111/j.1471-4159.2008.05664.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Tang XN, Cairns B, Cairns N, Yenari MA. Apocynin improves outcome in experimental stroke with a narrow dose range. Neuroscience. 2008;154(2):556–562. doi: 10.1016/j.neuroscience.2008.03.090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Chen H, Song YS, Chan PH. Inhibition of NADPH oxidase is neuroprotective after ischemia-reperfusion. J Cereb Blood Flow Metab. 2009;29(7):1262–1272. doi: 10.1038/jcbfm.2009.47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Infanger DW, Sharma RV, Davisson RL. NADPH oxidases of the brain: distribution, regulation, and function. Antioxid Redox Signal. 2006;8(9–10):1583–1596. doi: 10.1089/ars.2006.8.1583. [DOI] [PubMed] [Google Scholar]
- 228.Kim MJ, Shin KS, Chung YB, Jung KW, Cha CI, Shin DH. Immunohistochemical study of p47Phox and gp91Phox distributions in rat brain. Brain Res. 2005;1040(1–2):178–186. doi: 10.1016/j.brainres.2005.01.066. [DOI] [PubMed] [Google Scholar]
- 229.Serrano F, Kolluri NS, Wientjes FB, Card JP, Klann E. NADPH oxidase immunoreactivity in the mouse brain. Brain Res. 2003;988(1–2):193–198. doi: 10.1016/s0006-8993(03)03364-x. [DOI] [PubMed] [Google Scholar]
- 230.Tejada-Simon MV, Serrano F, Villasana LE, Kanterewicz BI, Wu GY, Quinn MT, Klann E. Synaptic localization of a functional NADPH oxidase in the mouse hippocampus. Mol Cell Neurosci. 2005;29(1):97–106. doi: 10.1016/j.mcn.2005.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Vallet P, Charnay Y, Steger K, Ogier-Denis E, Kovari E, Herrmann F, Michel JP, Szanto I. Neuronal expression of the NADPH oxidase NOX4, and its regulation in mouse experimental brain ischemia. Neuroscience. 2005;132(2):233–238. doi: 10.1016/j.neuroscience.2004.12.038. [DOI] [PubMed] [Google Scholar]
- 232.Tang XN, Zheng Z, Giffard RG, Yenari MA. Significance of marrow-derived nicotinamide adenine dinucleotide phosphate oxidase in experimental ischemic stroke. Ann Neurol. 2011;70(4):606–615. doi: 10.1002/ana.22476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Walder CE, Green SP, Darbonne WC, Mathias J, Rae J, Dinauer MC, Curnutte JT, Thomas GR. Ischemic stroke injury is reduced in mice lacking a functional NADPH oxidase. Stroke. 1997;28(11):2252–2258. doi: 10.1161/01.str.28.11.2252. [DOI] [PubMed] [Google Scholar]
- 234.Suh SW, Shin BS, Ma H, Van Hoecke M, Brennan AM, Yenari MA, Swanson RA. Glucose and NADPH oxidase drive neuronal superoxide formation in stroke. Ann Neurol. 2008;64(6):654–663. doi: 10.1002/ana.21511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Brennan AM, Suh SW, Won SJ, Narasimhan P, Kauppinen TM, Lee H, Edling Y, Chan PH, Swanson RA. NADPH oxidase is the primary source of superoxide induced by NMDA receptor activation. Nat Neurosci. 2009;12(7):857–863. doi: 10.1038/nn.2334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Kahles T, Luedike P, Endres M, Galla HJ, Steinmetz H, Busse R, Neumann-Haefelin T, Brandes RP. NADPH oxidase plays a central role in blood-brain barrier damage in experimental stroke. Stroke. 2007;38(11):3000–3006. doi: 10.1161/STROKEAHA.107.489765. [DOI] [PubMed] [Google Scholar]
- 237.Tang SC, Arumugam TV, Xu X, Cheng A, Mughal MR, Jo DG, Lathia JD, Siler DA, Chigurupati S, Ouyang X, Magnus T, Camandola S, Mattson MP. Pivotal role for neuronal Toll-like receptors in ischemic brain injury and functional deficits. Proc Natl Acad Sci U S A. 2007;104(34):13798–13803. doi: 10.1073/pnas.0702553104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Wang Q, Tompkins KD, Simonyi A, Korthuis RJ, Sun AY, Sun GY. Apocynin protects against global cerebral ischemia-reperfusion-induced oxidative stress and injury in the gerbil hippocampus. Brain Res. 2006;1090(1):182–189. doi: 10.1016/j.brainres.2006.03.060. [DOI] [PubMed] [Google Scholar]
- 239.Takizawa S, Aratani Y, Fukuyama N, Maeda N, Hirabayashi H, Koyama H, Shinohara Y, Nakazawa H. Deficiency of myeloperoxidase increases infarct volume and nitrotyrosine formation in mouse brain. J Cereb Blood Flow Metab. 2002;22(1):50–54. doi: 10.1097/00004647-200201000-00006. [DOI] [PubMed] [Google Scholar]
- 240.Rosenberg GA. Matrix metalloproteinases in neuroinflammation. Glia. 2002;39(3):279–291. doi: 10.1002/glia.10108. [DOI] [PubMed] [Google Scholar]
- 241.Kelly MA, Shuaib A, Todd KG. Matrix metalloproteinase activation and blood-brain barrier breakdown following thrombolysis. Exp Neurol. 2006 doi: 10.1016/j.expneurol.2006.01.032. [DOI] [PubMed] [Google Scholar]
- 242.Yamashita T, Kamiya T, Deguchi K, Inaba T, Zhang H, Shang J, Miyazaki K, Ohtsuka A, Katayama Y, Abe K. Dissociation and protection of the neurovascular unit after thrombolysis and reperfusion in ischemic rat brain. J Cereb Blood Flow Metab. 2009;29(4):715–725. doi: 10.1038/jcbfm.2008.164. [DOI] [PubMed] [Google Scholar]
- 243.Rosenberg GA, Cunningham LA, Wallace J, Alexander S, Estrada EY, Grossetete M, Razhagi A, Miller K, Gearing A. Immunohistochemistry of matrix metalloproteinases in reperfusion injury to rat brain: activation of MMP-9 linked to stromelysin-1 and microglia in cell cultures. Brain Res. 2001;893(1–2):104–112. doi: 10.1016/s0006-8993(00)03294-7. [DOI] [PubMed] [Google Scholar]
- 244.Rosenberg GA, Estrada EY, Dencoff JE. Matrix metalloproteinases and TIMPs are associated with blood-brain barrier opening after reperfusion in rat brain. Stroke. 1998;29(10):2189–2195. doi: 10.1161/01.str.29.10.2189. [DOI] [PubMed] [Google Scholar]
- 245.Romanic AM, White RF, Arleth AJ, Ohlstein EH, Barone FC. Matrix metalloproteinase expression increases after cerebral focal ischemia in rats: inhibition of matrix metalloproteinase-9 reduces infarct size. Stroke. 1998;29(5):1020–1030. doi: 10.1161/01.str.29.5.1020. [DOI] [PubMed] [Google Scholar]
- 246.Pfefferkorn T, Rosenberg GA. Closure of the blood-brain barrier by matrix metalloproteinase inhibition reduces rtPA-mediated mortality in cerebral ischemia with delayed reperfusion. Stroke. 2003;34(8):2025–2030. doi: 10.1161/01.STR.0000083051.93319.28. [DOI] [PubMed] [Google Scholar]
- 247.Asahi M, Asahi K, Jung JC, del Zoppo GJ, Fini ME, Lo EH. Role for matrix metalloproteinase 9 after focal cerebral ischemia: effects of gene knockout and enzyme inhibition with BB-94. J Cereb Blood Flow Metab. 2000;20(12):1681–1689. doi: 10.1097/00004647-200012000-00007. [DOI] [PubMed] [Google Scholar]
- 248.Asahi M, Sumii T, Fini ME, Itohara S, Lo EH. Matrix metalloproteinase 2 gene knockout has no effect on acute brain injury after focal ischemia. Neuroreport. 2001 Sep 17;:3003–3007. doi: 10.1097/00001756-200109170-00050. [DOI] [PubMed] [Google Scholar]
- 249.Montaner J, Alvarez-Sabin J, Molina C, Angles A, Abilleira S, Arenillas J, Gonzalez MA, Monasterio J. Matrix metalloproteinase expression after human cardioembolic stroke: temporal profile and relation to neurological impairment. Stroke. 2001;32(8):1759–1766. doi: 10.1161/01.str.32.8.1759. [DOI] [PubMed] [Google Scholar]
- 250.Gidday JM, Gasche YG, Copin JC, Shah AR, Perez RS, Shapiro SD, Chan PH, Park TS. Leukocyte-derived matrix metalloproteinase-9 mediates blood-brain barrier breakdown and is proinflammatory after transient focal cerebral ischemia. Am J Physiol Heart Circ Physiol. 2005;289(2):H558–568. doi: 10.1152/ajpheart.01275.2004. [DOI] [PubMed] [Google Scholar]
- 251.Lee SR, Kim HY, Rogowska J, Zhao BQ, Bhide P, Parent JM, Lo EH. Involvement of matrix metalloproteinase in neuroblast cell migration from the subventricular zone after stroke. J Neurosci. 2006;26(13):3491–3495. doi: 10.1523/JNEUROSCI.4085-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Zhao BQ, Wang S, Kim HY, Storrie H, Rosen BR, Mooney DJ, Wang X, Lo EH. Role of matrix metalloproteinases in delayed cortical responses after stroke. Nat Med. 2006;12(4):441–445. doi: 10.1038/nm1387. [DOI] [PubMed] [Google Scholar]
- 253.Baeuerle PA, Henkel T, editors. Function and activation of NF-kappa B in the immune system. 1994. [DOI] [PubMed] [Google Scholar]
- 254.Cechetto DF. Role of nuclear factor kappa B in neuropathological mechanisms. Prog Brain Res. 2001;132:391–404. doi: 10.1016/S0079-6123(01)32090-3. [DOI] [PubMed] [Google Scholar]
- 255.Schneider A, Martin-Villalba A, Weih F, Vogel J, Wirth T, Schwaninger M. NF-kappaB is activated and promotes cell death in focal cerebral ischemia. Nat Med. 1999;5(5):554–559. doi: 10.1038/8432. [DOI] [PubMed] [Google Scholar]
- 256.Herrmann O, Baumann B, de Lorenzi R, Muhammad S, Zhang W, Kleesiek J, Malfertheiner M, Kohrmann M, Potrovita I, Maegele I, Beyer C, Burke JR, Hasan MT, Bujard H, Wirth T, Pasparakis M, Schwaninger M. IKK mediates ischemia-induced neuronal death. Nat Med. 2005;11(12):1322–1329. doi: 10.1038/nm1323. [DOI] [PubMed] [Google Scholar]
- 257.Ueno T, Sawa Y, Kitagawa-Sakakida S, Nishimura M, Morishita R, Kaneda Y, Kohmura E, Yoshimine T, Matsuda H. Nuclear factor-kappa B decoy attenuates neuronal damage after global brain ischemia: a future strategy for brain protection during circulatory arrest. J Thorac Cardiovasc Surg. 2001;122(4):720–727. doi: 10.1067/mtc.2001.115917. [DOI] [PubMed] [Google Scholar]
- 258.Hill WD, Hess DC, Carroll JE, Wakade CG, Howard EF, Chen Q, Cheng C, Martin-Studdard A, Waller JL, Beswick RA. The NF-kappaB inhibitor diethyldithiocarbamate (DDTC) increases brain cell death in a transient middle cerebral artery occlusion model of ischemia. Brain Res Bull. 2001;55(3):375–386. doi: 10.1016/s0361-9230(01)00503-2. [DOI] [PubMed] [Google Scholar]
- 259.Irving EA, Bamford M. Role of mitogen- and stress-activated kinases in ischemic injury. J Cereb Blood Flow Metab. 2002;22(6):631–647. doi: 10.1097/00004647-200206000-00001. [DOI] [PubMed] [Google Scholar]
- 260.Irving EA, Barone FC, Reith AD, Hadingham SJ, Parsons AA. Differential activation of MAPK/ERK and p38/SAPK in neurones and glia following focal cerebral ischaemia in the rat. Brain Res Mol Brain Res. 2000;77(1):65–75. doi: 10.1016/s0169-328x(00)00043-7. [DOI] [PubMed] [Google Scholar]
- 261.Sugino T, Nozaki K, Takagi Y, Hattori I, Hashimoto N, Moriguchi T, Nishida E. Activation of mitogen-activated protein kinases after transient forebrain ischemia in gerbil hippocampus. J Neurosci. 2000;20(12):4506–4514. doi: 10.1523/JNEUROSCI.20-12-04506.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Kyriakis JM, Avruch J. Mammalian mitogen-activated protein kinase signal transduction pathways activated by stress and inflammation. Physiol Rev. 2001;81(2):807–869. doi: 10.1152/physrev.2001.81.2.807. [DOI] [PubMed] [Google Scholar]
- 263.Zhang QG, Wang RM, Yin XH, Pan J, Xu TL, Zhang GY. Knockdown of POSH expression is neuroprotective through down-regulating activation of the MLK3-MKK4-JNK pathway following cerebral ischaemia in the rat hippocampal CA1 subfield. J Neurochem. 2005;95(3):784–795. doi: 10.1111/j.1471-4159.2005.03435.x. [DOI] [PubMed] [Google Scholar]
- 264.Gao Q, Li Y, Chopp M. Bone marrow stromal cells increase astrocyte survival via upregulation of phosphoinositide 3-kinase/threonine protein kinase and mitogen-activated protein kinase kinase/extracellular signal-regulated kinase pathways and stimulate astrocyte trophic factor gene expression after anaerobic insult. Neuroscience. 2005;136(1):123–134. doi: 10.1016/j.neuroscience.2005.06.091. [DOI] [PubMed] [Google Scholar]
- 265.Walton KM, DiRocco R, Bartlett BA, Koury E, Marcy VR, Jarvis B, Schaefer EM, Bhat RV. Activation of p38MAPK in microglia after ischemia. J Neurochem. 1998;70(4):1764–1767. doi: 10.1046/j.1471-4159.1998.70041764.x. [DOI] [PubMed] [Google Scholar]
- 266.Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002;10(2):417–426. doi: 10.1016/s1097-2765(02)00599-3. [DOI] [PubMed] [Google Scholar]
- 267.Thauerer B, Zur Nedden S, Baier-Bitterlich G. Purine nucleosides: endogenous neuroprotectants in hypoxic brain. J Neurochem. 2012;121(3):329–342. doi: 10.1111/j.1471-4159.2012.07692.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Liew FY, Xu D, Brint EK, O’Neill LA. Negative regulation of toll-like receptor-mediated immune responses. Nat Rev Immunol. 2005;5(6):446–458. doi: 10.1038/nri1630. [DOI] [PubMed] [Google Scholar]
- 269.Piccinini AM, Midwood KS. DAMPening inflammation by modulating TLR signalling. Mediators Inflamm. 2010;2010 doi: 10.1155/2010/672395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Carty M, Bowie AG. Evaluating the role of Toll-like receptors in diseases of the central nervous system. Biochem Pharmacol. 2011;81(7):825–837. doi: 10.1016/j.bcp.2011.01.003. [DOI] [PubMed] [Google Scholar]
- 271.Kong Y, Le Y. Toll-like receptors in inflammation of the central nervous system. Int Immunopharmacol. 2011;11(10):1407–1414. doi: 10.1016/j.intimp.2011.04.025. [DOI] [PubMed] [Google Scholar]
- 272.Yang QW, Lu FL, Zhou Y, Wang L, Zhong Q, Lin S, Xiang J, Li JC, Fang CQ, Wang JZ. HMBG1 mediates ischemia-reperfusion injury by TRIF-adaptor independent Toll-like receptor 4 signaling. J Cereb Blood Flow Metab. 2011;31(2):593–605. doi: 10.1038/jcbfm.2010.129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273.Shichita T, Hasegawa E, Kimura A, Morita R, Sakaguchi R, Takada I, Sekiya T, Ooboshi H, Kitazono T, Yanagawa T, Ishii T, Takahashi H, Mori S, Nishibori M, Kuroda K, Akira S, Miyake K, Yoshimura A. Peroxiredoxin family proteins are key initiators of post-ischemic inflammation in the brain. Nat Med. 2012;18(6):911–917. doi: 10.1038/nm.2749. [DOI] [PubMed] [Google Scholar]
- 274.Marsh B, Stevens SL, Packard AE, Gopalan B, Hunter B, Leung PY, Harrington CA, Stenzel-Poore MP. Systemic lipopolysaccharide protects the brain from ischemic injury by reprogramming the response of the brain to stroke: a critical role for IRF3. J Neurosci. 2009;29(31):9839–9849. doi: 10.1523/JNEUROSCI.2496-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Takeda K, Akira S. Toll-like receptors in innate immunity. Int Immunol. 2005;17(1):1–14. doi: 10.1093/intimm/dxh186. [DOI] [PubMed] [Google Scholar]
- 276.Ziegler G, Harhausen D, Schepers C, Hoffmann O, Rohr C, Prinz V, Konig J, Lehrach H, Nietfeld W, Trendelenburg G. TLR2 has a detrimental role in mouse transient focal cerebral ischemia. Biochem Biophys Res Commun. 2007;359(3):574–579. doi: 10.1016/j.bbrc.2007.05.157. [DOI] [PubMed] [Google Scholar]
- 277.Lalancette-Hebert M, Phaneuf D, Soucy G, Weng YC, Kriz J. Live imaging of Toll-like receptor 2 response in cerebral ischaemia reveals a role of olfactory bulb microglia as modulators of inflammation. Brain. 2009;132(Pt 4):940–954. doi: 10.1093/brain/awn345. [DOI] [PubMed] [Google Scholar]
- 278.Tu XK, Yang WZ, Shi SS, Wang CH, Zhang GL, Ni TR, Chen CM, Wang R, Jia JW, Song QM. Spatio-temporal distribution of inflammatory reaction and expression of TLR2/4 signaling pathway in rat brain following permanent focal cerebral ischemia. Neurochem Res. 2010;35(8):1147–1155. doi: 10.1007/s11064-010-0167-6. [DOI] [PubMed] [Google Scholar]
- 279.Lv M, Liu Y, Zhang J, Sun L, Liu Z, Zhang S, Wang B, Su D, Su Z. Roles of inflammation response in microglia cell through Toll-like receptors 2/interleukin-23/interleukin-17 pathway in cerebral ischemia/reperfusion injury. Neuroscience. 2011;176:162–172. doi: 10.1016/j.neuroscience.2010.11.066. [DOI] [PubMed] [Google Scholar]
- 280.Yao H, Felfly H, Wang J, Zhou D, Haddad GG. DIDS protects against neuronal injury by blocking Toll-like receptor 2 activated-mechanisms. J Neurochem. 2009;108(3):835–846. doi: 10.1111/j.1471-4159.2008.05838.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Abe T, Shimamura M, Jackman K, Kurinami H, Anrather J, Zhou P, Iadecola C. Key role of CD36 in Toll-like receptor 2 signaling in cerebral ischemia. Stroke. 2010;41(5):898–904. doi: 10.1161/STROKEAHA.109.572552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282.Ziegler G, Freyer D, Harhausen D, Khojasteh U, Nietfeld W, Trendelenburg G. Blocking TLR2 in vivo protects against accumulation of inflammatory cells and neuronal injury in experimental stroke. J Cereb Blood Flow Metab. 2011;31(2):757–766. doi: 10.1038/jcbfm.2010.161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Hua F, Ma J, Ha T, Kelley JL, Kao RL, Schweitzer JB, Kalbfleisch JH, Williams DL, Li C. Differential roles of TLR2 and TLR4 in acute focal cerebral ischemia/reperfusion injury in mice. Brain Res. 2009;1262:100–108. doi: 10.1016/j.brainres.2009.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284.Hyakkoku K, Hamanaka J, Tsuruma K, Shimazawa M, Tanaka H, Uematsu S, Akira S, Inagaki N, Nagai H, Hara H. Toll-like receptor 4 (TLR4), but not TLR3 or TLR9, knock-out mice have neuroprotective effects against focal cerebral ischemia. Neuroscience. 2010;171(1):258–267. doi: 10.1016/j.neuroscience.2010.08.054. [DOI] [PubMed] [Google Scholar]
- 285.Caso JR, Pradillo JM, Hurtado O, Lorenzo P, Moro MA, Lizasoain I. Toll-like receptor 4 is involved in brain damage and inflammation after experimental stroke. Circulation. 2007;115(12):1599–1608. doi: 10.1161/CIRCULATIONAHA.106.603431. [DOI] [PubMed] [Google Scholar]
- 286.Lehnardt S, Lachance C, Patrizi S, Lefebvre S, Follett PL, Jensen FE, Rosenberg PA, Volpe JJ, Vartanian T. The toll-like receptor TLR4 is necessary for lipopolysaccharide-induced oligodendrocyte injury in the CNS. J Neurosci. 2002;22(7):2478–2486. doi: 10.1523/JNEUROSCI.22-07-02478.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287.Yang QW, Li JC, Lu FL, Wen AQ, Xiang J, Zhang LL, Huang ZY, Wang JZ. Upregulated expression of toll-like receptor 4 in monocytes correlates with severity of acute cerebral infarction. J Cereb Blood Flow Metab. 2008;28(9):1588–1596. doi: 10.1038/jcbfm.2008.50. [DOI] [PubMed] [Google Scholar]
- 288.Cao CX, Yang QW, Lv FL, Cui J, Fu HB, Wang JZ. Reduced cerebral ischemia-reperfusion injury in Toll-like receptor 4 deficient mice. Biochem Biophys Res Commun. 2007;353(2):509–514. doi: 10.1016/j.bbrc.2006.12.057. [DOI] [PubMed] [Google Scholar]
- 289.Caso JR, Pradillo JM, Hurtado O, Leza JC, Moro MA, Lizasoain I. Toll-like receptor 4 is involved in subacute stress-induced neuroinflammation and in the worsening of experimental stroke. Stroke. 2008;39(4):1314–1320. doi: 10.1161/STROKEAHA.107.498212. [DOI] [PubMed] [Google Scholar]
- 290.Kilic U, Kilic E, Matter CM, Bassetti CL, Hermann DM. TLR-4 deficiency protects against focal cerebral ischemia and axotomy-induced neurodegeneration. Neurobiol Dis. 2008;31(1):33–40. doi: 10.1016/j.nbd.2008.03.002. [DOI] [PubMed] [Google Scholar]
- 291.Skaper SD. Ion channels on microglia: therapeutic targets for neuroprotection. CNS Neurol Disord Drug Targets. 2011;10(1):44–56. doi: 10.2174/187152711794488638. [DOI] [PubMed] [Google Scholar]
- 292.Melani A, Amadio S, Gianfriddo M, Vannucchi MG, Volonte C, Bernardi G, Pedata F, Sancesario G. P2X7 receptor modulation on microglial cells and reduction of brain infarct caused by middle cerebral artery occlusion in rat. J Cereb Blood Flow Metab. 2006;26(7):974–982. doi: 10.1038/sj.jcbfm.9600250. [DOI] [PubMed] [Google Scholar]
- 293.Yanagisawa D, Kitamura Y, Takata K, Hide I, Nakata Y, Taniguchi T. Possible involvement of P2X7 receptor activation in microglial neuroprotection against focal cerebral ischemia in rats. Biol Pharm Bull. 2008;31(6):1121–1130. doi: 10.1248/bpb.31.1121. [DOI] [PubMed] [Google Scholar]
- 294.Arbeloa J, Perez-Samartin A, Gottlieb M, Matute C. P2X7 receptor blockade prevents ATP excitotoxicity in neurons and reduces brain damage after ischemia. Neurobiol Dis. 2012;45(3):954–961. doi: 10.1016/j.nbd.2011.12.014. [DOI] [PubMed] [Google Scholar]
- 295.Chu K, Yin B, Wang J, Peng G, Liang H, Xu Z, Du Y, Fang M, Xia Q, Luo B. Inhibition of P2X7 receptor ameliorates transient global cerebral ischemia/reperfusion injury via modulating inflammatory responses in the rat hippocampus. J Neuroinflammation. 2012;9:69. doi: 10.1186/1742-2094-9-69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296.Inoue K. Purinergic systems in microglia. Cell Mol Life Sci. 2008;65(19):3074–3080. doi: 10.1007/s00018-008-8210-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297.Irino Y, Nakamura Y, Inoue K, Kohsaka S, Ohsawa K. Akt activation is involved in P2Y12 receptor-mediated chemotaxis of microglia. J Neurosci Res. 2008;86(7):1511–1519. doi: 10.1002/jnr.21610. [DOI] [PubMed] [Google Scholar]
- 298.Webster CM, McManus A, Hokari M, Tang XN, Yenari MA. P2Y12 receptors in ischemic inflammation: A new role for clopidogrel? Stroke. 2011;42(3):e296. [Google Scholar]
- 299.Ajmo CT, Jr, Vernon DO, Collier L, Hall AA, Garbuzova-Davis S, Willing A, Pennypacker KR. The spleen contributes to stroke-induced neurodegeneration. J Neurosci Res. 2008;86(10):2227–2234. doi: 10.1002/jnr.21661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300.Gendron A, Teitelbaum J, Cossette C, Nuara S, Dumont M, Geadah D, du Souich P, Kouassi E. Temporal effects of left versus right middle cerebral artery occlusion on spleen lymphocyte subsets and mitogenic response in Wistar rats. Brain Res. 2002;955(1–2):85–97. doi: 10.1016/s0006-8993(02)03368-1. [DOI] [PubMed] [Google Scholar]
- 301.Martin A, Aguirre J, Sarasa-Renedo A, Tsoukatou D, Garofalakis A, Meyer H, Mamalaki C, Ripoll J, Planas AM. Imaging changes in lymphoid organs in vivo after brain ischemia with three-dimensional fluorescence molecular tomography in transgenic mice expressing green fluorescent protein in T lymphocytes. Mol Imaging. 2008;7(4):157–167. [PubMed] [Google Scholar]
- 302.Prass K, Braun JS, Dirnagl U, Meisel C, Meisel A. Stroke propagates bacterial aspiration to pneumonia in a model of cerebral ischemia. Stroke. 2006;37(10):2607–2612. doi: 10.1161/01.STR.0000240409.68739.2b. [DOI] [PubMed] [Google Scholar]
- 303.Aslanyan S, Weir CJ, Diener HC, Kaste M, Lees KR. Pneumonia and urinary tract infection after acute ischaemic stroke: a tertiary analysis of the GAIN International trial. Eur J Neurol. 2004;11(1):49–53. doi: 10.1046/j.1468-1331.2003.00749.x. [DOI] [PubMed] [Google Scholar]
- 304.Hilker R, Poetter C, Findeisen N, Sobesky J, Jacobs A, Neveling M, Heiss WD. Nosocomial pneumonia after acute stroke: implications for neurological intensive care medicine. Stroke. 2003;34(4):975–981. doi: 10.1161/01.STR.0000063373.70993.CD. [DOI] [PubMed] [Google Scholar]
- 305.Langhorne P, Stott DJ, Robertson L, MacDonald J, Jones L, McAlpine C, Dick F, Taylor GS, Murray G. Medical complications after stroke: a multicenter study. Stroke. 2000;31(6):1223–1229. doi: 10.1161/01.str.31.6.1223. [DOI] [PubMed] [Google Scholar]
- 306.Urra X, Cervera A, Villamor N, Planas AM, Chamorro A. Harms and benefits of lymphocyte subpopulations in patients with acute stroke. Neuroscience. 2009;158(3):1174–1183. doi: 10.1016/j.neuroscience.2008.06.014. [DOI] [PubMed] [Google Scholar]
- 307.Pavlov VA, Wang H, Czura CJ, Friedman SG, Tracey KJ. The cholinergic anti-inflammatory pathway: a missing link in neuroimmunomodulation. Mol Med. 2003;9(5–8):125–134. [PMC free article] [PubMed] [Google Scholar]
- 308.Tracey KJ. Physiology and immunology of the cholinergic antiinflammatory pathway. J Clin Invest. 2007;117(2):289–296. doi: 10.1172/JCI30555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 309.Enlimomab. Use of anti-ICAM-1 therapy in ischemic stroke: results of the Enlimomab Acute Stroke Trial. Neurology. 2001;57(8):1428–1434. doi: 10.1212/wnl.57.8.1428. [DOI] [PubMed] [Google Scholar]
- 310.Becker KJ. Anti-leukocyte antibodies: LeukArrest (Hu23F2G) and Enlimomab (R6.5) in acute stroke. Curr Med Res Opin. 2002;18(Suppl 2):s18–22. doi: 10.1185/030079902125000688. [DOI] [PubMed] [Google Scholar]
- 311.Schneider D, Berrouschot J, Brandt T, Hacke W, Ferbert A, Norris SH, Polmar SH, Schafer E. Safety, pharmacokinetics and biological activity of enlimomab (anti-ICAM-1 antibody): an open-label, dose escalation study in patients hospitalized for acute stroke. Eur Neurol. 1998;40(2):78–83. doi: 10.1159/000007962. [DOI] [PubMed] [Google Scholar]
- 312.Krams M, Lees KR, Hacke W, Grieve AP, Orgogozo JM, Ford GA. Acute Stroke Therapy by Inhibition of Neutrophils (ASTIN): an adaptive dose-response study of UK-279,276 in acute ischemic stroke. Stroke. 2003;34(11):2543–2548. doi: 10.1161/01.STR.0000092527.33910.89. [DOI] [PubMed] [Google Scholar]
- 313.Jiang N, Moyle M, Soule HR, Rote WE, Chopp M. Neutrophil inhibitory factor is neuroprotective after focal ischemia in rats. Ann Neurol. 1995;38(6):935–942. doi: 10.1002/ana.410380615. [DOI] [PubMed] [Google Scholar]
- 314.Caimi G, Canino B, Ferrara F, Montana M, Musso M, Porretto F, Carollo C, Catania A, Lo Presti R. Granulocyte integrins before and after activation in acute ischaemic stroke. J Neurol Sci. 2001;186(1–2):23–26. doi: 10.1016/s0022-510x(01)00495-6. [DOI] [PubMed] [Google Scholar]
- 315.Campanella M, Sciorati C, Tarozzo G, Beltramo M. Flow cytometric analysis of inflammatory cells in ischemic rat brain. Stroke. 2002;33(2):586–592. doi: 10.1161/hs0202.103399. [DOI] [PubMed] [Google Scholar]
- 316.Del Zoppo GJ. Why do all drugs work in animals but none in stroke patients? 1. Drugs promoting cerebral blood flow. J Intern Med. 1995;237(1):79–88. doi: 10.1111/j.1365-2796.1995.tb01144.x. [DOI] [PubMed] [Google Scholar]
- 317.Yenari MA, Han HS. Neuroprotective mechanisms of hypothermia in brain ischaemia. Nat Rev Neurosci. 2012;13(4):267–278. doi: 10.1038/nrn3174. [DOI] [PubMed] [Google Scholar]
- 318.Busto R, Dietrich WD, Globus MY, Ginsberg MD. Postischemic moderate hypothermia inhibits CA1 hippocampal ischemic neuronal injury. Neurosci Lett. 1989;101(3):299–304. doi: 10.1016/0304-3940(89)90549-1. [DOI] [PubMed] [Google Scholar]
- 319.Edwards AD, Yue X, Squier MV, Thoresen M, Cady EB, Penrice J, Cooper CE, Wyatt JS, Reynolds EO, Mehmet H. Specific inhibition of apoptosis after cerebral hypoxia-ischaemia by moderate post-insult hypothermia. Biochem Biophys Res Commun. 1995;217(3):1193–1199. doi: 10.1006/bbrc.1995.2895. [DOI] [PubMed] [Google Scholar]
- 320.Baldwin WA, Kirsch JR, Hurn PD, Toung WS, Traystman RJ. Hypothermic cerebral reperfusion and recovery from ischemia. Am J Physiol. 1991;261(3 Pt 2):H774–781. doi: 10.1152/ajpheart.1991.261.3.H774. [DOI] [PubMed] [Google Scholar]
- 321.Deng H, Han HS, Cheng D, Sun GH, Yenari MA. Mild hypothermia inhibits inflammation after experimental stroke and brain inflammation. Stroke. 2003;34(10):2495–2501. doi: 10.1161/01.STR.0000091269.67384.E7. [DOI] [PubMed] [Google Scholar]
- 322.Wang GJ, Deng HY, Maier CM, Sun GH, Yenari MA. Mild hypothermia reduces ICAM-1 expression, neutrophil infiltration and microglia/monocyte accumulation following experimental stroke. Neuroscience. 2002;114(4):1081–1090. doi: 10.1016/s0306-4522(02)00350-0. [DOI] [PubMed] [Google Scholar]
- 323.Perrone S, Szabo M, Bellieni CV, Longini M, Bango M, Kelen D, Treszl A, Negro S, Tataranno ML, Buonocore G. Whole body hypothermia and oxidative stress in babies with hypoxic-ischemic brain injury. Pediatr Neurol. 2010;43(4):236–240. doi: 10.1016/j.pediatrneurol.2010.05.009. [DOI] [PubMed] [Google Scholar]
- 324.Meybohm P, Gruenewald M, Zacharowski KD, Albrecht M, Lucius R, Fosel N, Hensler J, Zitta K, Bein B. Mild hypothermia alone or in combination with anesthetic post-conditioning reduces expression of inflammatory cytokines in the cerebral cortex of pigs after cardiopulmonary resuscitation. Crit Care. 2010;14(1):R21. doi: 10.1186/cc8879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 325.Webster CM, Kelly S, Koike MA, Chock VY, Giffard RG, Yenari MA. Inflammation and NFkappaB activation is decreased by hypothermia following global cerebral ischemia. Neurobiol Dis. 2009;33(2):301–312. doi: 10.1016/j.nbd.2008.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 326.Schmitt KR, Diestel A, Lehnardt S, Schwartlander R, Lange PE, Berger F, Ullrich O, Abdul-Khaliq H. Hypothermia suppresses inflammation via ERK signaling pathway in stimulated microglial cells. J Neuroimmunol. 2007;189(1–2):7–16. doi: 10.1016/j.jneuroim.2007.06.010. [DOI] [PubMed] [Google Scholar]
- 327.Choi JS, Park J, Suk K, Moon C, Park YK, Han HS. Mild Hypothermia Attenuates Intercellular Adhesion Molecule-1 Induction via Activation of Extracellular Signal-Regulated Kinase-1/2 in a Focal Cerebral Ischemia Model. Stroke Res Treat. 2011;2011:846716. doi: 10.4061/2011/846716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 328.Matsui T, Kakeda T. IL-10 production is reduced by hypothermia but augmented by hyperthermia in rat microglia. J Neurotrauma. 2008;25(6):709–715. doi: 10.1089/neu.2007.0482. [DOI] [PubMed] [Google Scholar]
- 329.Truettner JS, Suzuki T, Dietrich WD. The effect of therapeutic hypothermia on the expression of inflammatory response genes following moderate traumatic brain injury in the rat. Brain Res Mol Brain Res. 2005;138(2):124–134. doi: 10.1016/j.molbrainres.2005.04.006. [DOI] [PubMed] [Google Scholar]
- 330.Wang X, Zhu S, Drozda M, Zhang W, Stavrovskaya IG, Cattaneo E, Ferrante RJ, Kristal BS, Friedlander RM. Minocycline inhibits caspase-independent and -dependent mitochondrial cell death pathways in models of Huntington’s disease. Proc Natl Acad Sci U S A. 2003;100(18):10483–10487. doi: 10.1073/pnas.1832501100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 331.Tikka T, Fiebich BL, Goldsteins G, Keinanen R, Koistinaho J. Minocycline, a tetracycline derivative, is neuroprotective against excitotoxicity by inhibiting activation and proliferation of microglia. J Neurosci. 2001;21(8):2580–2588. doi: 10.1523/JNEUROSCI.21-08-02580.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 332.Koistinaho M, Malm TM, Kettunen MI, Goldsteins G, Starckx S, Kauppinen RA, Opdenakker G, Koistinaho J. Minocycline protects against permanent cerebral ischemia in wild type but not in matrix metalloprotease-9-deficient mice. J Cereb Blood Flow Metab. 2005;25(4):460–467. doi: 10.1038/sj.jcbfm.9600040. [DOI] [PubMed] [Google Scholar]
- 333.Liu Z, Fan Y, Won SJ, Neumann M, Hu D, Zhou L, Weinstein PR, Liu J. Chronic treatment with minocycline preserves adult new neurons and reduces functional impairment after focal cerebral ischemia. Stroke. 2007;38(1):146–152. doi: 10.1161/01.STR.0000251791.64910.cd. [DOI] [PubMed] [Google Scholar]
- 334.Tang XN, Wang Q, Koike MA, Cheng D, Goris ML, Blankenberg FG, Yenari MA. Monitoring the protective effects of minocycline treatment with radiolabeled annexin V in an experimental model of focal cerebral ischemia. J Nucl Med. 2007;48(11):1822–1828. doi: 10.2967/jnumed.107.041335. [DOI] [PubMed] [Google Scholar]
- 335.Lampl Y, Boaz M, Gilad R, Lorberboym M, Dabby R, Rapoport A, Anca-Hershkowitz M, Sadeh M. Minocycline treatment in acute stroke: an open-label, evaluator-blinded study. Neurology. 2007;69(14):1404–1410. doi: 10.1212/01.wnl.0000277487.04281.db. [DOI] [PubMed] [Google Scholar]
- 336.Fagan SC, Waller JL, Nichols FT, Edwards DJ, Pettigrew LC, Clark WM, Hall CE, Switzer JA, Ergul A, Hess DC. Minocycline to improve neurologic outcome in stroke (MINOS): a dose-finding study. Stroke. 2010;41(10):2283–2287. doi: 10.1161/STROKEAHA.110.582601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 337.Pelletier D, Hafler DA. Fingolimod for multiple sclerosis. N Engl J Med. 2012;366(4):339–347. doi: 10.1056/NEJMct1101691. [DOI] [PubMed] [Google Scholar]
- 338.Baumruker T, Billich A, Brinkmann V. FTY720, an immunomodulatory sphingolipid mimetic: translation of a novel mechanism into clinical benefit in multiple sclerosis. Expert Opin Investig Drugs. 2007;16(3):283–289. doi: 10.1517/13543784.16.3.283. [DOI] [PubMed] [Google Scholar]
- 339.Billich A, Bornancin F, Devay P, Mechtcheriakova D, Urtz N, Baumruker T. Phosphorylation of the immunomodulatory drug FTY720 by sphingosine kinases. J Biol Chem. 2003;278(48):47408–47415. doi: 10.1074/jbc.M307687200. [DOI] [PubMed] [Google Scholar]
- 340.Pfeilschifter W, Czech-Zechmeister B, Sujak M, Mirceska A, Koch A, Rami A, Steinmetz H, Foerch C, Huwiler A, Pfeilschifter J. Activation of sphingosine kinase 2 is an endogenous protective mechanism in cerebral ischemia. Biochem Biophys Res Commun. 2011;413(2):212–217. doi: 10.1016/j.bbrc.2011.08.070. [DOI] [PubMed] [Google Scholar]
- 341.Brinkmann V, Davis MD, Heise CE, Albert R, Cottens S, Hof R, Bruns C, Prieschl E, Baumruker T, Hiestand P, Foster CA, Zollinger M, Lynch KR. The immune modulator FTY720 targets sphingosine 1-phosphate receptors. J Biol Chem. 2002;277(24):21453–21457. doi: 10.1074/jbc.C200176200. [DOI] [PubMed] [Google Scholar]
- 342.Czech B, Pfeilschifter W, Mazaheri-Omrani N, Strobel MA, Kahles T, Neumann-Haefelin T, Rami A, Huwiler A, Pfeilschifter J. The immunomodulatory sphingosine 1-phosphate analog FTY720 reduces lesion size and improves neurological outcome in a mouse model of cerebral ischemia. Biochem Biophys Res Commun. 2009;389(2):251–256. doi: 10.1016/j.bbrc.2009.08.142. [DOI] [PubMed] [Google Scholar]
- 343.Hasegawa Y, Suzuki H, Sozen T, Rolland W, Zhang JH. Activation of sphingosine 1-phosphate receptor-1 by FTY720 is neuroprotective after ischemic stroke in rats. Stroke. 2010;41(2):368–374. doi: 10.1161/STROKEAHA.109.568899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 344.Wei Y, Yemisci M, Kim HH, Yung LM, Shin HK, Hwang SK, Guo S, Qin T, Alsharif N, Brinkmann V, Liao JK, Lo EH, Waeber C. Fingolimod provides long-term protection in rodent models of cerebral ischemia. Ann Neurol. 2011;69(1):119–129. doi: 10.1002/ana.22186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 345.Kimura A, Ohmori T, Kashiwakura Y, Ohkawa R, Madoiwa S, Mimuro J, Shimazaki K, Hoshino Y, Yatomi Y, Sakata Y. Antagonism of sphingosine 1-phosphate receptor-2 enhances migration of neural progenitor cells toward an area of brain. Stroke. 2008;39(12):3411–3417. doi: 10.1161/STROKEAHA.108.514612. [DOI] [PubMed] [Google Scholar]
- 346.Yagi H, Kamba R, Chiba K, Soga H, Yaguchi K, Nakamura M, Itoh T. Immunosuppressant FTY720 inhibits thymocyte emigration. Eur J Immunol. 2000;30(5):1435–1444. doi: 10.1002/(SICI)1521-4141(200005)30:5<1435::AID-IMMU1435>3.0.CO;2-O. [DOI] [PubMed] [Google Scholar]
- 347.Brinkmann V, Pinschewer DD, Feng L, Chen S. FTY720: altered lymphocyte traffic results in allograft protection. Transplantation. 2001;72(5):764–769. doi: 10.1097/00007890-200109150-00002. [DOI] [PubMed] [Google Scholar]
- 348.Jung CG, Kim HJ, Miron VE, Cook S, Kennedy TE, Foster CA, Antel JP, Soliven B. Functional consequences of S1P receptor modulation in rat oligodendroglial lineage cells. Glia. 2007;55(16):1656–1667. doi: 10.1002/glia.20576. [DOI] [PubMed] [Google Scholar]
- 349.Stessin AM, Gursel DB, Schwartz A, Parashar B, Kulidzhanov FG, Sabbas AM, Boockvar J, Nori D, Wernicke AG. FTY720, sphingosine 1-phosphate receptor modulator, selectively radioprotects hippocampal neural stem cells. Neurosci Lett. 2012;516(2):253–258. doi: 10.1016/j.neulet.2012.04.004. [DOI] [PubMed] [Google Scholar]
- 350.Liu J, Zhang C, Tao W, Liu M. Systematic review and meta-analysis of the efficacy of sphingosine-1-phosphate (S1P) receptor agonist FTY720 (fingolimod) in animal models of stroke. Int J Neurosci. 2013;123(3):163–169. doi: 10.3109/00207454.2012.749255. [DOI] [PubMed] [Google Scholar]