Skip to main content
Antioxidants & Redox Signaling logoLink to Antioxidants & Redox Signaling
. 2015 Jan 10;22(2):161–174. doi: 10.1089/ars.2013.5767

NADPH Oxidase-2: Linking Glucose, Acidosis, and Excitotoxicity in Stroke

Angela M Brennan-Minnella 1,,2, Seok Joon Won 1,,2, Raymond A Swanson 1,,2,
PMCID: PMC4281853  PMID: 24628477

Abstract

Significance: Neuronal superoxide production contributes to cell death in both glutamate excitotoxicity and brain ischemia (stroke). NADPH oxidase-2 (NOX2) is the major source of neuronal superoxide production in these settings, and regulation of NOX2 activity can thereby influence outcome in stroke. Recent Advances: Reduced NOX2 activity can rescue cells from oxidative stress and cell death that otherwise occur in excitotoxicity and ischemia. NOX2 activity is regulated by several factors previously shown to affect outcome in stroke, including glucose availability, intracellular pH, protein kinase ζ/δ, casein kinase 2, phosphoinositide-3-kinase, Rac1/2, and phospholipase A2. The newly identified functions of these factors as regulators of NOX2 activity suggest alternative mechanisms for their effects on ischemic brain injury. Critical Issues: Key aspects of these regulatory influences remain unresolved, including the mechanisms by which rac1 and phospholipase activities are coupled to N-methyl-D-aspartate (NMDA) receptors, and whether superoxide production by NOX2 triggers subsequent superoxide production by mitochondria. Future Directions: It will be important to establish whether interventions targeting the signaling pathways linking NMDA receptors to NOX2 in brain ischemia can provide a greater neuroprotective efficacy or a longer time window to treatment than provided by NMDA receptor blockade alone. It will likewise be important to determine whether dissociating superoxide production from the other signaling events initiated by NMDA receptors can mitigate the deleterious effects of NMDA receptor blockade. Antioxid. Redox Signal. 22, 161–174.

Introduction

Ischemic stroke results from critically reduced blood flow in one or more arteries of the brain or spinal cord. If blood flow is not promptly restored, cells in the ischemic territory die of energy failure. If blood flow is restored (ischemia–reperfusion), cells may nevertheless go on to die from excitotoxicity, effects of inflammation, or programmed cell death. These processes may also kill cells that are not themselves subjected to critical ischemia, but are in the vicinity of ischemic tissues. Consequently, these processes—excitotoxicity, inflammation, and programmed cell death—are a primary focus of current stroke research (48, 59).

Of these injury mechanisms, neuronal excitotoxicity is the one triggered most rapidly after ischemia. Excitotoxicity results from sustained action of glutamate at neuronal glutamate receptors, primarily N-methyl-D-aspartate (NMDA)-type glutamate receptors (31, 105). In stroke, this is caused by the combined effects of neurotransmitter glutamate release, impaired glutamate reuptake, and reversed action of glutamate uptake transporters (62, 153). The importance of excitotoxicity as a cell death mechanism in stroke has been firmly established in a variety of stroke models and animal species, using pharmacological, genetic, and other approaches to block NMDA receptor activation (1, 47, 58, 126, 135, 137, 141, 162, 166, 168). Excitotoxicity is a particularly important cell death mechanism in stroke involving ischemia–reperfusion (43, 57, 141). However, the beneficial effects of NMDA receptor blockade in stroke require intervention at relatively short time points after onset of ischemia or reperfusion, typically within 3 h (167, 174). Clinical studies corroborate this result; interventions targeting excitotoxic injury have had no demonstrable efficacy when initiated many hours after ischemia onset, but more rapid initiation has shown salutary effects (74).

Glutamate excitotoxicity was first identified as a cell death mechanism in 1957 (113), and has since become recognized as a pathogenic mechanism in many acute and chronic neurological disorders (30, 53, 109, 122, 154). Despite this long history, our understanding of the events linking receptor activation to cell death continues to evolve. Early studies identified Ca2+ influx as a requisite event in this process (28) and correlated the magnitude of this Ca2+ influx (52, 69) and the subsequent mitochondrial Ca2+ uptake (170) with neuronal death. It was later established that the gaseous signaling molecules, nitric oxide and superoxide, were both produced during sustained NMDA receptor activation by Ca2+-dependent processes, and that production of both was required for excitotoxic death (17, 138, 158). Neither of these signaling molecules is intrinsically highly reactive or toxic, but in combination they form the highly reactive species peroxynitrite, which in turn can form additional reactive species such as hydroxyl radical and carboxyl radical, all of which readily and irreversibly react with DNA, proteins, lipids, and other crucial cell constituents (138). Subsequent events, notably poly(ADP-ribose) polymerase-1 activation and mitochondrial failure, lead to secondary energy depletion and cell death (6, 55, 97, 110, 116, 128, 188).

Nitric oxide is nonpolar, lipid permeable, and has a relatively long half-life and diffusion distance in the brain (138). Superoxide, by contrast, is polar, largely lipid impermeable, and has a relatively short half-life and diffusion distance, due in part to the ubiquitous distribution of extracellular and intracellular superoxide dismutase (138). Nitric oxide is produced by neuronal nitric oxide synthase in response to Ca2+ entry through NMDA receptors (158). The source and regulation of NMDA-induced superoxide production have been less clear, but accruing evidence points to neuronal NADPH oxidase (NOX) as the dominant source in this setting. In this study, we review this evidence, the regulatory mechanisms linking NMDA receptor activation to NOX2 activation, and the relevance of these factors to stroke pathogenesis.

NOX in the Central Nervous System

NOX has been best characterized in phagocytes and other cells of the immune system, where it plays a crucial role in bacterial killing. It has more recently been established that NOX is also expressed by many other cell types, in which it serves a variety of signaling functions. NOX is a multisubunit protein complex distinguished on the basis of their catalytic and associated subunits: NOX1–NOX5, DUOX 1, and DUOX2 (10). Of these, NOX2 is the dominant form expressed by phagocytes, and it is also the dominant form expressed by central nervous system (CNS) neurons, astrocytes, and microglia. NOX2 is composed of three cytosolic subunits, p47phox, p67phox, and p40phox, which when activated bind with two membrane-bound subunits, p22phox and gp91phox (the catalytic unit) and the small GTPase Rac1 to form an active transmembrane enzyme complex. NOX2 generates superoxide by the transfer of electrons to molecular oxygen on one side of the membrane, while oxidizing NADPH to NADP and H+ on the other side (Fig. 1). Once activated, the continued function of NOX2 in phagocytes requires a continuous source of NADPH and a rapid excretion of H+ to prevent intracellular acidification and membrane depolarization.

FIG. 1.

FIG. 1.

NADPH oxidase (NOX). The NADPH oxidase-2 (NOX2) complex generates superoxide on one side of the membrane, while oxidizing NADPH to NADP and H+ on the other side. Activation of the enzymatic complex requires translocation of the cytosolic subunits (light blue) p47phox, p67phox, and p40phox to the membrane along with Rac1 and phospholipase A2 (PLA2), where the membrane-bound subunits (dark blue) gp91 and p22phox are located. To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/ars

Other NOX isoforms are also found in the CNS, most notably NOX4 in endothelial cells (5, 10). The different NOX isoforms utilize different (but somewhat overlapping) organizing subunits and have differing regulation (10). Furthermore, some NOX isoforms localize to subcellular compartments, as determined, in part, by differences in isoform dependence on specific organizing subunits (71). NOX4 may be unique in producing hydrogen peroxide, rather than superoxide, as its final product (10, 120), and NOX4 was recently reported to colocalize with mitochondria in endothelial cells (99) and neurons (26).

NOX2 as a Source of Superoxide in Excitotoxicity and Stroke

Potential superoxide sources in neurons include mitochondria, cytosolic dehydrogenases, phospholipases, cyclooxygenases, and other oxidases, in addition to NOX. Of these, mitochondria were long thought to be the primary source of superoxide production in excitotoxicity. Support for this idea was based, in part, on the mitochondrial localization of oxidant-sensitive fluorescent indicators during neuronal glutamate exposure and a reduction in this signal with mitochondrial inhibitors (11, 50, 51, 151). However, the localization of oxidized dyes to the mitochondria does not itself establish mitochondria to be the source of oxidant production, as demonstrated by oxidation of mitoSOX in neuronal mitochondria by xanthine/xanthine oxidase added to the culture medium (82). A reduced fluorescent signal in response to mitochondrial inhibitors is likewise insufficient to establish mitochondria as the oxidant source, because these inhibitors also cause mitochondrial and plasma membrane depolarization, with resultant dye efflux (133). A quantitative assessment of mitochondrial superoxide production in excitotoxicity has been difficult because there are at least seven potential sources of superoxide production within mitochondria (9), and it is not feasible to block all of these or to make viable neurons devoid of mitochondria altogether. In addition, mitochondria normally act as sinks for extramitochondrial sources of reactive oxygen species (9), such that impaired mitochondrial function can artificially mimic superoxide production by eliminating this scavenging capacity.

Several lines of evidence point to the NOX2 isoform of NOX as the primary source of excitotoxic neuronal superoxide production. First, inhibition of NOX2 with either apocynin (139, 169, 172), diphenylene iodonium (7), or a tat-conjugated peptide inhibitor of NOX2 assembly (16, 17, 150) all block superoxide formation and neuronal death that otherwise result from NMDA receptor activation. Second, p47phox−/− neurons, which cannot form a functional NOX2 complex, likewise show markedly attenuated superoxide formation and cell death in response to NMDA (17, 60, 65, 150), and these are restored in p47phox−/− neurons transfected to express p47phox (17, 150). A third line of evidence arises from the specific requirement for glucose to fuel NADPH regeneration through the pentose phosphate pathway (45, 67). When flux through this pathway is blocked with glucose-free medium, 2-deoxyglucose, or 6-aminonicotinamide, there is again reduced superoxide formation and neuronal death (17, 171, 172). Fourth, neurons treated with a peptide inhibitor of protein kinase C (zeta) (PKCζ), which is required for neuronal p47phox phosphorylation (41, 54), show reduced superoxide formation and reduced cell death after NMDA exposures (16, 17). These findings were corroborated by studies using NMDA injections into mouse hippocampus in situ: NMDA injections induced neuronal superoxide production and subsequent neuronal death, and these were prevented by apocynin and by p47phox gene deficiency (17).

The possibility remains that superoxide produced by NOX2 might induce a secondary, possibly greater production of superoxide in mitochondria by disrupting sensitive iron–sulfur domains of mitochondrial electron transport complexes. Evidence for this has been reported in cardiac myocytes (14, 39, 94), and there is also evidence that superoxide generated in one mitochondrion can induce superoxide production in the neighboring mitochondria in this cell type (196). However, cardiac myocytes have a uniquely high mitochondrial density and stacked mitochondrial organization, and it is currently unknown if a similar superoxide-induced mitochondrial superoxide production occurs in neurons. Weighing against this idea are the observations that NMDA-induced superoxide production is not reduced in neurons that overexpress mitochondrial superoxide dismutase or in wild-type neurons treated with inhibitors of mitochondrial electron transport (17).

As noted above, several reports suggest that some NOX isoforms, including NOX2, can be localized intracellularly (26, 61, 99). The role of these intracellular NOX isoforms in excitotoxic injury remains uncertain, but the phenomenon of cell-to-cell transmission of excitotoxic injury (excitotoxic spread) further supports NOX2 at the plasma membrane to be the primary source of NMDA-induced neuronal superoxide production. Excitotoxic spread would be favored by superoxide released into the extracellular space, whereas superoxide released into the intracellular space would more likely affect the cell in which it was released. These alternatives were evaluated using neuronal cultures in which only a small subset of neurons expressed functional NOX2. The addition of NMDA to these cultures produced oxidative damage and death primarily in neurons nearby to NOX2-expressing neurons, and much less frequently in the NOX2-expressing neurons themselves (Fig. 2). Moreover, the cell-to-cell transmission of oxidative stress was blocked by the addition of superoxide dismutase to the medium, thereby confirming the extracellular oxidant species to be superoxide (150).

FIG. 2.

FIG. 2.

Neuronal NOX2 activation leads to oxidative stress in neighboring neurons. p47phox−/− neurons, which cannot assemble a functional NOX2 complex, were transfected at low efficiency with GFP-labeled p47phox (green) to reconstitute the NOX2 activity in a small fraction of cultured neurons. The cultures were treated with 100 μM N-methyl-D-aspartate (NMDA) for 30 min. 4-hydroxynonenal (4HNE) formation (red) identifies oxidative stress in many nontransfected neurons contiguous with processes of the two NOX2-competent neurons in the field (green). Neuronal soma and processes are labeled blue by immunostaining for MAP2. Reprinted from Reyes et al. (150). To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/ars

As in excitotoxicity, a crucial role for NOX in cell death has been identified in animal models of stroke. Ischemia–reperfusion increases the NOX2 activity in the brain, and both inhibitors of NOX2 activity and genetic downregulation or deficiency of NOX2 components reduce oxidative stress and infarct size in focal cerebral ischemia (81, 89, 90, 178, 186). These manipulations likewise reduce oxidative stress and neuronal death after transient forebrain ischemia (77, 172, 193, 195). The preponderance of evidence points to NOX2 as the dominant NOX isoform generating superoxide in ischemia [as reviewed by Kahles and Brandes (83, 84)], but evidence suggesting a role of NOX4 in ischemic injury has also been presented (96). Responses in neonatal brain may differ; however, one study found that NOX2 inhibition and downregulation reduced neonatal hypoxic/ischemic injury (112), but an earlier study did not (49).

The role of NOX2 in cell death following permanent focal ischemia may be less important than in ischemia–reperfusion (91), a point that will be addressed below. It should also be noted that superoxide is additionally produced by astrocytes and microglia in the brain at later time points (hours to days) after ischemia as part of the innate immune response to injury (13, 27, 111, 191, 195). This inflammatory oxidative stress may be a significant cause of secondary ischemic brain injury.

Regulation of NOX2 During NMDA Receptor Stimulation

NOX2 activation in phagocytes requires several independent events, including phosphorylation and membrane translocation of the cytosolic p47phox subunit, activation and translocation of the small GTPase Rac (Rac1 and/or Rac2, depending upon cell type), the activation of phosphoinositide-3-kinase (PI3K), and activation of phospholipase A2 (PLA2) (10, 33). Under resting conditions, p47phox assumes an autoinhibitory conformation, which prevents binding to the membrane-bound p22phox subunit (Fig. 3). Extensive phosphorylation of serine residues on p47phox leads to unfolding and unmasking of key binding domains, including a PX (phox) binding domain, which is required for binding to p22phox and membrane lipids, and SH3 (Src homology) binding domains, which target p47phox to a proline-rich region located in the C-terminus of gp91phox (63, 104). In phagocytes, these serine residues are phosphorylated by PKC, which is in turn activated by changes in intracellular calcium following receptor-mediated influx of extracellular calcium or release from internal stores. Neurons, unlike phagocytes and other cell types, uniquely activate NOX2 in response to NMDA receptor stimulation, and consequently, p47phox phosphorylation is achieved in a somewhat different way.

FIG. 3.

FIG. 3.

Coupling of NOX2 activation to NMDA receptor activation in neurons. Calcium influx via NR2B-containing NMDA-type glutamate receptors induces phosphoinositide-3-kinase (PI3K) to form phosphatidylinositol (3,4,5) trisphosphate (PIP3). PIP3 activates protein kinase C (zeta) (PKCζ), which in turn phosphorylates the p47phox organizing subunit of NOX2. Phosphorylated p47phox induces assembly of the NOX2 complex at the cell surface. The active NOX2 complex additionally requires binding to an activated GTPase, Rac1. To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/ars

Calcium influx through NR2B-containing NMDA receptors

Calcium influx has long been established as a requisite event in excitotoxicity (29), but it has been difficult to resolve whether the route of calcium influx is important in this process (23, 118, 165). Tymianski and colleagues showed that NMDA-induced nitric oxide production requires calcium influx specifically through NMDA receptors (157, 158). The pathway leading to NOX2 activation similarly requires calcium influx selectively via NMDA receptors.

NMDA receptors are usually composed of two NR1 subunits and two NR2 subunits, of which four subtypes have been described: NR2A, NR2B, NR2C, and NR2D (127, 181). Neuronal NMDA-induced NOX2 activation is blocked by selective antagonists of NR2B subunits (16), by NR2B deletion, or by replacing the NR2B C-terminus with the NR2A C-terminus (Brennan-Minnella, unpublished results). This specific requirement for NR2B in NMDA-induced NOX2 activation is consistent with recent studies identifying NR2B-containing NMDA receptors as responsible for excitotoxic neuronal death (118). Specificity for the route of Ca2+ influx in this process has additionally been shown by studies in which, Ca2+ elevations of comparable magnitude induced by ionomycin, as measured with both high- and low-affinity calcium-sensitive dyes, do not induce p47phox phosphorylation or NOX2 activation in neurons (16). However, ionomycin-induced calcium elevations, to levels much higher than occur with NMDA receptor activation, can trigger neuronal superoxide production by mechanisms independent of NOX2 (16, 165).

Roles of PKCζ and PI3K

Neurons, unlike phagocytes, activate p47phox primarily via the PKC isoforms, PKCζ and PKCδ. Peptide inhibitors of both PKCζ and PKCδ have been shown to block neuronal death induced by NMDA receptor activation and ischemia (18, 35, 98), and the role of these PKC isoforms in activating NOX2 thus provides a potential mechanism for this neuroprotective effect. PKCζ and PKCδ are not directly activated by cytosolic Ca2+ elevations (36), consistent with the failure of ionomycin-gated calcium to activate neuronal NOX2 (16). Instead, PKCζ (and possibly PKCδ) is activated by a pathway involving PI3K (Fig. 3).

Unlike conventional PKCs, which are activated by calcium and/or diacyl glycerol, PKCζ requires binding to phospholipids generated by PI3K, specifically phosphatidylinositol (3,4,5) trisphosphate (32). PI3K phosphorylates the 3 position of hydroxyl groups on the inositol ring of specific phosphatidylinositol membrane phospholipids (25), which are concentrated at the cytosolic surface of plasma membranes. These phosphorylated lipid substrates in turn recruit signaling proteins with lipid binding domains, the most common being pleckstrin homology (PH) domains, FYVE domains, and importantly for NOX2 activation, PX homology domains (107).

Both the PI3K inhibitor wortmannin and a PKCζ inhibitory peptide prevent p47phox phosphorylation, superoxide formation, and cell death, without blocking NMDA-induced calcium influx or mitochondrial depolarization (16). These factors suggest that PI3K activation is upstream of PKCζ in the pathway linking neuronal NMDA receptors to NOX2. Moreover, the wortmannin-dependent inhibition of NMDA-induced superoxide production can be overcome by either transfection with a constitutively active form of PKCζ or by exogenous supply of phosphatidylinositol (3,4,5) trisphosphate (16).

NMDA receptor activation is known to induce phosphorylation of Akt, an established downstream target of PI3K (185); however, the mechanism by which NMDA-gated Ca2+ influx triggers PI3K activation remains unresolved. Early reports suggested that the SH2 binding domain of the p85 subunit of PI3K could bind directly to phosphorylated tyrosine residues on the NR2B subunit of NMDA receptors. More recently, however, it has been shown that the PI3K activation by NMDA receptors may require the adaptor protein APPL1 (75, 131, 189).

The role of PI3K in excitotoxicity and oxidative stress contrasts with the well-established, prosurvival effects of PI3K mediated by the Akt pathway (19). Previous work has demonstrated that the prosurvival pathways required for the survival of developing neurons are dependent on NMDA-induced activation of PI3K (102, 140), and that selective disruption of NMDA-induced Akt signaling results in widespread neuronal apoptosis (189). The contrasting toxic and trophic roles of NMDA-induced PI3K activity may be reconciled by the observations that PI3K and superoxide are both mediators of intercellular signaling and synaptic plasticity in the nervous system (37, 93, 121). The cytotoxic, PI3K-dependent superoxide production that occurs during sustained NMDA receptor activation may thus be viewed as an exaggerated, pathological manifestation of a process that normally functions in neuronal plasticity.

In addition to their role in physiological signaling, there is evidence that PI3K and NOX-dependent superoxide are required for ischemic preconditioning. Ischemic preconditioning is a process whereby cells exposed to sublethal ischemia acquire resistance to subsequent otherwise lethal insults (56). In both an in vitro model using oxygen glucose deprivation in cultured astrocytes (155) or with transient global ischemia in mice (124), the protective effects of ischemic preconditioning were abolished when the selective inhibitor of PI3K, LY294002 was administered. Intriguingly, NAPDH oxidase activity is also required for ischemic preconditioning, at least in certain settings. The protective preconditioning effect achieved with ethanol is reversed by the NOX inhibitor apocynin (187), and similarly, lipopolysaccharide (LPS) administration fails to elicit ischemic preconditioning in NOX2-deficient animals (80, 88, 101). These observations suggest that the protective effects of ischemic preconditioning may require both PI3K activation and NOX superoxide production.

Regulation of NOX2 by Rac and protein kinase CK2

Rac1 (or in some cells, Rac2) is an additional requisite component of functional NOX2 oxidase, and it may also be activated by PI3K (25). Rac1 is a Rho GTPase signaling protein and functions as a guanine nucleotide exchange factor to catalyze the conversion of inactive GDP-bound GTPases to active GTP-bound complexes. Rac1 contains lipid binding domains, and PI3K activity increases GTP-bound Rac1 (70, 78). Ligand-dependent activation of NOX in fibroblasts and astroglioma cells is prevented by a dominant negative Rac1, (106, 173), and conversely, the NOX activity is increased by a constitutively active form of Rac1 (173). Evidence suggests that Rac1 is required for translocation of the p67phox cytosolic subunit (123), and may also act upstream of other required lipid mediators of NOX activation such as PLA2 (106, 144). In a stroke model, Brann and colleagues reported that the Rac inhibitor NSC23766 attenuated NOX activation and reduced oxidative stress and neuronal death (149). Interestingly, 17 beta-estradiol leads to phosphorylation/inactivation of Rac1 by an extranuclear signaling mechanism, resulting in a profound attenuation of NOX activation and superoxide production in ischemia–reperfusion (15, 195); a factor that may contribute to the significant gender differences observed in ischemic brain injury (79).

Rac1 is additionally regulated by protein kinase CK2 (also known as casein kinase 2) a serine/threonine kinase found in neurons and most other cell types (164). CK2 consists of three different subunits; two catalytic (α and α′) and one regulatory subunit (β). Recent work showed that neuronal CK2 protein expression and kinase activity are significantly reduced in the ischemic cortex (90). Moreover, pharmacological or siRNA-mediated suppression of CK2 activity exacerbates neuronal death in both cell culture and animal models of brain ischemia (89, 90). Decreased levels of CK2 correlated with increased NOX assembly and NOX-dependent superoxide production. Coimmunoprecipitation studies showed a direct physical interaction between CK2 and Rac1, and the authors proposed a model in which, under basal conditions, CK2 binds Rac1, preventing activation of NOX (90). Following ischemia, proteasome-dependent degradation of CK2 allows for Rac1 release and facilitates NOX complex assembly and superoxide formation (90).

Effect of intracellular acidification on NOX2

Cerebral ischemia produces acidosis to varying degrees. Severe acidosis, below pH 6.4, exacerbates cell ischemic injury (100, 145, 146, 192), but lesser degrees of acidosis, in the range of pH 7.0–6.5, reduce both ischemic injury (161) and glutamate-induced neuronal death (86, 179). These effects may be attributable, in part, to an inhibitory effect of hydrogen ions on calcium influx through NMDA receptor channels, but this effect is minimal at pH values above 6.6, (176, 180, 184).

NOX2 activity is highly pH sensitive (68, 125, 160, 182), with a 50% reduction in activity observed with a 0.2 pH unit reduction in intracellular pH. In many cell types, H+ generated by NOX2 is released to the extracellular space through voltage-dependent hydrogen ion channels or by Na+/H+ exchangers, and inhibition of this release causes intracellular acidification and resultant cessation of NOX2 activity (111, 148, 159). The mechanism of this effect is unlikely to be a simple mass action, given its steep pH dependence; instead, evidence suggests that the H+ concentration influences the phosphorylation status of p47phox (159).

Given the pH sensitivity of NOX2 and the role of this enzyme in NMDA receptor-mediated cell death, modest reductions in pH might limit neurotoxicity by dissociating NMDA receptor activation from superoxide production (Fig. 4). Studies confirm that both the superoxide production and cell death resulting from neuronal NMDA receptor activation are extremely pH sensitive, with a near complete reduction observed at intracellular pH below 6.7 (normal is pH 7.0). Acidification induced by Na+/H+ exchange inhibition or NHE1 gene deficiency also blocks superoxide production and cell death induced by NMDA injected into the mouse striatum, and reduces neuronal injury after ischemia–reperfusion (103, 114).

FIG. 4.

FIG. 4.

Intracellular H+ inhibits NOX2 activity. The oxidation of NADPH during the enzymatic activation of NOX also produces H+, and removal of these H+ is required for sustained NOX activity. In most cells types, this is accomplished by the proton channel, Hv1, and/or the Na+/H+ exchanger, NHE1. In addition, to production of H+ by NOX, several other pathways may contribute to intracellular acidification during ischemia, and thus indirectly inhibit NOX2 activity. These include anaerobic metabolism of glucose to lactic acid, mitochondrial depolarization, and extrusion of Ca2+ by Ca2+/H+ exchange. To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/ars

Excessive superoxide production may be deleterious in ischemia and other settings in which sustained NMDA receptor stimulation, and thus, it is possible that intracellular acidosis may function in a teleologically advantageous way to limit superoxide production. Neurons acidify during ischemia as a result of anaerobic lactic acidosis and other processes (Fig. 4), and the reduced blood flow during ischemia prevents removal of accumulated H+. Importantly, reperfusion restores aerobic metabolism and clears accumulated H+, thereby restoring the NOX2 activity. This effect of reperfusion may thereby provide an explanation for why superoxide production is greater in reperfused than nonreperfused ischemic brain, and why both NOX2 inhibitors and NMDA receptor antagonists are more effective as neuroprotective agents in reperfused than nonreperfused brain (22, 91, 175).

PLA2 in stroke and NOX2 activation

PLA2 is a family of enzymes that catalyze the removal of fatty acids from membrane phospholipids at the sn-2 position to produce lysophospholipids and free fatty acids, such as arachidonic acid (2). Several lines of evidence implicate PLA2 in the pathogenesis of ischemic brain injury. PLA2 expression and activity are increased in the injured region of the brain after ischemia (3, 4, 34, 134), and both genetic (12) and pharmacological (76, 194) reductions in PLA2 activity reduce injury in animal models of stroke. Increased PLA2 expression is associated with increased oxidative stress after ischemic injury, and the increase in oxidative stress is attenuated in PLA2-deficient mice (95).

Phagocytes deficient in PLA2 exhibit reduced NOX2 activity in response to phorbol ester and other stimuli. Furthermore, exogenous addition of free arachidonic acid, but not other free fatty acids or metabolites of arachidonic acid, is sufficient to restore NOX function (40, 117). Studies using pharmacological inhibitors or antisense downregulation of PLA2 also show that arachidonic acid production is required for NOX superoxide production (108). The mechanism by which arachidonic acid regulates NOX function remains uncertain, but there is evidence in phagocytic cells that localized arachidonic acid production is required for the release of H+ formed by NOX (72). In addition to these direct effects, arachidonic acid can be further metabolized by cyclooxygenases and lipoxygenases to form bioactive eicosanoids that indirectly lead to oxidative stress (2) and facilitate NOX2 activation by an as-yet unresolved mechanism (42, 108).

Glucose, Hyperglycemia, and NOX2 Activity

The deleterious effect of elevated blood glucose (hyperglycemia) is one of the most robust and reproducible aspects of stroke, particularly in ischemia–reperfusion (119, 130, 143). A widely cited mechanism by which hyperglycemia can exacerbate brain injury is through fueling the accumulation of lactic acid in hypoxic tissues (146); however, elevated glucose concentrations also exacerbate injury in brain slice models of ischemia in which pH is tightly controlled (132, 156), and in animal models of brain ischemia in which tissue acidosis is relieved by reperfusion (38, 142, 183). Moreover, clinical studies suggest that hyperglycemia during reperfusion increases risk of poor outcomes, independent of diabetes or preischemic blood glucose concentrations (20, 152). These observations suggest that a mechanism other than lactic acidosis may contribute to the effect of glucose on reperfusion injury.

In phagocytes and other cell types, the activity of NOX requires the continuous metabolism of glucose by the hexose monophosphate shunt for supply of NADPH substrate (45, 67) (Fig. 5). In cultured neurons, NMDA-induced superoxide production and cell death are both blocked when glucose is omitted from the culture medium, suggesting a similar requirement for glucose in neuronal NOX2 function (17). The same pattern was observed in neuronal cultures exposed to transient oxygen/glucose deprivation, a model used to mimic brain ischemia–reperfusion. Neurons in these cultures showed a rapid increase in superoxide production upon return of oxygen and glucose to the medium, and this did not occur if either glucose was omitted or glucose metabolism to NADPH was pharmacologically inhibited (172). Conversely, in a mouse model of ischemia–reperfusion, the superoxide produced during reperfusion was augmented by hyperglycemia, and this effect of hyperglycemia was negated in apocynin-treated and p47phox−/− mice (which cannot form an active NOX2 complex). These reports suggest that hyperglycemia can exacerbate reperfusion injury by supplying increased glucose substrate for superoxide production by NOX (Fig. 6).

FIG. 5.

FIG. 5.

Glucose is required for NOX2 activity. NOX2 oxidizes NADPH to NADP+, and continuous NOX2 function requires a continuous NADPH supply. NADPH is regenerated by the pentose phosphate pathway, which uses glucose as an obligate substrate. To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/ars

FIG. 6.

FIG. 6.

Hyperglycemia promotes reperfusion injury and hemorrhage by fueling NOX2. Neuronal superoxide production is low under basal conditions. It remains low after arterial occlusion because of limited oxygen supply and lactic acidosis. Upon reperfusion, the oxygen supply is restored and lactic acid is washed out. NOX2 then produces high levels of superoxide in response to extracellular glutamate acting at NMDA-type glutamate receptors. Glucose availability can be rate limiting for NOX2 superoxide production, and under hyperglycemic conditions, the increased glucose supply leads to increased superoxide production. This in turn can increase damage to blood vessel walls and promote reperfusion hemorrhage. To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/ars

Hyperglycemia and Reperfusion Hemorrhage

The only medical treatment proven clinically effective for acute stroke is thrombolysis with tissue plasminogen activator (tPA). However, the use of tPA carries the risk of reperfusion hemorrhage, and this restricts its use to patients who can be treated within 3–6 h after the onset of symptoms (64). Clinical studies have established a strong association between admission hyperglycemia and the risk of hemorrhage with tPA use, independent of prior diabetes, with the risk of hemorrhage roughly doubling with each 5.5 mM (100 mg/dl) increase in blood glucose (8, 21, 44, 46, 73, 87, 163). The reason for this increased risk has not been established, but may also be attributable to an increased NOX2 activity (Fig. 5). Rats undergoing focal ischemia and given tPA at reperfusion were found, as in the clinical studies, to have increased brain hemorrhage if they were rendered hyperglycemic. The hyperglycemia also induced increased superoxide formation in the reperfused brain, and both the increased superoxide and increase hemorrhage rate were prevented by the NOX2 inhibitor apocynin (190). This result is consistent with other studies demonstrating a role of NOX2 in the blood–brain barrier disruption (85, 177).

Conclusions

Studies reviewed here indicate that neuronal superoxide production in stroke and excitotoxicity is not an inevitable physical consequence of mitochondrial calcium overload, but is instead the end result of a complex and regulated signaling pathway linking NMDA receptors and NOX2. Factors and regulatory steps involved in this pathway include glucose supply, intracellular pH, PI3K, Rac1, PLA2, and PKCζ/δ, csk2, and estrogen. Many of these factors have previously been identified as being critical mediators of cell death in stroke, and consequently, their roles as regulators of NOX2 activity provide new or alternative mechanisms for their effects on ischemic brain injury. Conversely, dysregulation of these regulatory factors could in principle amplify superoxide production and cause cell injury at otherwise benign levels of NMDA receptor stimulation.

The key aspects of these signaling pathways remain unresolved. In particular, it remains unclear whether PI3K is directly upstream of the Rac1 activation required for NOX2 activation, or if Rac1 is activated by an independent pathway. Similarly, it is unclear how PLA2 is activated during excitotoxicity and how, at a biochemical level, PLA2 regulates NOX2. The potential interactions between NOX2 and neuronal mitochondria also remain to be established, and it remains possible that superoxide produced by NOX2 could damage mitochondria and thereby induce mitochondrial superoxide release in a feed-forward manner. A fundamental unresolved question is the physiological function of the NMDA/NOX2 signaling pathway in the brain. Superoxide production and NMDA receptor activation have each, independently, been shown to influence synaptic plasticity, the process by which learning and memory occur (115, 121, 129). The cytotoxic superoxide production that occurs during sustained NMDA receptor activation in stroke might thus represent an exaggerated, pathological manifestation of a process that is unique to CNS.

Specific, small molecule inhibitors of NOX2 and other NOX isoforms are currently under development (92), and there is growing enthusiasm for using this approach in the treatment of acute stroke (24, 83, 147). This enthusiasm must be tempered, however, by the experience gained from prior studies of neuroprotectant therapy in stroke. To be clinically useful, NOX inhibitors must pass readily into the ischemic brain and have tolerable side effects. Most importantly, clinical efficacy with this approach will almost certainly require administration within relatively short periods (hours) of ischemic onset, given the rapid pace of ischemic cell death processes. Studies using NOX inhibitors in animal models of stroke also suggest that rapid administration will be needed (77, 81, 96, 172, 178, 193, 195). This being said, the potential for NOX inhibitors as neuroprotective agents during therapeutic thrombolysis or clot removal is significant, particularly, given the role of reperfusion in NOX2 activation.

Many of the regulatory factors shown to influence the NOX2 activity are pharmacologically accessible, and interventions targeting these factors have been shown to block both excitotoxic and acute ischemic injury. It remains to be established whether these interventions can provide a greater neuroprotective efficacy or a longer time window to treatment than provided by NOX2 or NMDA receptor antagonists, or if dissociating superoxide production from other downstream signaling effects of NMDA receptor activation can mitigate the toxic effects of NMDA receptor blockade (66, 136).

Abbreviations Used

4HNE

4 hydroxynonenal

CNS

central nervous system

CK2

protein kinase CK2 (also known as casein kinase 2)

LPS

lipopolysaccharide

NADPH

nicotinamide adenine dinucleotide phosphate (reduced form)

NMDA

N-methyl-D-aspartate

NOX

NADPH oxidase

PI3K

phosphoinositide-3-kinase

PIP3

phosphatidylinositol (3,4,5) trisphosphate

PKCδ

protein kinase C (delta)

PKCζ

protein kinase C (zeta)

PLA2

phospholipase A2

tPA

tissue plasminogen activator

Acknowledgments

This work was supported by the U.S. Dept. of Veterans Affairs and National Institutes of Health (grant NS081149). We thank Colleen Hefner for assistance with the article preparation.

References

  • 1.Aarts M, Liu Y, Liu L, Besshoh S, Arundine M, Gurd JW, Wang YT, Salter MW, and Tymianski M. Treatment of ischemic brain damage by perturbing NMDA receptor- PSD-95 protein interactions. Science 298: 846–850, 2002 [DOI] [PubMed] [Google Scholar]
  • 2.Adibhatla RM. and Hatcher JF. Phospholipase A(2), reactive oxygen species, and lipid peroxidation in CNS pathologies. BMB Rep 41: 560–567, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Adibhatla RM, Hatcher JF, and Dempsey RJ. Phospholipase A2, hydroxyl radicals, and lipid peroxidation in transient cerebral ischemia. Antioxid Redox Signal 5: 647–654, 2003 [DOI] [PubMed] [Google Scholar]
  • 4.Adibhatla RM, Hatcher JF, Larsen EC, Chen X, Sun D, and Tsao FH. CDP-choline significantly restores phosphatidylcholine levels by differentially affecting phospholipase A2 and CTP: phosphocholine cytidylyltransferase after stroke. J Biol Chem 281: 6718–6725, 2006 [DOI] [PubMed] [Google Scholar]
  • 5.Ago T, Kitazono T, Ooboshi H, Iyama T, Han YH, Takada J, Wakisaka M, Ibayashi S, Utsumi H, and Iida M. Nox4 as the major catalytic component of an endothelial NAD(P)H oxidase. Circulation 109: 227–233, 2004 [DOI] [PubMed] [Google Scholar]
  • 6.Alano CC, Garnier P, Ying W, Higashi Y, Kauppinen TM, and Swanson RA. NAD+ depletion is necessary and sufficient for poly(ADP-ribose) polymerase-1-mediated neuronal death. J Neurosci 30: 2967–2978, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Alekseenko AV, Lemeshchenko VV, Pekun TG, Waseem TV, and Fedorovich SV. Glutamate-induced free radical formation in rat brain synaptosomes is not dependent on intrasynaptosomal mitochondria membrane potential. Neurosci Lett 513: 238–242, 2012 [DOI] [PubMed] [Google Scholar]
  • 8.Alvarez-Sabin J, Molina CA, Montaner J, Arenillas JF, Huertas R, Ribo M, Codina A, and Quintana M. Effects of admission hyperglycemia on stroke outcome in reperfused tissue plasminogen activator—treated patients. Stroke 34: 1235–1241, 2003 [DOI] [PubMed] [Google Scholar]
  • 9.Andreyev AY, Kushnareva YE, and Starkov AA. Mitochondrial metabolism of reactive oxygen species. Biochemistry (Mosc) 70: 200–214, 2005 [DOI] [PubMed] [Google Scholar]
  • 10.Bedard K. and Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev 87: 245–313, 2007 [DOI] [PubMed] [Google Scholar]
  • 11.Bindokas VP, Jordan J, Lee CC, and Miller RJ. Superoxide production in rat hippocampal neurons: selective imaging with hydroethidine. J Neurosci 16: 1324–1336, 1996 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bonventre JV, Huang Z, Taheri MR, O'Leary E, Li E, Moskowitz MA, and Sapirstein A. Reduced fertility and postischaemic brain injury in mice deficient in cytosolic phospholipase A2. Nature 390: 622–625, 1997 [DOI] [PubMed] [Google Scholar]
  • 13.Brait VH, Jackman KA, Walduck AK, Selemidis S, Diep H, Mast AE, Guida E, Broughton BR, Drummond GR, and Sobey CG. Mechanisms contributing to cerebral infarct size after stroke: gender, reperfusion, T lymphocytes, and Nox2-derived superoxide. J Cereb Blood Flow Metab 30: 1306–1317, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Brandes RP. Triggering mitochondrial radical release: a new function for NADPH oxidases. Hypertension 45: 847–848, 2005 [DOI] [PubMed] [Google Scholar]
  • 15.Brann D, Raz L, Wang R, Vadlamudi R, and Zhang Q. Oestrogen signalling and neuroprotection in cerebral ischaemia. J Neuroendocrinol 24: 34–47, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Brennan-Minnella AM, Shen Y, El-Benna J, and Swanson RA. Phosphoinositide 3-kinase couples NMDA receptors to superoxide release in excitotoxic neuronal death. Cell Death Dis 4: e624, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Brennan AM, Suh SW, Won SJ, Narasimhan P, Kauppinen TM, Lee H, Edling Y, Chan PH, and Swanson RA. NADPH oxidase is the primary source of superoxide induced by NMDA receptor activation. Nat Neurosci 12: 857–863, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bright R, Raval AP, Dembner JM, Perez-Pinzon MA, Steinberg GK, Yenari MA, and Mochly-Rosen D. Protein kinase C delta mediates cerebral reperfusion injury in vivo. J Neurosci 24: 6880–6888, 2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Brunet A, Datta SR, and Greenberg ME. Transcription-dependent and -independent control of neuronal survival by the PI3K-Akt signaling pathway. Curr Opin Neurobiol 11: 297–305, 2001 [DOI] [PubMed] [Google Scholar]
  • 20.Bruno A, Biller J, Adams HP, Jr., Clarke WR, Woolson RF, Williams LS, and Hansen MD. Acute blood glucose level and outcome from ischemic stroke. Trial of ORG 10172 in Acute Stroke Treatment (TOAST) Investigators. Neurology 52: 280–284, 1999 [DOI] [PubMed] [Google Scholar]
  • 21.Bruno A, Levine SR, Frankel MR, Brott TG, Lin Y, Tilley BC, Lyden PD, Broderick JP, Kwiatkowski TG, and Fineberg SE. Admission glucose level and clinical outcomes in the NINDS rt-PA Stroke Trial. Neurology 59: 669–674, 2002 [DOI] [PubMed] [Google Scholar]
  • 22.Buchan A. and Pulsinelli WA. Hypothermia but not the N-methyl-D-aspartate antagonist, MK-801, attenuates neuronal damage in gerbils subjected to transient global ischemia. J Neurosci 10: 311–316, 1990 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Budd SL. and Nicholls DG. Mitochondria, calcium regulation, and acute glutamate excitotoxicity in cultured cerebellar granule cells. J Neurochem 67: 2282–2291, 1996 [DOI] [PubMed] [Google Scholar]
  • 24.Cairns B, Kim JY, Tang XN, and Yenari MA. NOX inhibitors as a therapeutic strategy for stroke and neurodegenerative disease. Curr Drug Targets 13: 199–206, 2012 [DOI] [PubMed] [Google Scholar]
  • 25.Cantley LC. The phosphoinositide 3-kinase pathway. Science 296: 1655–1657, 2002 [DOI] [PubMed] [Google Scholar]
  • 26.Case AJ, Li S, Basu U, Tian J, and Zimmerman MC. Mitochondrial-localized NADPH oxidase 4 is a source of superoxide in angiotensin II-stimulated neurons. Am J Physiol Heart Circ Physiol 305: H19–H28, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Chen H, Kim GS, Okami N, Narasimhan P, and Chan PH. NADPH oxidase is involved in post-ischemic brain inflammation. Neurobiol Dis 42: 341–348, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Choi DW. Glutamate neurotoxicity in cortical cell culture is calcium dependent. Neurosci Lett 58: 293–297, 1985 [DOI] [PubMed] [Google Scholar]
  • 29.Choi DW. Ionic dependence of glutamate neurotoxicity. J Neurosci 7: 369–379, 1987 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Choi DW. Glutamate neurotoxicity and diseases of the nervous system. Neuron 1: 623–634, 1988 [DOI] [PubMed] [Google Scholar]
  • 31.Choi DW. Excitotoxic cell death. J Neurobiol 23: 1261–1276, 1992 [DOI] [PubMed] [Google Scholar]
  • 32.Chou MM, Hou W, Johnson J, Graham LK, Lee MH, Chen CS, Newton AC, Schaffhausen BS, and Toker A. Regulation of protein kinase C zeta by PI 3-kinase and PDK-1. Curr Biol 8: 1069–1077, 1998 [DOI] [PubMed] [Google Scholar]
  • 33.Clark RA, Volpp BD, Leidal KG, and Nauseef WM. Two cytosolic components of the human neutrophil respiratory burst oxidase translocate to the plasma membrane during cell activation. J Clin Invest 85: 714–721, 1990 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Clemens JA, Stephenson DT, Smalstig EB, Roberts EF, Johnstone EM, Sharp JD, Little SP, and Kramer RM. Reactive glia express cytosolic phospholipase A2 after transient global forebrain ischemia in the rat. Stroke 27: 527–535, 1996 [DOI] [PubMed] [Google Scholar]
  • 35.Crisanti P, Leon A, Lim DM, and Omri B. Aspirin prevention of NMDA-induced neuronal death by direct protein kinase Czeta inhibition. J Neurochem 93: 1587–1593, 2005 [DOI] [PubMed] [Google Scholar]
  • 36.Csukai M. and Mochly-Rosen D. Pharmacologic modulation of protein kinase C isozymes: the role of RACKs and subcellular localisation. Pharmacol Res 39: 253–259, 1999 [DOI] [PubMed] [Google Scholar]
  • 37.Cuesto G, Enriquez-Barreto L, Carames C, Cantarero M, Gasull X, Sandi C, Ferrus A, Acebes A, and Morales M. Phosphoinositide-3-kinase activation controls synaptogenesis and spinogenesis in hippocampal neurons. J Neurosci 31: 2721–2733, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.D'Alecy LG, Lundy EF, Barton KJ, and Zelenock GB. Dextrose containing intravenous fluid impairs outcome and increases death after eight minutes of cardiac arrest and resuscitation in dogs. Surgery 100: 505–511, 1986 [PubMed] [Google Scholar]
  • 39.Daiber A. Redox signaling (cross-talk) from and to mitochondria involves mitochondrial pores and reactive oxygen species. Biochim Biophys Acta 1797: 897–906, 2010 [DOI] [PubMed] [Google Scholar]
  • 40.Dana R, Leto TL, Malech HL, and Levy R. Essential requirement of cytosolic phospholipase A2 for activation of the phagocyte NADPH oxidase. J Biol Chem 273: 441–445, 1998 [DOI] [PubMed] [Google Scholar]
  • 41.Dang PM, Fontayne A, Hakim J, El Benna J, and Perianin A. Protein kinase C zeta phosphorylates a subset of selective sites of the NADPH oxidase component p47phox and participates in formyl peptide-mediated neutrophil respiratory burst. J Immunol 166: 1206–1213, 2001 [DOI] [PubMed] [Google Scholar]
  • 42.Daniels I, Lindsay MA, Keany CI, Burden RP, Fletcher J, and Haynes AP. Role of arachidonic acid and its metabolites in the priming of NADPH oxidase in human polymorphonuclear leukocytes by peritoneal dialysis effluent. Clin Diagn Lab Immunol 5: 683–689, 1998 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Dawson DA, Graham DI, McCulloch J, and Macrae IM. Anti-ischaemic efficacy of a nitric oxide synthase inhibitor and a N-methyl-D-aspartate receptor antagonist in models of transient and permanent focal cerebral ischaemia. Br J Pharmacol 113: 247–253, 1994 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.De Silva DA, Ebinger M, Christensen S, Parsons MW, Levi C, Butcher K, Barber PA, Bladin C, Donnan GA, and Davis SM. Baseline diabetic status and admission blood glucose were poor prognostic factors in the EPITHET trial. Cerebrovasc Dis 29: 14–21, 2010 [DOI] [PubMed] [Google Scholar]
  • 45.Decoursey TE. and Ligeti E. Regulation and termination of NADPH oxidase activity. Cell Mol Life Sci 62: 2173–2193, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Demchuk AM, Morgenstern LB, Krieger DW, Linda Chi T, Hu W, Wein TH, Hardy RJ, Grotta JC, and Buchan AM. Serum glucose level and diabetes predict tissue plasminogen activator-related intracerebral hemorrhage in acute ischemic stroke. Stroke 30: 34–39, 1999 [DOI] [PubMed] [Google Scholar]
  • 47.Diemer NH, Johansen FF, Benveniste H, Bruhn T, Berg M, Valente E, and Jorgensen MB. Ischemia as an excitotoxic lesion: protection against hippocampal nerve cell loss by denervation. Acta Neurochir Suppl (Wien) 57: 94–101, 1993 [DOI] [PubMed] [Google Scholar]
  • 48.Dirnagl U, Iadecola C, and Moskowitz MA. Pathobiology of ischaemic stroke: an integrated view. Trends Neurosci 22: 391–397, 1999 [DOI] [PubMed] [Google Scholar]
  • 49.Doverhag C, Keller M, Karlsson A, Hedtjarn M, Nilsson U, Kapeller E, Sarkozy G, Klimaschewski L, Humpel C, Hagberg H, Simbruner G, Gressens P, and Savman K. Pharmacological and genetic inhibition of NADPH oxidase does not reduce brain damage in different models of perinatal brain injury in newborn mice. Neurobiol Dis 31: 133–144, 2008 [DOI] [PubMed] [Google Scholar]
  • 50.Duan Y, Gross RA, and Sheu SS. Ca2+-dependent generation of mitochondrial reactive oxygen species serves as a signal for poly(ADP-ribose) polymerase-1 activation during glutamate excitotoxicity. J Physiol 585: 741–758, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Dugan LL, Sensi SL, Canzoniero LM, Handran SD, Rothman SM, Lin TS, Goldberg MP, and Choi DW. Mitochondrial production of reactive oxygen species in cortical neurons following exposure to N-methyl-D-aspartate. J Neurosci 15: 6377–6388, 1995 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Eimerl S. and Schramm M. The quantity of calcium that appears to induce neuronal death. J Neurochem 62: 1223–1226, 1994 [DOI] [PubMed] [Google Scholar]
  • 53.Faden AI, Demediuk P, Panter SS, and Vink R. The role of excitatory amino acids and NMDA receptors in traumatic brain injury. Science 244: 798–800, 1989 [DOI] [PubMed] [Google Scholar]
  • 54.Fontayne A, Dang PM, Gougerot-Pocidalo MA, and El-Benna J. Phosphorylation of p47phox sites by PKC alpha, beta II, delta, and zeta: effect on binding to p22phox and on NADPH oxidase activation. Biochemistry 41: 7743–7750, 2002 [DOI] [PubMed] [Google Scholar]
  • 55.Formentini L, Macchiarulo A, Cipriani G, Camaioni E, Rapizzi E, Pellicciari R, Moroni F, and Chiarugi A. Poly(ADP-ribose) catabolism triggers AMP-dependent mitochondrial energy failure. J Biol Chem 284: 17668–17676, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Gidday JM. Cerebral preconditioning and ischaemic tolerance. Nat Rev Neurosci 7: 437–448, 2006 [DOI] [PubMed] [Google Scholar]
  • 57.Gill R, Brazell C, Woodruff GN, and Kemp JA. The neuroprotective action of dizocilpine (MK-801) in the rat middle cerebral artery occlusion model of focal ischaemia. Br J Pharmacol 103: 2030–2036, 1991 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Gill R, Foster AC, and Woodruff GN. Systemic administration of MK-801 protects against ischemia-induced hippocampal neurodegeneration in the gerbil. J Neurosci 7: 3343–3349, 1987 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ginsberg MD. Neuroprotection for ischemic stroke: past, present and future. Neuropharmacology 55: 363–389, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Girouard H, Wang G, Gallo EF, Anrather J, Zhou P, Pickel VM, and Iadecola C. NMDA receptor activation increases free radical production through nitric oxide and NOX2. J Neurosci 29: 2545–2552, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Glass MJ, Huang J, Oselkin M, Tarsitano MJ, Wang G, Iadecola C, and Pickel VM. Subcellular localization of nicotinamide adenine dinucleotide phosphate oxidase subunits in neurons and astroglia of the rat medial nucleus tractus solitarius: relationship with tyrosine hydroxylase immunoreactive neurons. Neuroscience 143: 547–564, 2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Gouix E, Leveille F, Nicole O, Melon C, Had-Aissouni L, and Buisson A. Reverse glial glutamate uptake triggers neuronal cell death through extrasynaptic NMDA receptor activation. Mol Cell Neurosci 40: 463–473, 2009 [DOI] [PubMed] [Google Scholar]
  • 63.Groemping Y, Lapouge K, Smerdon SJ, and Rittinger K. Molecular basis of phosphorylation-induced activation of the NADPH oxidase. Cell 113: 343–355, 2003 [DOI] [PubMed] [Google Scholar]
  • 64.Group NIoNDaSr-PSS. Tissue plasminogen activator for acute ischemic stroke. The National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group. N Engl J Med 333: 1581–1587, 1995 [DOI] [PubMed] [Google Scholar]
  • 65.Guemez-Gamboa A, Estrada-Sanchez AM, Montiel T, Paramo B, Massieu L, and Moran J. Activation of NOX2 by the stimulation of ionotropic and metabotropic glutamate receptors contributes to glutamate neurotoxicity in vivo through the production of reactive oxygen species and calpain activation. J Neuropathol Exp Neurol 70: 1020–1035, 2011 [DOI] [PubMed] [Google Scholar]
  • 66.Gunduz-Bruce H. The acute effects of NMDA antagonism: from the rodent to the human brain. Brain Res Rev 60: 279–286, 2009 [DOI] [PubMed] [Google Scholar]
  • 67.Gupte SA, Levine RJ, Gupte RS, Young ME, Lionetti V, Labinskyy V, Floyd BC, Ojaimi C, Bellomo M, Wolin MS, and Recchia FA. Glucose-6-phosphate dehydrogenase-derived NADPH fuels superoxide production in the failing heart. J Mol Cell Cardiol 41: 340–349, 2006 [DOI] [PubMed] [Google Scholar]
  • 68.Gyllenhammar H. Effects of extracellular pH on neutrophil superoxide anion production, and chemiluminescence augmented with luminol, lucigenin or DMNH. J Clin Lab Immunol 28: 97–102, 1989 [PubMed] [Google Scholar]
  • 69.Hartley DM, Kurth MC, Bjerkness L, Weiss JH, and Choi DW. Glutamate receptor-induced 45Ca2+ accumulation in cortical cell culture correlates with subsequent neuronal degeneration. J Neurosci 13: 1993–2000, 1993 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Hawkins PT, Eguinoa A, Qiu RG, Stokoe D, Cooke FT, Walters R, Wennstrom S, Claesson-Welsh L, Evans T, Symons M, et al. PDGF stimulates an increase in GTP-Rac via activation of phosphoinositide 3-kinase. Curr Biol 5: 393–403, 1995 [DOI] [PubMed] [Google Scholar]
  • 71.Helmcke I, Heumuller S, Tikkanen R, Schroder K, and Brandes RP. Identification of structural elements in Nox1 and Nox4 controlling localization and activity. Antioxid Redox Signal 11: 1279–1287, 2009 [DOI] [PubMed] [Google Scholar]
  • 72.Henderson LM, Thomas S, Banting G, and Chappell JB. The arachidonate-activatable, NADPH oxidase-associated H+ channel is contained within the multi-membrane-spanning N-terminal region of gp91-phox. Biochem J 325 (Pt 3): 701–705, 1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Hill MD. and Buchan AM. Thrombolysis for acute ischemic stroke: results of the Canadian Alteplase for Stroke Effectiveness Study. CMAJ 172: 1307–1312, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hill MD, Martin RH, Mikulis D, Wong JH, Silver FL, Terbrugge KG, Milot G, Clark WM, Macdonald RL, Kelly ME, Boulton M, Fleetwood I, McDougall C, Gunnarsson T, Chow M, Lum C, Dodd R, Poublanc J, Krings T, Demchuk AM, Goyal M, Anderson R, Bishop J, Garman D, and Tymianski M. Safety and efficacy of NA-1 in patients with iatrogenic stroke after endovascular aneurysm repair (ENACT): a phase 2, randomised, double-blind, placebo-controlled trial. Lancet Neurol 11: 942–950, 2013 [DOI] [PubMed] [Google Scholar]
  • 75.Hisatsune C, Umemori H, Mishina M, and Yamamoto T. Phosphorylation-dependent interaction of the N-methyl-D-aspartate receptor epsilon 2 subunit with phosphatidylinositol 3-kinase. Genes Cells 4: 657–666, 1999 [DOI] [PubMed] [Google Scholar]
  • 76.Hoda MN, Singh I, Singh AK, and Khan M. Reduction of lipoxidative load by secretory phospholipase A2 inhibition protects against neurovascular injury following experimental stroke in rat. J Neuroinflammation 6: 21, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Hong H, Zeng JS, Kreulen DL, Kaufman DI, and Chen AF. Atorvastatin protects against cerebral infarction via inhibition of NADPH oxidase-derived superoxide in ischemic stroke. Am J Physiol Heart Circ Physiol 291: H2210–H2215, 2006 [DOI] [PubMed] [Google Scholar]
  • 78.Hordijk PL. Regulation of NADPH oxidases: the role of Rac proteins. Circ Res 98: 453–462, 2006 [DOI] [PubMed] [Google Scholar]
  • 79.Hurn PD. and Macrae IM. Estrogen as a neuroprotectant in stroke. J Cereb Blood Flow Metab 20: 631–652, 2000 [DOI] [PubMed] [Google Scholar]
  • 80.Iadecola C, Kahles T, Gallo EF, and Anrather J. Neurovascular protection by ischaemic tolerance: role of nitric oxide. J Physiol 589: 4137–4145, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Jackman KA, Miller AA, De Silva TM, Crack PJ, Drummond GR, and Sobey CG. Reduction of cerebral infarct volume by apocynin requires pretreatment and is absent in Nox2-deficient mice. Br J Pharmacol 156: 680–688, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Johnson-Cadwell LI, Jekabsons MB, Wang A, Polster BM, and Nicholls DG. ‘Mild Uncoupling’ does not decrease mitochondrial superoxide levels in cultured cerebellar granule neurons but decreases spare respiratory capacity and increases toxicity to glutamate and oxidative stress. J Neurochem 101: 1619–1631, 2007 [DOI] [PubMed] [Google Scholar]
  • 83.Kahles T. and Brandes RP. NADPH oxidases as therapeutic targets in ischemic stroke. Cell Mol Life Sci 69: 2345–2363, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Kahles T. and Brandes RP. Which NADPH oxidase isoform is relevant for ischemic stroke? The case for nox 2. Antioxid Redox Signal 18: 1400–1417, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kahles T, Luedike P, Endres M, Galla HJ, Steinmetz H, Busse R, Neumann-Haefelin T, and Brandes RP. NADPH oxidase plays a central role in blood-brain barrier damage in experimental stroke. Stroke 38: 3000–3006, 2007 [DOI] [PubMed] [Google Scholar]
  • 86.Kaku DA, Giffard RG, and Choi DW. Neuroprotective effects of glutamate antagonists and extracellular acidity. Science 260: 1516–1518, 1993 [DOI] [PubMed] [Google Scholar]
  • 87.Kase CS, Furlan AJ, Wechsler LR, Higashida RT, Rowley HA, Hart RG, Molinari GF, Frederick LS, Roberts HC, Gebel JM, Sila CA, Schulz GA, Roberts RS, and Gent M. Cerebral hemorrhage after intra-arterial thrombolysis for ischemic stroke: the PROACT II trial. Neurology 57: 1603–1610, 2001 [DOI] [PubMed] [Google Scholar]
  • 88.Kawano T, Kunz A, Abe T, Girouard H, Anrather J, Zhou P, and Iadecola C. iNOS-derived NO and nox2-derived superoxide confer tolerance to excitotoxic brain injury through peroxynitrite. J Cereb Blood Flow Metab 27: 1453–1462, 2007 [DOI] [PubMed] [Google Scholar]
  • 89.Kim GS, Jung JE, Narasimhan P, Sakata H, Yoshioka H, Song YS, Okami N, and Chan PH. Release of mitochondrial apoptogenic factors and cell death are mediated by CK2 and NADPH oxidase. J Cereb Blood Flow Metab 32: 720–730, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Kim GS, Jung JE, Niizuma K, and Chan PH. CK2 is a novel negative regulator of NADPH oxidase and a neuroprotectant in mice after cerebral ischemia. J Neurosci 29: 14779–14789, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Kim HA, Brait VH, Lee S, De Silva TM, Diep H, Eisenhardt A, Drummond GR, and Sobey CG. Brain infarct volume after permanent focal ischemia is not dependent on Nox2 expression. Brain Res 1483: 105–111, 2012 [DOI] [PubMed] [Google Scholar]
  • 92.Kim JA, Neupane GP, Lee ES, Jeong BS, Park BC, and Thapa P. NADPH oxidase inhibitors: a patent review. Expert Opin Ther Pat 21: 1147–1158, 2011 [DOI] [PubMed] [Google Scholar]
  • 93.Kim JI, Lee HR, Sim SE, Baek J, Yu NK, Choi JH, Ko HG, Lee YS, Park SW, Kwak C, Ahn SJ, Choi SY, Kim H, Kim KH, Backx PH, Bradley CA, Kim E, Jang DJ, Lee K, Kim SJ, Zhuo M, Collingridge GL, and Kaang BK. PI3Kgamma is required for NMDA receptor-dependent long-term depression and behavioral flexibility. Nat Neurosci 14: 1447–1454, 2011 [DOI] [PubMed] [Google Scholar]
  • 94.Kimura S, Zhang GX, Nishiyama A, Shokoji T, Yao L, Fan YY, Rahman M, Suzuki T, Maeta H, and Abe Y. Role of NAD(P)H oxidase- and mitochondria-derived reactive oxygen species in cardioprotection of ischemic reperfusion injury by angiotensin II. Hypertension 45: 860–866, 2005 [DOI] [PubMed] [Google Scholar]
  • 95.Kishimoto K, Li RC, Zhang J, Klaus JA, Kibler KK, Dore S, Koehler RC, and Sapirstein A. Cytosolic phospholipase A2 alpha amplifies early cyclooxygenase-2 expression, oxidative stress and MAP kinase phosphorylation after cerebral ischemia in mice. J Neuroinflammation 7: 42, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Kleinschnitz C, Grund H, Wingler K, Armitage ME, Jones E, Mittal M, Barit D, Schwarz T, Geis C, Kraft P, Barthel K, Schuhmann MK, Herrmann AM, Meuth SG, Stoll G, Meurer S, Schrewe A, Becker L, Gailus-Durner V, Fuchs H, Klopstock T, de Angelis MH, Jandeleit-Dahm K, Shah AM, Weissmann N, and Schmidt HH. Post-stroke inhibition of induced NADPH oxidase type 4 prevents oxidative stress and neurodegeneration. PLoS Biol 8: e1000479, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Koh DW, Dawson TM, and Dawson VL. Mediation of cell death by poly(ADP-ribose) polymerase-1. Pharmacol Res 52: 5–14, 2005 [DOI] [PubMed] [Google Scholar]
  • 98.Koponen S, Kurkinen K, Akerman KE, Mochly-Rosen D, Chan PH, and Koistinaho J. Prevention of NMDA-induced death of cortical neurons by inhibition of protein kinase Czeta. J Neurochem 86: 442–450, 2003 [DOI] [PubMed] [Google Scholar]
  • 99.Koziel R, Pircher H, Kratochwil M, Lener B, Hermann M, Dencher NA, and Jansen- Dürr P. Mitochondrial respiratory chain complex I is inactivated by NADPH oxidase Nox4. Biochem J 452: 231–239, 2013 [DOI] [PubMed] [Google Scholar]
  • 100.Kraig RP, Petito CK, Plum F, and Pulsinelli WA. Hydrogen ions kill brain at concentrations reached in ischemia. J Cereb Blood Flow Metab 7: 379–386, 1987 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Kunz A, Park L, Abe T, Gallo EF, Anrather J, Zhou P, and Iadecola C. Neurovascular protection by ischemic tolerance: role of nitric oxide and reactive oxygen species. J Neurosci 27: 7083–7093, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Lafon-Cazal M, Perez V, Bockaert J, and Marin P. Akt mediates the anti-apoptotic effect of NMDA but not that induced by potassium depolarization in cultured cerebellar granule cells. Eur J Neurosci 16: 575–583, 2002 [DOI] [PubMed] [Google Scholar]
  • 103.Lam TI, Brennan-Minella AM, Won S-J, Shen Y, and Swanson RA. Intracellular acidification prevents exitotoxic and ischemic neuronal death by inhibiting NADPH oxidase. Proc Natl Acad Sci U S A 110: 4362–4368, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Lambeth JD. NOX enzymes and the biology of reactive oxygen. Nat Rev Immunol 4: 181–189, 2004 [DOI] [PubMed] [Google Scholar]
  • 105.Lau A. and Tymianski M. Glutamate receptors, neurotoxicity and neurodegeneration. Pflugers Arch 460: 525–542, 2010 [DOI] [PubMed] [Google Scholar]
  • 106.Lee M, You HJ, Cho SH, Woo CH, Yoo MH, Joe EH, and Kim JH. Implication of the small GTPase Rac1 in the generation of reactive oxygen species in response to beta-amyloid in C6 astroglioma cells. Biochem J 366: 937–943, 2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Leevers SJ, Vanhaesebroeck B, and Waterfield MD. Signalling through phosphoinositide 3-kinases: the lipids take centre stage. Curr Opin Cell Biol 11: 219–225, 1999 [DOI] [PubMed] [Google Scholar]
  • 108.Levy R. The role of cytosolic phospholipase A2-alfa in regulation of phagocytic functions. Biochim Biophys Acta 1761: 1323–1334, 2006 [DOI] [PubMed] [Google Scholar]
  • 109.Lipton SA. Failures and successes of NMDA receptor antagonists: molecular basis for the use of open-channel blockers like memantine in the treatment of acute and chronic neurologic insults. NeuroRx 1: 101–110, 2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Liu D, Gharavi R, Pitta M, Gleichmann M, and Mattson MP. Nicotinamide prevents NAD+ depletion and protects neurons against excitotoxicity and cerebral ischemia: NAD+ consumption by SIRT1 may endanger energetically compromised neurons. Neuromol Med 11: 28–42, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Liu Y, Kintner DB, Chanana V, Algharabli J, Chen X, Gao Y, Chen J, Ferrazzano P, Olson JK, and Sun D. Activation of microglia depends on Na+/H+ exchange-mediated H+ homeostasis. J Neurosci 30: 15210–15220, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Lu Q, Wainwright MS, Harris VA, Aggarwal S, Hou Y, Rau T, Poulsen DJ, and Black SM. Increased NADPH oxidase-derived superoxide is involved in the neuronal cell death induced by hypoxia-ischemia in neonatal hippocampal slice cultures. Free Radic Biol Med 53: 1139–1151, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Lucas DR. and Newhouse JP. The toxic effect of sodium L-glutamate on the inner layers of the retina. AMA Arch Ophthalmol 58: 193–201, 1957 [DOI] [PubMed] [Google Scholar]
  • 114.Luo J, Chen H, Kintner DB, Shull GE, and Sun D. Decreased neuronal death in Na+/H+ exchanger isoform 1-null mice after in vitro and in vivo ischemia. J Neurosci 25: 11256–11268, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Malenka RC. and Nicoll RA. NMDA-receptor-dependent synaptic plasticity: multiple forms and mechanisms. Trends Neurosci 16: 521–527, 1993 [DOI] [PubMed] [Google Scholar]
  • 116.Mandir AS, Poitras MF, Berliner AR, Herring WJ, Guastella DB, Feldman A, Poirier GG, Wang ZQ, Dawson TM, and Dawson VL. NMDA but not non-NMDA excitotoxicity is mediated by Poly(ADP-ribose) polymerase. J Neurosci 20: 8005–8011, 2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Mankelow TJ, Pessach E, Levy R, and Henderson LM. The requirement of cytosolic phospholipase A2 for the PMA activation of proton efflux through the N-terminal 230-amino-acid fragment of gp91phox. Biochem J 374: 315–319, 2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Martel MA, Ryan TJ, Bell KF, Fowler JH, McMahon A, Al-Mubarak B, Komiyama NH, Horsburgh K, Kind PC, Grant SG, Wyllie DJ, and Hardingham GE. The subtype of GluN2 C-terminal domain determines the response to excitotoxic insults. Neuron 74: 543–556, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Martini SR. and Kent TA. Hyperglycemia in acute ischemic stroke: a vascular perspective. J Cereb Blood Flow Metab 27: 435–451, 2007 [DOI] [PubMed] [Google Scholar]
  • 120.Martyn KD, Frederick LM, von Loehneysen K, Dinauer MC, and Knaus UG. Functional analysis of Nox4 reveals unique characteristics compared to other NADPH oxidases. Cell Signal 18: 69–82, 2006 [DOI] [PubMed] [Google Scholar]
  • 121.Massaad CA. and Klann E. Reactive oxygen species in the regulation of synaptic plasticity and memory. Antioxid Redox Signal 14: 2013–2054, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Meldrum BS. Excitotoxicity and selective neuronal loss in epilepsy. Brain Pathol 3: 405–412, 1993 [DOI] [PubMed] [Google Scholar]
  • 123.Miyano K. and Sumimoto H. Role of the small GTPase Rac in p22phox-dependent NADPH oxidases. Biochimie 89: 1133–1144, 2007 [DOI] [PubMed] [Google Scholar]
  • 124.Miyawaki T, Mashiko T, Ofengeim D, Flannery RJ, Noh KM, Fujisawa S, Bonanni L, Bennett MV, Zukin RS, and Jonas EA. Ischemic preconditioning blocks BAD translocation, Bcl-xL cleavage, and large channel activity in mitochondria of postischemic hippocampal neurons. Proc Natl Acad Sci U S A 105: 4892–4897, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Morgan D, Cherny VV, Murphy R, Katz BZ, and DeCoursey TE. The pH dependence of NADPH oxidase in human eosinophils. J Physiol 569: 419–431, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Morikawa E, Mori H, Kiyama Y, Mishina M, Asano T, and Kirino T. Attenuation of focal ischemic brain injury in mice deficient in the epsilon1 (NR2A) subunit of NMDA receptor. J Neurosci 18: 9727–9732, 1998 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Moriyoshi K, Masu M, Ishii T, Shigemoto R, Mizuno N, and Nakanishi S. Molecular cloning and characterization of the rat NMDA receptor. Nature 354: 31–37, 1991 [DOI] [PubMed] [Google Scholar]
  • 128.Moroni F. Poly(ADP-ribose)polymerase 1 (PARP-1) and postischemic brain damage. Curr Opin Pharmacol 8: 96–103, 2008 [DOI] [PubMed] [Google Scholar]
  • 129.Munnamalai V. and Suter DM. Reactive oxygen species regulate F-actin dynamics in neuronal growth cones and neurite outgrowth. J Neurochem 108: 644–661, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Myers RE. and Yamaguchi S. Nervous system effects of cardiac arrest in monkeys. Preservation of vision. Arch Neurol 34: 65–74, 1977 [DOI] [PubMed] [Google Scholar]
  • 131.Nakazawa T, Komai S, Tezuka T, Hisatsune C, Umemori H, Semba K, Mishina M, Manabe T, and Yamamoto T. Characterization of Fyn-mediated tyrosine phosphorylation sites on GluR epsilon 2 (NR2B) subunit of the N-methyl-D-aspartate receptor. J Biol Chem 276: 693–699, 2001 [DOI] [PubMed] [Google Scholar]
  • 132.Newell DW, Barth A, Papermaster V, and Malouf AT. Glutamate and non-glutamate receptor mediated toxicity caused by oxygen and glucose deprivation in organotypic hippocampal cultures. J Neurosci 15: 7702–7711, 1995 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Nicholls DG. Simultaneous monitoring of ionophore- and inhibitor-mediated plasma and mitochondrial membrane potential changes in cultured neurons. J Biol Chem 281: 14864–14874, 2006 [DOI] [PubMed] [Google Scholar]
  • 134.Nito C, Kamada H, Endo H, Niizuma K, Myer DJ, and 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 28: 1686–1696, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Olney JW, Ikonomidou C, Mosinger JL, and Frierdich G. MK-801 prevents hypobaric-ischemic neuronal degeneration in infant rat brain. J Neurosci 9: 1701–1704, 1989 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Olney JW, Labruyere J, Wang G, Wozniak DF, Price MT, and Sesma MA. NMDA antagonist neurotoxicity: mechanism and prevention. Science 254: 1515–1518, 1991 [DOI] [PubMed] [Google Scholar]
  • 137.Ozyurt E, Graham DI, Woodruff GN, and McCulloch J. Protective effect of the glutamate antagonist, MK-801 in focal cerebral ischemia in the cat. J Cereb Blood Flow Metab 8: 138–143, 1988 [DOI] [PubMed] [Google Scholar]
  • 138.Pacher P, Beckman JS, and Liaudet L. Nitric oxide and peroxynitrite in health and disease. Physiol Rev 87: 315–424, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Pamenter ME, Ali SS, Tang Q, Finley JC, Gu XQ, Dugan LL, and Haddad GG. An in vitro ischemic penumbral mimic perfusate increases NADPH oxidase-mediated superoxide production in cultured hippocampal neurons. Brain Res 1452: 165–172, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Papadia S, Stevenson P, Hardingham NR, Bading H, and Hardingham GE. Nuclear Ca2+ and the cAMP response element-binding protein family mediate a late phase of activity-dependent neuroprotection. J Neurosci 25: 4279–4287, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Park CK, Nehls DG, Graham DI, Teasdale GM, and McCulloch J. The glutamate antagonist MK-801 reduces focal ischemic brain damage in the rat. Ann Neurol 24: 543–551, 1988 [DOI] [PubMed] [Google Scholar]
  • 142.Park WS, Chang YS, and Lee M. Effects of hyperglycemia or hypoglycemia on brain cell membrane function and energy metabolism during the immediate reoxygenation-reperfusion period after acute transient global hypoxia-ischemia in the newborn piglet. Brain Res 901: 102–108, 2001 [DOI] [PubMed] [Google Scholar]
  • 143.Parsons MW, Barber PA, Desmond PM, Baird TA, Darby DG, Byrnes G, Tress BM, and Davis SM. Acute hyperglycemia adversely affects stroke outcome: a magnetic resonance imaging and spectroscopy study. Ann Neurol 52: 20–28, 2002 [DOI] [PubMed] [Google Scholar]
  • 144.Peppelenbosch MP, Qiu RG, de Vries-Smits AM, Tertoolen LG, de Laat SW, McCormick F, Hall A, Symons MH, and Bos JL. Rac mediates growth factor-induced arachidonic acid release. Cell 81: 849–856, 1995 [DOI] [PubMed] [Google Scholar]
  • 145.Petito CK, Kraig RP, and Pulsinelli WA. Light and electron microscopic evaluation of hydrogen ion-induced brain necrosis. J Cereb Blood Flow Metab 7: 625–632, 1987 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Plum F. What causes infarction in ischemic brain?: The Robert Wartenberg Lecture. Neurology 33: 222–223, 1983 [DOI] [PubMed] [Google Scholar]
  • 147.Radermacher KA, Wingler K, Langhauser F, Altenhofer S, Kleikers P, Hermans JJ, Hrabe de Angelis M, Kleinschnitz C, and Schmidt HH. Neuroprotection after stroke by targeting NOX4 as a source of oxidative stress. Antioxid Redox Signal 18: 1418–1427, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Ramsey IS, Ruchti E, Kaczmarek JS, and Clapham DE. Hv1 proton channels are required for high-level NADPH oxidase-dependent superoxide production during the phagocyte respiratory burst. Proc Natl Acad Sci U S A 106: 7642–7647, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Raz L, Zhang QG, Zhou CF, Han D, Gulati P, Yang LC, Yang F, Wang RM, and Brann DW. Role of Rac1 GTPase in NADPH oxidase activation and cognitive impairment following cerebral ischemia in the rat. PLoS One 5: e12606, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Reyes RC, Brennan AM, Shen Y, Baldwin Y, and Swanson RA. Activation of neuronal NMDA receptors induces superoxide-mediated oxidative stress in neighboring neurons and astrocytes. J Neurosci 32: 12973–12978, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Reynolds IJ. and Hastings TG. Glutamate induces the production of reactive oxygen species in cultured forebrain neurons following NMDA receptor activation. J Neurosci 15: 3318–3327, 1995 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Ribo M, Molina C, Montaner J, Rubiera M, Delgado-Mederos R, Arenillas JF, Quintana M, and Alvarez-Sabin J. Acute hyperglycemia state is associated with lower tPA-induced recanalization rates in stroke patients. Stroke 36: 1705–1709, 2005 [DOI] [PubMed] [Google Scholar]
  • 153.Rossi DJ, Oshima T, and Attwell D. Glutamate release in severe brain ischaemia is mainly by reversed uptake. Nature 403: 316–321, 2000 [DOI] [PubMed] [Google Scholar]
  • 154.Rothstein JD. Current hypotheses for the underlying biology of amyotrophic lateral sclerosis. Ann Neurol 65Suppl 1: S3–S9, 2009 [DOI] [PubMed] [Google Scholar]
  • 155.Ruscher K, Freyer D, Karsch M, Isaev N, Megow D, Sawitzki B, Priller J, Dirnagl U, and Meisel A. Erythropoietin is a paracrine mediator of ischemic tolerance in the brain: evidence from an in vitro model. J Neurosci 22: 10291–10301, 2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Rytter A, Cronberg T, Asztely F, Nemali S, and Wieloch T. Mouse hippocampal organotypic tissue cultures exposed to in vitro “ischemia” show selective and delayed CA1 damage that is aggravated by glucose. J Cereb Blood Flow Metab 23: 23–33, 2003 [DOI] [PubMed] [Google Scholar]
  • 157.Sattler R, Charlton MP, Hafner M, and Tymianski M. Distinct influx pathways, not calcium load, determine neuronal vulnerability to calcium neurotoxicity. J Neurochem 71: 2349–2364, 1998 [DOI] [PubMed] [Google Scholar]
  • 158.Sattler R, Xiong Z, Lu WY, Hafner M, MacDonald JF, and Tymianski M. Specific coupling of NMDA receptor activation to nitric oxide neurotoxicity by PSD-95 protein. Science 284: 1845–1848, 1999 [DOI] [PubMed] [Google Scholar]
  • 159.Shi Y, Chanana V, Watters JJ, Ferrazzano P, and Sun D. Role of sodium/hydrogen exchanger isoform 1 in microglial activation and proinflammatory responses in ischemic brains. J Neurochem 119: 124–135, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Simchowitz L. Intracellular pH modulates the generation of superoxide radicals by human neutrophils. J Clin Invest 76: 1079–1089, 1985 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Simon RP, Niro M, and Gwinn R. Brain acidosis induced by hypercarbic ventilation attenuates focal ischemic injury. J Pharmacol Exp Ther 267: 1428–1431, 1993 [PubMed] [Google Scholar]
  • 162.Simon RP, Swan JH, Griffiths T, and Meldrum BS. Blockade of N-methyl-D-aspartate receptors may protect against ischemic damage in the brain. Science 226: 850–852, 1984 [DOI] [PubMed] [Google Scholar]
  • 163.Simpson MA, Dewey HM, Churilov L, Ahmed N, Bladin CF, Schultz D, Markus R, Sturm JW, Levi CR, Blacker DJ, Jannes J, Lindley RI, and Parsons MW. Thrombolysis for acute stroke in Australia: outcomes from the Safe Implementation of Thrombolysis in Stroke registry (2002–2008). Med J Aust 193: 439–443, 2010 [DOI] [PubMed] [Google Scholar]
  • 164.Soto D, Pancetti F, Marengo JJ, Sandoval M, Sandoval R, Orrego F, and Wyneken U. Protein kinase CK2 in postsynaptic densities: phosphorylation of PSD-95/SAP90 and NMDA receptor regulation. Biochem Biophys Res Commun 322: 542–550, 2004 [DOI] [PubMed] [Google Scholar]
  • 165.Stanika RI, Villanueva I, Kazanina G, Andrews SB, and Pivovarova NB. Comparative impact of voltage-gated calcium channels and NMDA receptors on mitochondria-mediated neuronal injury. J Neurosci 32: 6642–6650, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Steinberg GK, George CP, DeLaPaz R, Shibata DK, and Gross T. Dextromethorphan protects against cerebral injury following transient focal ischemia in rabbits. Stroke 19: 1112–1118, 1988 [DOI] [PubMed] [Google Scholar]
  • 167.Steinberg GK, Panahian N, Perez-Pinzon MA, Sun GH, Modi MW, and Sepinwall J. Narrow temporal therapeutic window for NMDA antagonist protection against focal cerebral ischaemia. Neurobiol Dis 2: 109–118, 1995 [DOI] [PubMed] [Google Scholar]
  • 168.Steinberg GK, Saleh J, Kunis D, DeLaPaz R, and Zarnegar SR. Protective effect of N-methyl-D-aspartate antagonists after focal cerebral ischemia in rabbits. Stroke 20: 1247–1252, 1989 [DOI] [PubMed] [Google Scholar]
  • 169.Stolk J, Hiltermann TJ, Dijkman JH, and Verhoeven AJ. Characteristics of the inhibition of NADPH oxidase activation in neutrophils by apocynin, a methoxy-substituted catechol. Am J Respir Cell Mol Biol 11: 95–102, 1994 [DOI] [PubMed] [Google Scholar]
  • 170.Stout AK, Raphael HM, Kanterewicz BI, Klann E, and Reynolds IJ. Glutamate-induced neuron death requires mitochondrial calcium uptake. Nat Neurosci 1: 366–373, 1998 [DOI] [PubMed] [Google Scholar]
  • 171.Suh SW, Gum ET, Hamby AM, Chan PH, and Swanson RA. Hypoglycemic neuronal death is triggered by glucose reperfusion and activation of neuronal NADPH oxidase. J Clin Invest 117: 910–918, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Suh SW, Shin BS, Ma H, Van Hoecke M, Brennan AM, Yenari MA, and Swanson RA. Glucose and NADPH oxidase drive neuronal superoxide formation in stroke. Ann Neurol 64: 654–663, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Sundaresan M, Yu ZX, Ferrans VJ, Sulciner DJ, Gutkind JS, Irani K, Goldschmidt-Clermont PJ, and Finkel T. Regulation of reactive-oxygen-species generation in fibroblasts by Rac1. Biochem J 318 (Pt 2): 379–382, 1996 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Swan JH. and Meldrum BS. Protection by NMDA antagonists against selective cell loss following transient ischaemia. J Cereb Blood Flow Metab 10: 343–351, 1990 [DOI] [PubMed] [Google Scholar]
  • 175.Takizawa S, Fukuyama N, Hirabayashi H, Nakazawa H, and Shinohara Y. Reperfusion enhances nitrotyrosine formation in rat focal cerebral ischemia. J Stroke Cerebrovasc Dis 12: 196–200, 2003 [DOI] [PubMed] [Google Scholar]
  • 176.Tang CM, Dichter M, and Morad M. Modulation of the N-methyl-D-aspartate channel by extracellular H+. Proc Natl Acad Sci U S A 87: 6445–6449, 1990 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Tang J, Liu J, Zhou C, Ostanin D, Grisham MB, Neil Granger D, and Zhang JH. Role of NADPH oxidase in the brain injury of intracerebral hemorrhage. J Neurochem 94: 1342–1350, 2005 [DOI] [PubMed] [Google Scholar]
  • 178.Tang XN, Cairns B, Cairns N, and Yenari MA. Apocynin improves outcome in experimental stroke with a narrow dose range. Neuroscience 154: 556–562, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Tombaugh GC. and Sapolsky RM. Mild acidosis protects hippocampal neurons from injury induced by oxygen and glucose deprivation. Brain Res 506: 343–345, 1990 [DOI] [PubMed] [Google Scholar]
  • 180.Traynelis SF. and Cull-Candy SG. Proton inhibition of N-methyl-D-aspartate receptors in cerebellar neurons. Nature 345: 347–350, 1990 [DOI] [PubMed] [Google Scholar]
  • 181.Traynelis SF, Wollmuth LP, McBain CJ, Menniti FS, Vance KM, Ogden KK, Hansen KB, Yuan H, Myers SJ, and Dingledine R. Glutamate receptor ion channels: structure, regulation, and function. Pharmacol Rev 62: 405–496, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Trevani AS, Andonegui G, Giordano M, Lopez DH, Gamberale R, Minucci F, and Geffner JR. Extracellular acidification induces human neutrophil activation. J Immunol 162: 4849–4857, 1999 [PubMed] [Google Scholar]
  • 183.Venables GS, Miller SA, Gibson G, Hardy JA, and Strong AJ. The effects of hyperglycaemia on changes during reperfusion following focal cerebral ischaemia in the cat. J Neurol Neurosurg Psychiatry 48: 663–669, 1985 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Vyklicky L, Jr., Vlachova V, and Krusek J. The effect of external pH changes on responses to excitatory amino acids in mouse hippocampal neurones. J Physiol 430: 497–517, 1990 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Waite K. and Eickholt BJ. The neurodevelopmental implications of PI3K signaling. Curr Top Microbiol Immunol 346: 245–265, 2010 [DOI] [PubMed] [Google Scholar]
  • 186.Walder CE, Green SP, Darbonne WC, Mathias J, Rae J, Dinauer MC, Curnutte JT, and Thomas GR. Ischemic stroke injury is reduced in mice lacking a functional NADPH oxidase. Stroke 28: 2252–2258, 1997 [DOI] [PubMed] [Google Scholar]
  • 187.Wang Q, Sun AY, Simonyi A, Kalogeris TJ, Miller DK, Sun GY, and Korthuis RJ. Ethanol preconditioning protects against ischemia/reperfusion-induced brain damage: role of NADPH oxidase-derived ROS. Free Radic Biol Med 43: 1048–1060, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Wang S, Xing Z, Vosler PS, Yin H, Li W, Zhang F, Signore AP, Stetler RA, Gao Y, and Chen J. Cellular NAD replenishment confers marked neuroprotection against ischemic cell death: role of enhanced DNA repair. Stroke 39: 2587–2595, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Wang YB, Wang JJ, Wang SH, Liu SS, Cao JY, Li XM, Qiu S, and Luo JH. Adaptor protein APPL1 couples synaptic NMDA receptor with neuronal prosurvival phosphatidylinositol 3-kinase/Akt pathway. J Neurosci 32: 11919–11929, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Won SJ, Tang XN, Suh SW, Yenari MA, and Swanson RA. Hyperglycemia promotes tissue plasminogen activator-induced hemorrhage by Increasing superoxide production. Ann Neurol 70: 583–590, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Wu LJ, Wu G, Akhavan Sharif MR, Baker A, Jia Y, Fahey FH, Luo HR, Feener EP, and Clapham DE. The voltage-gated proton channel Hv1 enhances brain damage from ischemic stroke. Nat Neurosci 15: 565–573, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Xiong ZG, Zhu XM, Chu XP, Minami M, Hey J, Wei WL, MacDonald JF, Wemmie JA, Price MP, Welsh MJ, and Simon RP. Neuroprotection in ischemia: blocking calcium-permeable acid-sensing ion channels. Cell 118: 687–698, 2004 [DOI] [PubMed] [Google Scholar]
  • 193.Yoshioka H, Niizuma K, Katsu M, Okami N, Sakata H, Kim GS, Narasimhan P, and Chan PH. NADPH oxidase mediates striatal neuronal injury after transient global cerebral ischemia. J Cereb Blood Flow Metab 31: 868–880, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Zhang J, Barasch N, Li RC, and Sapirstein A. Inhibition of cytosolic phospholipase A(2) alpha protects against focal ischemic brain damage in mice. Brain Res 1471: 129–137, 2012 [DOI] [PubMed] [Google Scholar]
  • 195.Zhang QG, Raz L, Wang R, Han D, De Sevilla L, Yang F, Vadlamudi RK, and Brann DW. Estrogen attenuates ischemic oxidative damage via an estrogen receptor alpha-mediated inhibition of NADPH oxidase activation. J Neurosci 29: 13823–13836, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Zorov DB, Juhaszova M, and Sollott SJ. Mitochondrial ROS-induced ROS release: an update and review. Biochim Biophys Acta 1757: 509–517, 2006 [DOI] [PubMed] [Google Scholar]

Articles from Antioxidants & Redox Signaling are provided here courtesy of Mary Ann Liebert, Inc.

RESOURCES