Abstract
Neuroprotection as approach to stroke therapy has recently seen a revival of sorts, fueled in part by the continuing necessity to improve acute stroke care, and in part by the identification of novel drug targets. 12/15-Lipoxygenase (12/15-LOX), one of the key enzymes of the arachidonic acid cascade, contributes to both neuronal cell death and vascular injury. Inhibition of 12/15-LOX may thus provide multifactorial protection against ischemic injury. Targeting 12/15-LOX and related eicosanoid pathways is the subject of this brief review.
Keywords: Ischemic stroke, neuroprotection, oxidative stress, eicosanoid, lipoxygenase, arachidonic acid cascade
Introduction
Neuroprotection in recent years has had a difficult standing in the stroke research field. Many failed clinical drug trials, notably the Saint II trial for the radical scavenger NXY-059[1], have contributed to a nihilistic view in both Industry and parts of Academia. With the identification of novel targets however, and given the continuing need for effective acute stroke treatment options, this stance is being re-evaluated. Several excellent recent reviews have covered various aspects of new approaches to neuroprotection, such as protection of the neurovascular unit, reduction of white matter injury, blockade of the inflammatory cascade, and others [2–6]. The approach of blocking the activity of 12/15-lipoxygenase (12/15-LOX) will be the major focus of this short review, and it touches upon several of these themes. In addition, we will very briefly highlight recent approaches to neuroprotection targeting other eicosanoid-producing pathways, including the cyclooxygenase and cytochrome P450 pathways.
The arachidonic acid cascade and lipoxygenase pathways
Free arachidonic acid is increased in the ischemic brain, mostly liberated from membrane phospholipids by phospholipases, especially phospholipase A2. The arachidonic acid cascade is a series of enzymatic reactions initiated by lipoxygenases, cyclooxygenases, and members of the cytochrome P450 family of proteins; collectively, the oxidation products are termed eicosanoids [7]. Lipoxygenases oxidize arachidonic acid at specific carbon atoms to generate a hydroperoxide (termed HPETE for hydroperoxyeicosatetraenoic acid), which is then reduced to the corresponding hydroxide (termed HETE). In this way, 5-LOX oxidizes arachidonic acid at C-5, generating 5-HPETE, which is then converted to 5-HETE and, in follow-up reactions by other enzymes, to pro-inflammatory leukotrienes. In a similar manner, 12/15-LOX generates a mixture of 12-HPETE and 15-HPETE, which again get reduced to the corresponding HETEs and undergo various follow-up reactions. In conjunction with other enzymes, this leads to production of both pro-inflammatory signaling molecules like the hepoxilins[8, 9], and anti-inflammatories like lipoxins[10]. Other polyunsaturated fatty acids besides arachidonic acid can serve as substrate; e.g., linoleic acid is oxidized to 9- and 13-HODE. Remarkably though, 12/15-LOX can also oxidize arachidonic acid and linoleic acid when they are esterified as phospholipids, meaning it can directly cause lipid peroxidation, and this may be its major contribution to ischemic brain damage (see below). In addition to 5-and 12/15-LOX, several other lipoxygenases exist, but little is known about their expression in the brain[11].
Another important class of eicosanoids are the prostaglandins, pro-inflammatory signaling molecules generated through the oxidation and cyclization of arachidonic acid by the cyclooxygenases COX-1 and COX-2. These enzymes are the target of non-steroidal anti-inflammatory drugs (NSAIDS), which have been extensively evaluated as stroke therapeutics. Finally, cytochrome P450 oxidizes arachidonic acid to 20-HETE as well as several epoxides (EETs), which are major vasoreactive molecules.
The role of 12/15-LOX in stroke
Brain injury subsequent to an ischemic stroke typically follows a pattern with distinct temporal characteristics. The initial infarct core, where a sudden massive drop in the blood supply causes rapid cell death, is surrounded by a penumbra, where tissue is at risk for further injury, but not yet irreversibly affected. Blood flow to the penumbra region is reduced, and several additional factors contribute to the progression of the infarct over hours and days. One of these is the acidification of tissue, with pH drops to around pH 6.5, sometimes lower. In addition, depolarized neurons and dying cells from both the core and the penumbra release large amounts of glutamate, leading to excitotoxicity in neurons whose glutamate receptors become overloaded. The blood – brain barrier becomes leaky, leading to edema and massive secondary injury[12]. Oxidative stress-related injury underlies many of these pathologies, and 12/15-LOX (also known as 15-LOX-1, leukocyte-type 12-LOX, or 12/15-LO) may be a major contributor.
In the meantime, a fair body of evidence has accumulated to document the importance of 12/15-LOX in ischemic stroke. Early evidence of arachidonic acid metabolizing enzymes being involved in ischemic injury came from the work of Nic Bazan, who documented increased levels of arachidonic acid in the brain following ischemia [13]. This was followed by Pak Chan and Robert Fishman’s demonstration that arachidonic acid injected into the brain can cause edema[14, 15]. Finally, Mike Moskowitz and colleagues showed in 1984 that lipoxygenase metabolites, including both 5-LOX derived leukotrienes and the 12/15-LOX metabolite 12-HETE, were increased in the gerbil forebrain following ischemia[16]. These early findings set the stage, but the next steps were not taken until much later. Fast forward to 1997, when Li, Maher, and Schubert showed an involvement of 12/15-LOX in oxidative stress-induced cell death in cultured neurons[17]. Then, in 2004/2005, the lab of Chandan Sen and ours independently demonstrated the specific involvement of 12/15-LOX in experimental stroke in rodents [18–20]. On the clinical side, a recent study showed elevated cerebrospinal fluid (CSF) levels of 12-HETE in patients with subarachnoid hemorrhage[21], and elevated levels of 12-HETE were also found in the CSF following traumatic brain injury[22]. Similar measurements in ischemic stroke patients have to our knowledge not been reported. Intriguingly, we have recently found 12/15-LOX increased in the peri-infarct cortex of two stroke patients, suggesting 12/15-LOX may contribute to stroke injury in humans, as well[23].
To date, most studies of 12/15-LOX in stroke have been carried out in rodents. In mice, inducing an experimental stroke by transiently occluding the middle cerebral artery with a filament (transient MCAO) leads initially to a striatal infarct that expands into the cortex over time. This cortical region that is adjacent to the core infarct and goes on to die is seen as representative for the penumbra region in human stroke patients. 12/15-LOX was found by immunohistochemistry to be expressed in this peri-infarct region of the cortex[20]. 12/15-LOX expression progressively increases over 24 hours. In the same cells, but with a slight time delay, apoptosis-inducing factor (AIF) is also increased[24], linking 12/15-LOX to an apoptotic cell death mechanism. Importantly, 12/15-LOX is increased both in neurons and in endothelial cells, suggesting it may contribute to both neuronal cell death and vascular injury[25].
Cell death through organelle damage: the toxic mechanism of 12/15-LOX
The enzymology of 12/15-LOX is complex, but important for its mechanism of action[11, 26–28]. 12/15-LOX contains a non-heme iron, which shuttles between Fe3+ and Fe2+ during the oxidation reaction. An oxidative environment is thus required for enzyme activity. The enzyme inserts molecular oxygen into a C-H bond, which thus forms an unstable hydroperoxide that can undergo various follow-up reactions. As substrate, 12/15-LOX uses polyunsaturated fatty acids (PUFAs), mostly arachidonic and linoleic acid. In contrast to other lipoxygenases, cyclooxygenases, and cytochrome P450s, 12/15-LOX can oxidize PUFAs even when they are membrane-bound, as phospholipids. The important consequence of this is that 12/15-LOX can directly attack and modify membranes. It does so by binding to the membrane, generating pores that release soluble proteins from the lumenal space of organelles, including mitochondria and endoplasmic reticulum (ER) membranes[29, 30].
Regulation of 12/15-LOX
In reticulocytes, the red blood cell precursors, organelle permeabilization is a desired effect, because it is the first step on the way to degrading the organelle. This is part of red blood cell maturation[31, 32], where mitochondria are absent in the mature erythrocyte, and it may also apply to lens cells of the eye[29, 33, 34]. But in almost all other cell types, organelle degradation is a detrimental process, and the cell thus limits 12/15-LOX activity through transcriptional, translational, and post-translational control. Depending on the cell type, transcriptional regulators of 12/15-LOX include STAT6[35], IL-4/IL-13[36], and GATA-6[37]. Translation of 12/15-LOX is regulated by proteins binding to the 3′-UTR of its mRNA, including hnRNP k and hnRNP E1[38, 39]. Nonetheless, the actual transcriptional or translational regulators leading to increased 12/15-LOX expression in the ischemic brain have not been reported to date. Post-translational control is exerted through a requirement for calcium to enable membrane binding[40], and through the intracellular antioxidant glutathione[17, 41]. Both of these become dysregulated in the ischemic brain, with glutathione levels dropping[42] and intracellular calcium levels rising[43]. This allows the 12/15-LOX to become activated and to start damaging organelles. Further activity increases are caused by the availability of free arachidonic acid, which is generated by phospholipase A2 under ischemic conditions[13]. To make matters worse, the enzyme is also activated by the hydroperoxides it produces, resulting in a destructive feed-forward loop[26]. These events are summarized in Fig. 1.
Figure 1. Model for activation of 12/15-LOX in the ischemic brain.
Under ischemic conditions, cPLA2 is activated to release arachidonic acid from membrane phospholipids. This free arachidonic acid, together with a calcium influx from the extracellular milieu and the depletion of glutathione, leads to activation of 12/15-LOX, which starts binding to and oxidizing mitochondrial and ER membranes. This permeabilizes the organelles, leading to release of calcium from ER stores and generation of ROS by mitochondria, which further activates 12/15-LOX. Mitochondria release the pro-apoptotic proteins cytochrome c and AIF, which then translocates to the nucleus. Abbreviations: AA, arachidonic acid; Cyt., cytochrome; AIF, apoptosis-inducing factor; ROS, reactive oxygen species.
Inhibition of 12/15-LOX is protective in models of oxidative stress-related cell death in several types of brain cells
On a cellular level, much has been learned from a model of cell death through glutathione depletion, termed oxidative glutamate toxicity or oxytosis [44, 45]. Immature neurons, when challenged with high concentrations of glutamate, die despite the absence of functional glutamate receptors[44, 46]. The reason is a cystine/glutamate antiporter in the cell membrane termed Xc−, which supplies a building block for glutathione to the cells. Glutathione is the major endogenous antioxidant of neurons, and glutathione depletion causes oxidative stress and subsequently, cell death. This cell death model is replicated in the neuronal cell line HT22, which has been used to study its mechanisms. In both immature neurons and HT22 cells, oxytosis is dependent on 12/15-LOX activity[17]. Besides neuronal cells, oligodendrocytes are also subject to a similar mode of cell death, elicited by removal of cysteine from the culture medium. In both mature oligodendrocytes and their precursors, cell death is prevented by 12/15-LOX inhibition[47–49].
In HT22 cells, glutathione depletion occurs over a time period of several hours, after which the cells start experiencing oxidative stress. 12/15-LOX then becomes membrane-associated and activated[17]. Mitochondrial membranes are especially susceptible[24], and the mitochondrial membrane potential breaks down. This can be replicated in vitro by incubating 12/15-LOX with isolated mitochondria, which also leads to production of reactive oxygen species (ROS). Subsequently, cytochrome c is released to the cytosol[50], but without activation of caspase-3[51]. Instead, cell death proceeds through translocation of the apoptosis-inducing factor AIF from mitochondria to the nucleus. Pro-apoptotic Bid presumably contributes to the process[52]. This pathway may be highly relevant to neuronal death in the ischemic penumbra, where increased 12/15-LOX coincides with increased AIF in the peri-infarct region [24]. Further support comes from a study showing the importance of glutathione peroxidase-4 in preventing 12/15-LOX-and AIF-dependent cell death in cells and in vivo[53]. The link between GPX-4 and 12/15-LOX had previously been demonstrated in vitro[54]. Finally, proteasome inhibition blocks cell death in HT22 cells downstream of mitochondrial damage, but at present it is unclear which specific step of the cell death program requires proteasome activity[51].
Targeting 12/15-LOX in rodent models of stroke: gene knockout vs. pharmacological inhibition
Increased levels of 12/15-LOX in the ischemic peri-infarct area suggested that blocking its activity might be protective. Supporting this idea, 12/15-LOX knockout mice exhibit reduced infarct sizes[19, 20], and less edema than the corresponding wild-type mice[25]. Several direct inhibitors of 12/15-LOX have been successful in focal ischemia models in vivo.
a-tocotrienol
To block 12/15-LOX activity pharmacologically, Chandan Sen’s group focused on a vitamin E compound, a-tocotrienol[55, 56]. Given by oral gavage for several weeks before the ischemic insult, a-tocotrienol reduced infarct size in rats[19]. It is not known what the time window would be for using a-tocotrienol in acute stroke, but it also reduced severity of stroke injury when dosed chronically in canines, suggesting it may have utility in secondary stroke prevention[57]. In addition to inhibiting 12/15-LOX and a general antioxidant effect, a-tocotrienol may also protect by targeting Multidrug Resistance-Associated Protein 1[58].
Baicalein
In our experiments, we initially used baicalein, a flavonoid natural product from the root of Scutellaria Baicalensis[59]. It is a component of Chinese herbal medicine, and an inhibitor of both 12-LOX and 12/15-LOX[60]. In mice, it reduced infarct size and edema formation to about the same extent as the gene knockout[20, 25]. Beneficial effects have also been reported for baicalein in ischemia models in rats, and an embolic stroke model in rabbits[61–63]. It is however, not very specific, also blocking xanthine oxidase and having strong antioxidant activity[59]. In addition, we needed 300 mg/kg to provide neuroprotection in mice, although lower dosages have been reported to be protective in rats. The reason for the different protective dosages is not clear, but might relate to different sources of the compound.
Novel 12/15-LOX inhibitors
In order to develop new inhibitors of 12/15-LOX as drug candidates, we have collaborated with the group of Ted Holman at the University of California, Santa Cruz. Using an in vitro assay with recombinant human 15-LOX-1 and in silico modeling, they identified several candidate compounds that blocked 12/15-LOX activity[64]. We then used the oxidative glutamate toxicity model in HT-22 cells as secondary screen for novel neuroprotective inhibitors targeting 12/15-LOX. Two out of three compounds were protective in this model, and also successfully rescued primary neurons and oligodendrocytic cells against cell death through glutathione depletion[49]. These compounds, 2-amino-N-(3-chlorophenyl)-4,5,6,7-tetrahydro-1-benzothiophene-3-carboxamide and 4-ethyl-6-(4-phenoxy-1H-pyrazol-3-yl)-1,3-benzenediol (now termed LOXBlock-1 and -3, respectively) are currently being evaluated in several models of ischemic brain injury. Because of their identification through modeling to the active site of 12/15-LOX, and the finding that they have very little radical scavenging activity, they may show higher specificity than most previously established 12/15-LOX inhibitors, although cross-reactivity with other lipoxygenases cannot be ruled out. We have recently shown that LOXBlock-1 provides efficient neuroprotection in mice, even when given four hours after onset of ischemia. In addition, it does not worsen outcome in a hemorrhage model, suggesting it may be safe to give as first-line stroke treatment[23].
Direct and indirect 12/15-LOX inhibition: antioxidant inhibitors
In addition to the attempts to specifically inhibit 12/15-LOX activity to reduce stroke injury presented above, several other approaches to neuroprotection should also at least partially block 12/15-LOX activity. These neuroprotectants broadly fall into 2 categories: One, treatment with antioxidant drugs or chelators that either directly or indirectly reduce 12/15-LOX activation; two, increasing endogenous antioxidant defenses.
Drugs that block 12/15-LOX activity
Several antioxidants and some non-antioxidants that have been shown to reduce infarct size and/or edema following focal ischemia either directly or indirectly inhibit 12/15-LOX. Among these, edaravone stands out because it has clinical approval as stroke treatment in Japan[65, 66]. Known as a radical scavenger, it also inhibits 12/15-LOX activity in vitro[67]. Similarly, the glutathione peroxidase mimic ebselen is a lipoxygenase inhibitor. Ebselen has been investigated in clinical trials, where an average administration time of 25 hours post infarct presumably contributed to its lack of efficacy[68]. Other antioxidant 12/15-LOX inhibitors that have shown promise in animal studies include curcumin [69] and the green tea polyphenol epigallocatechin gallate[70]. AA-861, which is not an antioxidant, is an interesting case. Originally introduced as an inhibitor of 5-LOX, it has also been shown to block 12/15-LOX activity. Correspondingly, it protects neuronal cells against oxidative glutamate toxicity, and it also protects rodents against transient focal ischemia[71]. In all of these cases, other protective mechanisms (radical scavenging, general antioxidant effects) are possible, but 12/15-LOX pathology should be considered as injury mechanism.
Endogenous antioxidants
These include the peptide glutathione, as well as several important redox-active proteins. The best known of these is superoxide dismutase. Both the cytosolic (SOD1) and the mitochondrial (SOD2) version of superoxide dismutase protect against ischemic injury by scavenging ROS[72, 73], and in both cases this would be expected to reduce activation of 12/15-LOX. A direct connection between SOD and 12/15-LOX has to date not been made. In contrast, the glutathione peroxidase GPX-4 has been clearly linked to 12/15-LOX dependent cell death[53]; and overexpression of GPX-1 is protective in models of focal ischemia[74]. Another intracellular antioxidant in neurons is Peroxiredoxin-2, which when overexpressed protects against transient focal ischemia via a redox mechanism[75]. Finally, glutathione has long been a neuroprotection target. Because 12/15-LOX is activated when glutathione is depleted, raising glutathione levels should prevent activation of 12/15-LOX and thus, injury caused by 12/15-LOX. This cannot be achieved by glutathione itself, which cannot enter cells. Instead, either derivatives like glutathione methyl ester, or glutathione precursors may be beneficial[76].
The other side of the coin: anti-inflammatory lipoxins and protectins/neuroprotectins
Paradoxically, 12/15-LOX can also generate protective molecules from either arachidonic acid, or the w3-fatty acids eicosapentaenoic and docosahexaenoic acid. In both cases, other enzymes – typically 5-LOX - are involved in the biosynthetic pathway as well. The arachidonic acid derivative lipoxin A4 is well known for its role in inflammation resolution[10, 77]. Lipoxin A4 may contribute to the neuroprotective effects of rosiglitazone through activation of PPARg(Sobrado et al., 2009). Its synthetic methyl ester derivative has been shown to reduce inflammation following transient focal ischemia[78]. Strikingly, an epi-lipoxin with equivalent activity is made when aspirin-treated COX-1 replaces 12/15-LOX.
The laboratories of Nicolas Bazan in New Orleans, and of Charles Serhan in Boston, have demonstrated strong neuroprotective effects of novel docosahexaenoic acid derivatives, termed protectins or neuroprotectins [79–83]. These can be either generated endogenously following infusion of docosahexaenoic acid, or made synthetically and used in a straight-up neuroprotectant approach.
This would pose the dilemma that 12/15-LOX can have both good and bad effects on stroke pathology. At least in experimental stroke studies in rodents to date, the bad clearly outweighs the good, because the net effect of 12/15-LOX inhibition or gene knockout is an improved outcome. Luckily, there may be a way to have the best of both worlds: in a fascinating twist to the story, low-dose aspirin converts COX-2 into a pseudo-lipoxygenase, which generates the R-enantiomers of 12/15-LOX products, which are usually in S-configuration. These R-enantiomers are also active in resolving inflammation[10, 79, 80]. Importantly, the modified COX-2 would not be expected to be able to damage organelles in the way 12/15-LOX does. Therefore, combining low-dose aspirin with a 12/15-LOX inhibitor could provide dual protection, blocking intracellular organelle damage while allowing for the production of anti-inflammatory aspirin-triggered (AT) epi-lipoxin A4 and AT-neuroprotectin D1.
Do other lipoxygenases contribute to stroke injury?
As outlined above, the case for involvement of 12/15-LOX in stroke pathology is strong. But the lipoxygenases are a large family of proteins, with varying tissue distributions and activities[11]. So do other lipoxygenases contribute to ischemic brain injury as well? Presumably not via the same mechanism, because only 12/15-LOX can directly oxidize membrane-bound phospholipids. For example, 5-LOX needs Five-lipoxygenase activating protein FLAP to associate with membranes, but even then a phospholipase A2 is required to liberate polyunsaturated fatty acids from the membrane before they can be oxidized. This may relate to differences in the binding pocket between 5- and 12/15-LOX, which favor head-on insertion for 5-LOX, but tail first engagement for 12/15-LOX. While the attack on mitochondria and other organelles inside the neural cell appears to be restricted to 12/15-LOX, other mechanisms of injury may be elicited by lipoxygenases.
5-LOX
Initially, 5-LOX was a preferred target for stroke therapy, based on its role as generator of the pro-inflammatory leukotrienes. Inhibitors that target 5-LOX directly have been reported to reduce ischemic injury in several models. Nonetheless, 5-LOX knockout mice developed the same infarct size in both transient and permanent focal ischemia as the corresponding wild-type mice [84]. While this result has put a damper on targeting 5-LOX directly, its activating protein FLAP is still being investigated. In rats, the FLAP inhibitor MK-886 decreased leukotriene levels and reduced infarct size[85] 24h after onset of permanent ischemia. It is somewhat unclear how to reconcile these results with one another. In addition, polymorphisms in ALOX5AP (the gene encoding FLAP) have been implicated in stroke risk[86, 87], although this has recently been drawn into question[88]. Similarly, downstream enzymes of the leukotriene pathway have been identified as conferring an increased risk of stroke[89, 90].
12-LOX and other lipoxygenases
Besides 12/15-LOX, there is also a separate 12-LOX enzyme, termed platelet-type 12-LOX[91, 92]. While its distribution in the brain has not been thoroughly studied, it is being investigated as a possible target for anti-platelet therapy[93–95], and this could be beneficial for ischemic stroke. A variety of other lipoxygenases exist, including a 12R-LOX, epidermal LOX-3, and 15-LOX-2, but not much is known about their expression and possible functions in the brain[91].
Other enzymes of the arachidonic acid cascade: phospholipase A2, cyclooxygenase, and cytochrome P450
Besides the lipoxygenase family, other enzymes of the arachidonic acid cascade have also long been implicated in stroke-related brain injury. Phospholipases that liberate arachidonic acid from membrane phospholipids, cyclooxygenases which produce pro-inflammatory prostaglandins, and cytochrome P450 which generates vasoactive 20-HETE, all may contribute to ischemic neurotoxicity. A variety of excellent reviews is available for these subjects, so we will here just very briefly highlight some of the concepts involved.
Phospholipases
because phospholipases, especially of the extended A2 family (PLA2), increase the pool of free poly unsaturated fatty acids following ischemia, their importance was recognized very early[13]. Cytosolic phospholipase A2 (cPLA2) is a type IV phospholipase, and the major arachidonic acid-releasing enzyme in the neuronal cell. It can thus be seen as a master regulator of the arachidonic acid cascade. Besides supplying the arachidonic acid substrate when activated by calcium influx, it is also literally a regulator for the cyclooxygenase COX-2 following ischemia: COX-2 protein is increased two-fold in wild-type, but not in cPLA2 knockout mice following cerebral ischemia[96]. Correspondingly, cPLA2 knockout mice are protected against transient focal ischemia[97]. Either directly or indirectly, cPLA2 also contributes to disruption of the blood – brain barrier[98]. The drawback of being at the initiation step of the arachidonic acid cascade is that it may not be a good drug target. Because cPLA2 modulates early events, the time window for phospholipase inhibition after a stroke may be fairly short[96]. Another phospholipase that has received much attention for its role in atherosclerosis is the lipoprotein-associated phospholipase, Lp-PLA2. In the stroke field, it is being studied mostly for its contributions to stroke risk[99–101]. Isoforms of secretory PLA2 (sPLA2) may contribute to neuronal death[102, 103]. Conversely, another phospholipase PAF-acetylhydrolase II appears to be neuroprotective[104].
Cyclooxygenases
Similar to phospholipases, the cyclooxygenases COX-1 and COX-2 have long been studied for their involvement in stroke. Especially COX-2 is known to be increased, and contribute to injury, in animal models of stroke[105, 106], while COX-1 deficiency actually leads to increased infarct size[107], although recently the case has been made for reinvestigating COX-1 as drug target[108]. In part, COX-2 is modulated by NMDA receptor activity, at the same time it contributes to excitotoxicity[109]. However, because of the finding that COX-2 inhibitors like Vioxx and Bextra increase the risk of cardiovascular events[110, 111], recent research has mostly focused on downstream elements of the COX-2 pathway[112]. Here, especially the Prostaglandin E2 receptors EP1 – EP4 are seen as promising targets, as well as the microsomal Prostaglandin E synthase[113]. Receptors of the D and F series have also been targeted[114, 115].
Cytochrome P450
The Cytochrome P450 family of proteins generates a variety of blood flow regulators, including 20-HETE and EETs. These have been investigated as opportunities to increase blood flow following ischemia[116]. In addition, increasing the levels of EETs by inhibiting soluble epoxide hydrolase (sEH) is being investigated[117].
Summary and Outlook
Several important questions have been raised concerning the feasibility of neuroprotection to combat stroke[118–123]. While not all of these can be answered with theoretical arguments, the basic principle still applies: the injury that can be prevented, does not need to be healed later. Some of the novel approaches presented here, alone or in combination, may provide us with treatments that have proven elusive so far.
So can inhibition of 12/15-LOX be turned into a promising drug approach to treat stroke? Several findings suggest this may be the case. One, low-level expression of the enzyme in the non-ischemic mouse brain, and the relatively normal brain histology of 12/15-LOX knockout mice, both suggest blocking 12/15-LOX activity may not be too disruptive, and thus not saddled with overly severe side effects. Two, the increased expression in both neurons and endothelial cells of the peri-infarct zone, together with the reduction in both infarct size and edema formation through 12/15-LOX inhibition or knockout, show that despite hitting only one target, a suitable 12/15-LOX inhibitor may disrupt several injury pathways in the ischemic brain. Three, the finding of increased 12/15-LOX protein, as well as increased plasma levels of 12-HETE in human stroke patients, indicate that this injury mechanism may also apply to humans. Four, the time window for inhibitor administration appears to be large enough to make 12/15-LOX inhibition clinically relevant, with LOXBlock-1 providing significant neuroprotection when given four hours after onset of ischemia in mice. Although initial studies look promising, effects of 12/15-LOX inhibition on hemorrhage (both in intracerebral hemorrhage models, and in conjunction with tPA) need to be evaluated in more detail. This will help to define the settings in which a 12/15-LOX inhibitor can be employed. If hemorrhagic complications are not worsened, this would make the strategy more widely applicable. Furthermore, the relationship with possible beneficial effects of 12/15-LOX through protectins and similar anti-inflammatory mechanisms needs to be clarified. Finally, studies should be expanded to include aged animals[124], and additional models of stroke and cerebral ischemia in other organisms. Nonetheless, the promising results with 12/15-LOX inhibitors thus far establish that this is a strategy that should be pursued, and hopefully, the current impasse on neuroprotection as stroke treatment strategy can be broken.
Acknowledgments
I thank Dr. Eng Lo for helpful discussions and excellent advice.
List of Abbreviations
- LOX
lipoxygenase (e.g., 12/15-LOX)
- H(P)ETE
hydro(pero)xyeicosatetraenoic acid
- COX
cyclooxygenase
- NSAIDs
non-steroidal anti-inflammatory drugs
- EET
epoxyeicosatrienoic acid
- CSF
cerebrospinal fluid
- AIF
apoptosis-inducing factor
- PUFA
polyunsaturated fatty acid
- ER
endoplasmic reticulum
- ROS
reactive oxygen species
- SOD
superoxide dismutase
- GPX
glutathione peroxidase
- FLAP
five-lipoxygenase activating protein
- cPLA2
cytosolic phospholipase A2
- lp-PLA2
lipoprotein-associated phospholipase A2
Footnotes
Conflict of Interest
Support from the National Institutes of Health (R01NS049430 and R01NS069939) is gratefully acknowledged. A patent on the use of LOXBlock inhibitors to treat stroke has been applied for (U.S. Patent Application No.: 12/671,567).
Literature
- 1.Shuaib A, Lees KR, Lyden P, Grotta J, Davalos A, Davis SM, Diener HC, Ashwood T, Wasiewski WW, Emeribe U. NXY-059 for the treatment of acute ischemic stroke. N Engl J Med. 2007;357:562–71. doi: 10.1056/NEJMoa070240. [DOI] [PubMed] [Google Scholar]
- 2.Moskowitz MA, Lo EH, Iadecola C. The science of stroke: mechanisms in search of treatments. Neuron. 2010;67:181–98. doi: 10.1016/j.neuron.2010.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Zlokovic BV, Griffin JH. Cytoprotective protein C pathways and implications for stroke and neurological disorders. Trends Neurosci. 2011;34:198–209. doi: 10.1016/j.tins.2011.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Quaegebeur A, Lange C, Carmeliet P. The neurovascular link in health and disease: molecular mechanisms and therapeutic implications. Neuron. 2011;71:406–24. doi: 10.1016/j.neuron.2011.07.013. [DOI] [PubMed] [Google Scholar]
- 5.Lau A, Tymianski M. Glutamate receptors, neurotoxicity and neurodegeneration. Pflugers Arch. 2010;460:525–42. doi: 10.1007/s00424-010-0809-1. [DOI] [PubMed] [Google Scholar]
- 6.Besancon E, Guo S, Lok J, Tymianski M, Lo EH. Beyond NMDA and AMPA glutamate receptors: emerging mechanisms for ionic imbalance and cell death in stroke. Trends Pharmacol Sci. 2008;29:268–75. doi: 10.1016/j.tips.2008.02.003. [DOI] [PubMed] [Google Scholar]
- 7.Haeggstrom JZ, Rinaldo-Matthis A, Wheelock CE, Wetterholm A. Advances in eicosanoid research, novel therapeutic implications. Biochem Biophys Res Commun. 2010;396:135–9. doi: 10.1016/j.bbrc.2010.03.140. [DOI] [PubMed] [Google Scholar]
- 8.Nigam S, Zafiriou MP, Deva R, Ciccoli R, Roux-Van der Merwe R. Structure, biochemistry and biology of hepoxilins: an update. FEBS J. 2007;274:3503–12. doi: 10.1111/j.1742-4658.2007.05910.x. [DOI] [PubMed] [Google Scholar]
- 9.Pace-Asciak CR. The hepoxilins and some analogues: a review of their biology. Br J Pharmacol. 2009;158:972–81. doi: 10.1111/j.1476-5381.2009.00168.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Serhan CN. Lipoxins and aspirin-triggered 15-epi-lipoxins are the first lipid mediators of endogenous anti-inflammation and resolution. Prostaglandins Leukot Essent Fatty Acids. 2005;73:141–62. doi: 10.1016/j.plefa.2005.05.002. [DOI] [PubMed] [Google Scholar]
- 11.Brash AR. Lipoxygenases: occurrence, functions, catalysis, and acquisition of substrate. J Biol Chem. 1999;274:23679–82. doi: 10.1074/jbc.274.34.23679. [DOI] [PubMed] [Google Scholar]
- 12.Lo EH, Moskowitz MA, Jacobs TP. Exciting, radical, suicidal: how brain cells die after stroke. Stroke. 2005;36:189–92. doi: 10.1161/01.STR.0000153069.96296.fd. [DOI] [PubMed] [Google Scholar]
- 13.Bazan NG., Jr Effects of ischemia and electroconvulsive shock on free fatty acid pool in the brain. Biochim Biophys Acta. 1970;218:1–10. doi: 10.1016/0005-2760(70)90086-x. [DOI] [PubMed] [Google Scholar]
- 14.Chan PH, Fishman RA. Brain edema: induction in cortical slices by polyunsaturated fatty acids. Science. 1978;201:358–60. doi: 10.1126/science.663662. [DOI] [PubMed] [Google Scholar]
- 15.Chan PH, Fishman RA, Caronna J, Schmidley JW, Prioleau G, Lee J. Induction of brain edema following intracerebral injection of arachidonic acid. Ann Neurol. 1983;13:625–32. doi: 10.1002/ana.410130608. [DOI] [PubMed] [Google Scholar]
- 16.Moskowitz MA, Kiwak KJ, Hekimian K, Levine L. Synthesis of compounds with properties of leukotrienes C4 and D4 in gerbil brains after ischemia and reperfusion. Science. 1984;224:886–9. doi: 10.1126/science.6719118. [DOI] [PubMed] [Google Scholar]
- 17.Li Y, Maher P, Schubert D. A role for 12-lipoxygenase in nerve cell death caused by glutathione depletion. Neuron. 1997;19:453–63. doi: 10.1016/s0896-6273(00)80953-8. [DOI] [PubMed] [Google Scholar]
- 18.van Leyen K, Lee SR, Siddiq A, Ratan RR, Lo EH. Program No 1356, 2004 Abstract Viewer/Itinerary Planner. Washington, DC: Society for Neuroscience; 2004. 12/15-lipoxygenase and the proteasome as mediators of neuronal oxidative stress and stroke. [Google Scholar]
- 19.Khanna S, Roy S, Slivka A, Craft TK, Chaki S, Rink C, Notestine MA, DeVries AC, Parinandi NL, Sen CK. Neuroprotective properties of the natural vitamin E alpha-tocotrienol. Stroke. 2005;36:2258–64. doi: 10.1161/01.STR.0000181082.70763.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.van Leyen K, Kim HY, Lee SR, Jin G, Arai K, Lo EH. Baicalein and 12/15-lipoxygenase in the ischemic brain. Stroke. 2006;37:3014–8. doi: 10.1161/01.STR.0000249004.25444.a5. [DOI] [PubMed] [Google Scholar]
- 21.Poloyac SM, Reynolds RB, Yonas H, Kerr ME. Identification and quantification of the hydroxyeicosatetraenoic acids, 20-HETE and 12-HETE, in the cerebrospinal fluid after subarachnoid hemorrhage. J Neurosci Methods. 2005;144:257–63. doi: 10.1016/j.jneumeth.2004.11.015. [DOI] [PubMed] [Google Scholar]
- 22.Farias SE, Heidenreich KA, Wohlauer MV, Murphy RC, Moore EE. Lipid mediators in cerebral spinal fluid of traumatic brain injured patients. J Trauma. 2011;71:1211–8. doi: 10.1097/TA.0b013e3182092c62. [DOI] [PubMed] [Google Scholar]
- 23.Yigitkanli K, Pekcec A, Karatas H, Pallast S, Mandeville E, Joshi N, Smirnova N, Gazaryan I, Ratan RR, Witztum JL, Montaner J, Holman TR, Lo EH, van Leyen K. Inhibition of 12/15-Lipoxygenase as Therapeutic Strategy to Treat Stroke. Ann Neurol. 2013;73:129–135. doi: 10.1002/ana.23734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Pallast S, Arai K, Pekcec A, Yigitkanli K, Yu Z, Wang X, Lo EH, van Leyen K. Increased nuclear apoptosis-inducing factor after transient focal ischemia: a 12/15-lipoxygenase-dependent organelle damage pathway. J Cereb Blood Flow Metab. 2010;30:1157–67. doi: 10.1038/jcbfm.2009.281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Jin G, Arai K, Murata Y, Wang S, Stins MF, Lo EH, van Leyen K. Protecting against cerebrovascular injury: contributions of 12/15-lipoxygenase to edema formation after transient focal ischemia. Stroke. 2008;39:2538–43. doi: 10.1161/STROKEAHA.108.514927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Schewe T, Rapoport SM, Kuhn H. Enzymology and physiology of reticulocyte lipoxygenase: comparison with other lipoxygenases. Adv Enzymol Relat Areas Mol Biol. 1986;58:191–272. doi: 10.1002/9780470123041.ch6. [DOI] [PubMed] [Google Scholar]
- 27.Ivanov I, Heydeck D, Hofheinz K, Roffeis J, O’Donnell VB, Kuhn H, Walther M. Molecular enzymology of lipoxygenases. Arch Biochem Biophys. 2010;503:161–74. doi: 10.1016/j.abb.2010.08.016. [DOI] [PubMed] [Google Scholar]
- 28.Maccarrone M, Melino G, Finazzi-Agro A. Lipoxygenases and their involvement in programmed cell death. Cell Death Differ. 2001;8:776–84. doi: 10.1038/sj.cdd.4400908. [DOI] [PubMed] [Google Scholar]
- 29.van Leyen K, Duvoisin RM, Engelhardt H, Wiedmann M. A function for lipoxygenase in programmed organelle degradation. Nature. 1998;395:392–5. doi: 10.1038/26500. [DOI] [PubMed] [Google Scholar]
- 30.van Leyen K. Pictures in cell biology Un-LOXing the door to organelle degradation. Trends Cell Biol. 1998;8:436. doi: 10.1016/s0962-8924(98)01386-5. [DOI] [PubMed] [Google Scholar]
- 31.Grüllich C, Duvoisin RM, Wiedmann M, van Leyen K. Inhibition of 15-lipoxygenase leads to delayed organelle degradation in the reticulocyte. FEBS Lett. 2001;489:51–4. doi: 10.1016/s0014-5793(01)02080-4. [DOI] [PubMed] [Google Scholar]
- 32.Schewe T, Halangk W, Hiebsch C, Rapoport SM. A lipoxygenase in rabbit reticulocytes which attacks phospholipids and intact mitochondria. FEBS Lett. 1975;60:149–52. doi: 10.1016/0014-5793(75)80439-x. [DOI] [PubMed] [Google Scholar]
- 33.Bassnett S. On the mechanism of organelle degradation in the vertebrate lens. Exp Eye Res. 2009;88:133–9. doi: 10.1016/j.exer.2008.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Dahm R. Dying to see. Sci Am. 2004;291:82–9. doi: 10.1038/scientificamerican1004-82. [DOI] [PubMed] [Google Scholar]
- 35.Conrad DJ, Lu M. Regulation of human 12/15-lipoxygenase by Stat6-dependent transcription. Am J Respir Cell Mol Biol. 2000;22:226–34. doi: 10.1165/ajrcmb.22.2.3786. [DOI] [PubMed] [Google Scholar]
- 36.Huang JT, Welch JS, Ricote M, Binder CJ, Willson TM, Kelly C, Witztum JL, Funk CD, Conrad D, Glass CK. Interleukin-4-dependent production of PPAR-gamma ligands in macrophages by 12/15-lipoxygenase. Nature. 1999;400:378–82. doi: 10.1038/22572. [DOI] [PubMed] [Google Scholar]
- 37.Shureiqi I, Zuo X, Broaddus R, Wu Y, Guan B, Morris JS, Lippman SM. The transcription factor GATA-6 is overexpressed in vivo and contributes to silencing 15-LOX-1 in vitro in human colon cancer. FASEB J. 2007;21:743–53. doi: 10.1096/fj.06-6830com. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ostareck DH, Ostareck-Lederer A, Shatsky IN, Hentze MW. Lipoxygenase mRNA silencing in erythroid differentiation: The 3′UTR regulatory complex controls 60S ribosomal subunit joining. Cell. 2001;104:281–90. doi: 10.1016/s0092-8674(01)00212-4. [DOI] [PubMed] [Google Scholar]
- 39.Ostareck DH, Ostareck-Lederer A, Wilm M, Thiele BJ, Mann M, Hentze MW. mRNA silencing in erythroid differentiation: hnRNP K and hnRNP E1 regulate 15-lipoxygenase translation from the 3′ end. Cell. 1997;89:597–606. doi: 10.1016/s0092-8674(00)80241-x. [DOI] [PubMed] [Google Scholar]
- 40.Brinckmann R, Schnurr K, Heydeck D, Rosenbach T, Kolde G, Kuhn H. Membrane translocation of 15-lipoxygenase in hematopoietic cells is calcium-dependent and activates the oxygenase activity of the enzyme. Blood. 1998;91:64–74. [PubMed] [Google Scholar]
- 41.Imai H, Nakagawa Y. Biological significance of phospholipid hydroperoxide glutathione peroxidase (PHGPx, GPx4) in mammalian cells. Free Radic Biol Med. 2003;34:145–69. doi: 10.1016/s0891-5849(02)01197-8. [DOI] [PubMed] [Google Scholar]
- 42.Uemura Y, Miller JM, Matson WR, Beal MF. Neurochemical analysis of focal ischemia in rats. Stroke. 1991;22:1548–53. doi: 10.1161/01.str.22.12.1548. [DOI] [PubMed] [Google Scholar]
- 43.Bano D, Nicotera P. Ca2+ signals and neuronal death in brain ischemia. Stroke. 2007;38:674–6. doi: 10.1161/01.STR.0000256294.46009.29. [DOI] [PubMed] [Google Scholar]
- 44.Murphy TH, Schnaar RL, Coyle JT. Immature cortical neurons are uniquely sensitive to glutamate toxicity by inhibition of cystine uptake. FASEB J. 1990;4:1624–33. [PubMed] [Google Scholar]
- 45.Ratan RR, Murphy TH, Baraban JM. Oxidative stress induces apoptosis in embryonic cortical neurons. J Neurochem. 1994;62:376–9. doi: 10.1046/j.1471-4159.1994.62010376.x. [DOI] [PubMed] [Google Scholar]
- 46.Ratan RR, Ryu H, Lee J, Mwidau A, Neve RL. In vitro model of oxidative stress in cortical neurons. Methods Enzymol. 2002;352:183–90. doi: 10.1016/s0076-6879(02)52018-8. [DOI] [PubMed] [Google Scholar]
- 47.Wang H, Li J, Follett PL, Zhang Y, Cotanche DA, Jensen FE, Volpe JJ, Rosenberg PA. 12-Lipoxygenase plays a key role in cell death caused by glutathione depletion and arachidonic acid in rat oligodendrocytes. Eur J Neurosci. 2004;20:2049–58. doi: 10.1111/j.1460-9568.2004.03650.x. [DOI] [PubMed] [Google Scholar]
- 48.Zhang Y, Aizenman E, DeFranco DB, Rosenberg PA. Intracellular zinc release, 12-lipoxygenase activation and MAPK dependent neuronal and oligodendroglial death. Mol Med. 2007;13:350–5. doi: 10.2119/2007-00042.Zhang. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.van Leyen K, Arai K, Jin G, Kenyon V, Gerstner B, Rosenberg PA, Holman TR, Lo EH. Novel lipoxygenase inhibitors as neuroprotective reagents. J Neurosci Res. 2008;86:904–9. doi: 10.1002/jnr.21543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Pallast S, Arai K, Wang X, Lo EH, van Leyen K. 12/15-Lipoxygenase targets neuronal mitochondria under oxidative stress. J Neurochem. 2009;111:882–9. doi: 10.1111/j.1471-4159.2009.06379.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.van Leyen K, Siddiq A, Ratan RR, Lo EH. Proteasome inhibition protects HT22 neuronal cells from oxidative glutamate toxicity. J Neurochem. 2005;92:824–30. doi: 10.1111/j.1471-4159.2004.02915.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Tobaben S, Grohm J, Seiler A, Conrad M, Plesnila N, Culmsee C. Bid-mediated mitochondrial damage is a key mechanism in glutamate-induced oxidative stress and AIF-dependent cell death in immortalized HT-22 hippocampal neurons. Cell Death Differ. 2011;18:282–92. doi: 10.1038/cdd.2010.92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Seiler A, Schneider M, Forster H, Roth S, Wirth EK, Culmsee C, Plesnila N, Kremmer E, Radmark O, Wurst W, Bornkamm GW, Schweizer U, Conrad M. Glutathione peroxidase 4 senses and translates oxidative stress into 12/15-lipoxygenase dependent- and AIF-mediated cell death. Cell Metab. 2008;8:237–48. doi: 10.1016/j.cmet.2008.07.005. [DOI] [PubMed] [Google Scholar]
- 54.Huang HS, Chen CJ, Suzuki H, Yamamoto S, Chang WC. Inhibitory effect of phospholipid hydroperoxide glutathione peroxidase on the activity of lipoxygenases and cyclooxygenases. Prostaglandins Other Lipid Mediat. 1999;58:65–75. doi: 10.1016/s0090-6980(99)00017-9. [DOI] [PubMed] [Google Scholar]
- 55.Khanna S, Roy S, Parinandi NL, Maurer M, Sen CK. Characterization of the potent neuroprotective properties of the natural vitamin E alpha-tocotrienol. J Neurochem. 2006;98:1474–86. doi: 10.1111/j.1471-4159.2006.04000.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Khanna S, Roy S, Ryu H, Bahadduri P, Swaan PW, Ratan RR, Sen CK. Molecular basis of vitamin E action: tocotrienol modulates 12-lipoxygenase, a key mediator of glutamate-induced neurodegeneration. J Biol Chem. 2003;278:43508–15. doi: 10.1074/jbc.M307075200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Rink C, Christoforidis G, Khanna S, Peterson L, Patel Y, Abduljalil A, Irfanoglu O, Machiraju R, Bergdall VK, Sen CK. Tocotrienol vitamin E protects against preclinical canine ischemic stroke by inducing arteriogenesis. J Cereb Blood Flow Metab. 2011;31:2218–30. doi: 10.1038/jcbfm.2011.85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Park HA, Kubicki N, Gnyawali S, Chan YC, Roy S, Khanna S, Sen CK. Natural vitamin E alpha-tocotrienol protects against ischemic stroke by induction of multidrug resistance-associated protein 1. Stroke. 2011;42:2308–14. doi: 10.1161/STROKEAHA.110.608547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Huang Y, Tsang SY, Yao X, Chen ZY. Biological properties of baicalein in cardiovascular system. Curr Drug Targets Cardiovasc Haematol Disord. 2005;5:177–84. doi: 10.2174/1568006043586206. [DOI] [PubMed] [Google Scholar]
- 60.Deschamps JD, Kenyon VA, Holman TR. Baicalein is a potent in vitro inhibitor against both reticulocyte 15-human and platelet 12-human lipoxygenases. Bioorg Med Chem. 2006;14:4295–301. doi: 10.1016/j.bmc.2006.01.057. [DOI] [PubMed] [Google Scholar]
- 61.Lapchak PA, Maher P, Schubert D, Zivin JA. Baicalein, an antioxidant 12/15-lipoxygenase inhibitor improves clinical rating scores following multiple infarct embolic strokes. Neuroscience. 2007;150:585–91. doi: 10.1016/j.neuroscience.2007.09.033. [DOI] [PubMed] [Google Scholar]
- 62.Xu YW, Sun L, Liang H, Sun GM, Cheng Y. 12/15-Lipoxygenase inhibitor baicalein suppresses PPAR gamma expression and nuclear translocation induced by cerebral ischemia/reperfusion. Brain Res. 2010;1307:149–57. doi: 10.1016/j.brainres.2009.10.038. [DOI] [PubMed] [Google Scholar]
- 63.Hwang YS, Shin CY, Huh Y, Ryu JH. Hwangryun-Hae-Dok-tang (Huanglian-Jie-Du-Tang) extract and its constituents reduce ischemia-reperfusion brain injury and neutrophil infiltration in rats. Life Sci. 2002;71:2105–17. doi: 10.1016/s0024-3205(02)01920-3. [DOI] [PubMed] [Google Scholar]
- 64.Kenyon V, Chorny I, Carvajal WJ, Holman TR, Jacobson MP. Novel human lipoxygenase inhibitors discovered using virtual screening with homology models. J Med Chem. 2006;49:1356–63. doi: 10.1021/jm050639j. [DOI] [PubMed] [Google Scholar]
- 65.Yoshida H, Yanai H, Namiki Y, Fukatsu-Sasaki K, Furutani N, Tada N. Neuroprotective effects of edaravone: a novel free radical scavenger in cerebrovascular injury. CNS Drug Rev. 2006;12:9–20. doi: 10.1111/j.1527-3458.2006.00009.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Lee BJ, Egi Y, van Leyen K, Lo EH, Arai K. Edaravone, a free radical scavenger, protects components of the neurovascular unit against oxidative stress in vitro. Brain Res. 2010;1307:22–7. doi: 10.1016/j.brainres.2009.10.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Abe K, Yuki S, Kogure K. Strong attenuation of ischemic and postischemic brain edema in rats by a novel free radical scavenger. Stroke. 1988;19:480–5. doi: 10.1161/01.str.19.4.480. [DOI] [PubMed] [Google Scholar]
- 68.Yamaguchi T, Sano K, Takakura K, Saito I, Shinohara Y, Asano T, Yasuhara H. Ebselen in acute ischemic stroke: a placebo-controlled, double-blind clinical trial. Ebselen Study Group. Stroke. 1998;29:12–7. doi: 10.1161/01.str.29.1.12. [DOI] [PubMed] [Google Scholar]
- 69.Thiyagarajan M, Sharma SS. Neuroprotective effect of curcumin in middle cerebral artery occlusion induced focal cerebral ischemia in rats. Life Sci. 2004;74:969–85. doi: 10.1016/j.lfs.2003.06.042. [DOI] [PubMed] [Google Scholar]
- 70.Lee SY, Kim CY, Lee JJ, Jung JG, Lee SR. Effects of delayed administration of (−)-epigallocatechin gallate, a green tea polyphenol on the changes in polyamine levels and neuronal damage after transient forebrain ischemia in gerbils. Brain Res Bull. 2003;61:399–406. doi: 10.1016/s0361-9230(03)00139-4. [DOI] [PubMed] [Google Scholar]
- 71.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;85:112–6. doi: 10.3171/jns.1996.85.1.0112. [DOI] [PubMed] [Google Scholar]
- 72.Maier CM, Chan PH. Role of superoxide dismutases in oxidative damage and neurodegenerative disorders. Neuroscientist. 2002;8:323–34. doi: 10.1177/107385840200800408. [DOI] [PubMed] [Google Scholar]
- 73.Jung JE, Kim GS, Chen H, Maier CM, Narasimhan P, Song YS, Niizuma K, Katsu M, Okami N, Yoshioka H, Sakata H, Goeders CE, Chan PH. Reperfusion and neurovascular dysfunction in stroke: from basic mechanisms to potential strategies for neuroprotection. Mol Neurobiol. 2010;41:172–9. doi: 10.1007/s12035-010-8102-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Weisbrot-Lefkowitz M, Reuhl K, Perry B, Chan PH, Inouye M, Mirochnitchenko O. Overexpression of human glutathione peroxidase protects transgenic mice against focal cerebral ischemia/reperfusion damage. Brain Res Mol Brain Res. 1998;53:333–8. doi: 10.1016/s0169-328x(97)00313-6. [DOI] [PubMed] [Google Scholar]
- 75.Gan Y, Ji X, Hu X, Luo Y, Zhang L, Li P, Liu X, Yan F, Vosler P, Gao Y, Stetler RA, Chen J. Transgenic overexpression of peroxiredoxin-2 attenuates ischemic neuronal injury via suppression of a redox-sensitive pro-death signaling pathway. Antioxid Redox Signal. 2012;17:719–32. doi: 10.1089/ars.2011.4298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Anderson MF, Nilsson M, Eriksson PS, Sims NR. Glutathione monoethyl ester provides neuroprotection in a rat model of stroke. Neurosci Lett. 2004;354:163–5. doi: 10.1016/j.neulet.2003.09.067. [DOI] [PubMed] [Google Scholar]
- 77.Serhan CN, Yacoubian S, Yang R. Anti-inflammatory and proresolving lipid mediators. Annu Rev Pathol. 2008;3:279–312. doi: 10.1146/annurev.pathmechdis.3.121806.151409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Ye XH, Wu Y, Guo PP, Wang J, Yuan SY, Shang Y, Yao SL. Lipoxin A4 analogue protects brain and reduces inflammation in a rat model of focal cerebral ischemia reperfusion. Brain Res. 2010;1323:174–83. doi: 10.1016/j.brainres.2010.01.079. [DOI] [PubMed] [Google Scholar]
- 79.Bazan NG, Eady TN, Khoutorova L, Atkins KD, Hong S, Lu Y, Zhang C, Jun B, Obenaus A, Fredman G, Zhu M, Winkler JW, Petasis NA, Serhan CN, Belayev L. Novel aspirin-triggered neuroprotectin D1 attenuates cerebral ischemic injury after experimental stroke. Exp Neurol. 2012;236:122–30. doi: 10.1016/j.expneurol.2012.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Serhan CN, Fredman G, Yang R, Karamnov S, Belayev LS, Bazan NG, Zhu M, Winkler JW, Petasis NA. Novel proresolving aspirin-triggered DHA pathway. Chem Biol. 2011;18:976–87. doi: 10.1016/j.chembiol.2011.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Belayev L, Khoutorova L, Atkins KD, Eady TN, Hong S, Lu Y, Obenaus A, Bazan NG. Docosahexaenoic Acid Therapy of Experimental Ischemic Stroke. Transl Stroke Res. 2011;2:33–41. doi: 10.1007/s12975-010-0046-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Belayev L, Khoutorova L, Atkins KD, Bazan NG. Robust docosahexaenoic acid-mediated neuroprotection in a rat model of transient, focal cerebral ischemia. Stroke. 2009;40:3121–6. doi: 10.1161/STROKEAHA.109.555979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Marcheselli VL, Hong S, Lukiw WJ, Tian XH, Gronert K, Musto A, Hardy M, Gimenez JM, Chiang N, Serhan CN, Bazan NG. Novel docosanoids inhibit brain ischemia-reperfusion-mediated leukocyte infiltration and pro-inflammatory gene expression. J Biol Chem. 2003;278:43807–17. doi: 10.1074/jbc.M305841200. [DOI] [PubMed] [Google Scholar]
- 84.Kitagawa K, Matsumoto M, Hori M. Cerebral ischemia in 5-lipoxygenase knockout mice. Brain Res. 2004;1004:198–202. doi: 10.1016/j.brainres.2004.01.018. [DOI] [PubMed] [Google Scholar]
- 85.Ciceri P, Rabuffetti M, Monopoli A, Nicosia S. Production of leukotrienes in a model of focal cerebral ischaemia in the rat. Br J Pharmacol. 2001;133:1323–9. doi: 10.1038/sj.bjp.0704189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Helgadottir A, Manolescu A, Thorleifsson G, Gretarsdottir S, Jonsdottir H, Thorsteinsdottir U, Samani NJ, Gudmundsson G, Grant SF, Thorgeirsson G, Sveinbjornsdottir S, Valdimarsson EM, Matthiasson SE, Johannsson H, Gudmundsdottir O, Gurney ME, Sainz J, Thorhallsdottir M, Andresdottir M, Frigge ML, Topol EJ, Kong A, Gudnason V, Hakonarson H, Gulcher JR, Stefansson K. The gene encoding 5-lipoxygenase activating protein confers risk of myocardial infarction and stroke. Nat Genet. 2004;36:233–9. doi: 10.1038/ng1311. [DOI] [PubMed] [Google Scholar]
- 87.Domingues-Montanari S, Fernandez-Cadenas I, del Rio-Espinola A, Corbeto N, Krug T, Manso H, Gouveia L, Sobral J, Mendioroz M, Fernandez-Morales J, Alvarez-Sabin J, Ribo M, Rubiera M, Obach V, Marti-Fabregas J, Freijo M, Serena J, Ferro JM, Vicente AM, Oliveira SA, Montaner J. Association of a genetic variant in the ALOX5AP with higher risk of ischemic stroke: a case-control, meta-analysis and functional study. Cerebrovasc Dis. 2010;29:528–37. doi: 10.1159/000302738. [DOI] [PubMed] [Google Scholar]
- 88.Barral S, Fernandez-Cadenas I, Bis JC, Montaner J, Ikram AM, Launer LJ, Fornage M, Schmidt H, Brickman AM, Seshadri S, Mayeux R. No association of ALOX5AP polymorphisms with risk of MRI-defined brain infarcts. Neurobiol Aging. 2012;33:629, e1–3. doi: 10.1016/j.neurobiolaging.2011.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Bevan S, Dichgans M, Wiechmann HE, Gschwendtner A, Meitinger T, Markus HS. Genetic variation in members of the leukotriene biosynthesis pathway confer an increased risk of ischemic stroke: a replication study in two independent populations. Stroke. 2008;39:1109–14. doi: 10.1161/STROKEAHA.107.491969. [DOI] [PubMed] [Google Scholar]
- 90.Helgadottir A, Manolescu A, Helgason A, Thorleifsson G, Thorsteinsdottir U, Gudbjartsson DF, Gretarsdottir S, Magnusson KP, Gudmundsson G, Hicks A, Jonsson T, Grant SF, Sainz J, O’Brien SJ, Sveinbjornsdottir S, Valdimarsson EM, Matthiasson SE, Levey AI, Abramson JL, Reilly MP, Vaccarino V, Wolfe ML, Gudnason V, Quyyumi AA, Topol EJ, Rader DJ, Thorgeirsson G, Gulcher JR, Hakonarson H, Kong A, Stefansson K. A variant of the gene encoding leukotriene A4 hydrolase confers ethnicity-specific risk of myocardial infarction. Nat Genet. 2006;38:68–74. doi: 10.1038/ng1692. [DOI] [PubMed] [Google Scholar]
- 91.Schneider C, Strayhorn WD, Brantley DM, Nanney LB, Yull FE, Brash AR. Upregulation of 8-lipoxygenase in the dermatitis of IkappaB-alpha-deficient mice. J Invest Dermatol. 2004;122:691–8. doi: 10.1111/j.0022-202X.2004.22329.x. [DOI] [PubMed] [Google Scholar]
- 92.Funk CD, Chen XS, Johnson EN, Zhao L. Lipoxygenase genes and their targeted disruption. Prostaglandins Other Lipid Mediat. 2002;68–69:303–12. doi: 10.1016/s0090-6980(02)00036-9. [DOI] [PubMed] [Google Scholar]
- 93.Yeung J, Holinstat M. 12-lipoxygenase: a potential target for novel anti-platelet therapeutics. Cardiovasc Hematol Agents Med Chem. 2011;9:154–64. doi: 10.2174/187152511797037619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Deschamps JD, Gautschi JT, Whitman S, Johnson TA, Gassner NC, Crews P, Holman TR. Discovery of platelet-type 12-human lipoxygenase selective inhibitors by high-throughput screening of structurally diverse libraries. Bioorg Med Chem. 2007;15:6900–8. doi: 10.1016/j.bmc.2007.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Yeung J, Apopa PL, Vesci J, Kenyon V, Rai G, Jadhav A, Simeonov A, Holman TR, Maloney DJ, Boutaud O, Holinstat M. Protein kinase C regulation of 12-lipoxygenase-mediated human platelet activation. Mol Pharmacol. 2012;81:420–30. doi: 10.1124/mol.111.075630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Kishimoto K, Li RC, Zhang J, Klaus JA, Kibler KK, Dore S, Koehler RC, Sapirstein A. Cytosolic phospholipase A2 alpha amplifies early cyclooxygenase-2 expression, oxidative stress and MAP kinase phosphorylation after cerebral ischemia in mice. J Neuroinflammation. 2010;7:42. doi: 10.1186/1742-2094-7-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.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;390:622–5. doi: 10.1038/37635. [DOI] [PubMed] [Google Scholar]
- 98.Nito C, Kamada H, Endo H, Niizuma K, Myer DJ, Chan PH. Role of the p38 mitogen-activated protein kinase/cytosolic phospholipase A2 signaling pathway in blood-brain barrier disruption after focal cerebral ischemia and reperfusion. J Cereb Blood Flow Metab. 2008;28:1686–96. doi: 10.1038/jcbfm.2008.60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Massot A, Pelegri D, Penalba A, Arenillas J, Boada C, Giralt D, Ribo M, Molina CA, Rosell A, Alvarez-Sabin J, Chacon P, Rovira A, Delgado P, Montaner J. Lipoprotein-associated phospholipase A2 testing usefulness among patients with symptomatic intracranial atherosclerotic disease. Atherosclerosis. 2011;218:181–7. doi: 10.1016/j.atherosclerosis.2011.04.031. [DOI] [PubMed] [Google Scholar]
- 100.Gorelick PB. Lipoprotein-associated phospholipase A2 and risk of stroke. Am J Cardiol. 2008;101:34F–40F. doi: 10.1016/j.amjcard.2008.04.017. [DOI] [PubMed] [Google Scholar]
- 101.Elkind MS. Inflammatory markers and stroke. Curr Cardiol Rep. 2009;11:12–20. doi: 10.1007/s11886-009-0003-2. [DOI] [PubMed] [Google Scholar]
- 102.Muralikrishna Adibhatla R, Hatcher JF. Phospholipase A2, reactive oxygen species, and lipid peroxidation in cerebral ischemia. Free Radic Biol Med. 2006;40:376–87. doi: 10.1016/j.freeradbiomed.2005.08.044. [DOI] [PubMed] [Google Scholar]
- 103.Kolko M, de Turco EB, Diemer NH, Bazan NG. Secretory phospholipase A2-mediated neuronal cell death involves glutamate ionotropic receptors. Neuroreport. 2002;13:1963–6. doi: 10.1097/00001756-200210280-00026. [DOI] [PubMed] [Google Scholar]
- 104.Umemura K, Kato I, Hirashima Y, Ishii Y, Inoue T, Aoki J, Kono N, Oya T, Hayashi N, Hamada H, Endo S, Oda M, Arai H, Kinouchi H, Hiraga K. Neuroprotective role of transgenic PAF-acetylhydrolase II in mouse models of focal cerebral ischemia. Stroke. 2007;38:1063–8. doi: 10.1161/01.STR.0000257981.09329.d2. [DOI] [PubMed] [Google Scholar]
- 105.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;98:1294–9. doi: 10.1073/pnas.98.3.1294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Candelario-Jalil E. Nimesulide as a promising neuroprotectant in brain ischemia: new experimental evidences. Pharmacol Res. 2008;57:266–73. doi: 10.1016/j.phrs.2008.03.003. [DOI] [PubMed] [Google Scholar]
- 107.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;21:1436–41. doi: 10.1097/00004647-200112000-00008. [DOI] [PubMed] [Google Scholar]
- 108.Aid S, Bosetti F. Targeting cyclooxygenases-1 and -2 in neuroinflammation: Therapeutic implications. Biochimie. 2011;93:46–51. doi: 10.1016/j.biochi.2010.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Stark DT, Bazan NG. Synaptic and extrasynaptic NMDA receptors differentially modulate neuronal cyclooxygenase-2 function, lipid peroxidation, and neuroprotection. J Neurosci. 2011;31:13710–21. doi: 10.1523/JNEUROSCI.3544-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Grosser T, Yu Y, Fitzgerald GA. Emotion recollected in tranquility: lessons learned from the COX-2 saga. Annu Rev Med. 2010;61:17–33. doi: 10.1146/annurev-med-011209-153129. [DOI] [PubMed] [Google Scholar]
- 111.Cannon CP, Cannon PJ. Physiology COX-2 inhibitors and cardiovascular risk. Science. 2012;336:1386–7. doi: 10.1126/science.1224398. [DOI] [PubMed] [Google Scholar]
- 112.Iadecola C, Gorelick PB. The Janus face of cyclooxygenase-2 in ischemic stroke: shifting toward downstream targets. Stroke. 2005;36:182–5. doi: 10.1161/01.STR.0000153797.33611.d8. [DOI] [PubMed] [Google Scholar]
- 113.Andreasson K. Emerging roles of PGE2 receptors in models of neurological disease. Prostaglandins Other Lipid Mediat. 2010;91:104–12. doi: 10.1016/j.prostaglandins.2009.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Kim YT, Moon SK, Maruyama T, Narumiya S, Dore S. Prostaglandin FP receptor inhibitor reduces ischemic brain damage and neurotoxicity. Neurobiol Dis. 2012;48:58–65. doi: 10.1016/j.nbd.2012.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Ahmad AS, Ahmad M, Maruyama T, Narumiya S, Dore S. Prostaglandin D2 DP1 receptor is beneficial in ischemic stroke and in acute exicitotoxicity in young and old mice. Age (Dordr) 2010;32:271–82. doi: 10.1007/s11357-010-9135-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Liu M, Hurn PD, Alkayed NJ. Cytochrome P450 in neurological disease. Curr Drug Metab. 2004;5:225–34. doi: 10.2174/1389200043335540. [DOI] [PubMed] [Google Scholar]
- 117.Imig JD, Simpkins AN, Renic M, Harder DR. Cytochrome P450 eicosanoids and cerebral vascular function. Expert Rev Mol Med. 2011;13:e7. doi: 10.1017/S1462399411001773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.O’Collins VE, Macleod MR, Donnan GA, Horky LL, van der Worp BH, Howells DW. 1,026 experimental treatments in acute stroke. Ann Neurol. 2006;59:467–77. doi: 10.1002/ana.20741. [DOI] [PubMed] [Google Scholar]
- 119.Donnan GA, Davis SM. Neuroprotection: still achievable in humans. Stroke. 2008;39:525. doi: 10.1161/STROKEAHA.107.494807. [DOI] [PubMed] [Google Scholar]
- 120.Savitz SI, Schabitz WR. A Critique of SAINT II: wishful thinking, dashed hopes, and the future of neuroprotection for acute stroke. Stroke. 2008;39:1389–91. doi: 10.1161/STROKEAHA.107.504415. [DOI] [PubMed] [Google Scholar]
- 121.Ginsberg MD. Neuroprotection for ischemic stroke: past, present and future. Neuropharmacology. 2008;55:363–89. doi: 10.1016/j.neuropharm.2007.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Hussain MS, Shuaib A. Research into neuroprotection must continue. But with a different approach. Stroke. 2008;39:521–2. doi: 10.1161/STROKEAHA.107.494781. [DOI] [PubMed] [Google Scholar]
- 123.Rother J. Neuroprotection does not work! Stroke. 2008;39:523–4. doi: 10.1161/STROKEAHA.107.494799. [DOI] [PubMed] [Google Scholar]
- 124.Chen RL, Balami JS, Esiri MM, Chen LK, Buchan AM. Ischemic stroke in the elderly: an overview of evidence. Nat Rev Neurol. 2010;6:256–65. doi: 10.1038/nrneurol.2010.36. [DOI] [PubMed] [Google Scholar]