<<

Neurol Sci (2012) 33:223–237 DOI 10.1007/s10072-011-0828-5

REVIEW ARTICLE

The role of glutamate in neuronal ischemic injury: the role of spark in fire

Botros B. Kostandy

Received: 7 June 2010 / Accepted: 20 October 2011 / Published online: 2 November 2011 Ó Springer-Verlag 2011

Abstract Although being a physiologically important cellular and synaptic functions, cell death and survival, excitatory neurotransmitter, glutamate plays a pivotal role motor functions, learning and memory [1]. Its level in in various neurological disorders including ischemic neu- mammalian brain is 1,000 times more than other important rological diseases. Its level is increased during cerebral neurotransmitters such as dopamine or serotonin. Although ischemia with excessive neurological stimulation causing glutamate has important physiological functions in the the glutamate-induced neuronal toxicity, excitotoxicity, CNS it is suggested to take part in the pathophysiology of and this is considered the triggering spark in the ischemic many disease processes such as epilepsy, neurodegenera- neuronal damage. The stimulation will lead tive disorders and cerebrovascular [2]. Playing an to rise in the intracellular sodium and calcium, and the important role in ischemic neuronal damage, this review elevated intracellular calcium will lead to mitochondrial focuses on the mechanisms of injurious effect of glutamate dysfunction, activation of proteases, accumulation of during ischemia and possible therapeutic approaches. reactive oxygen species and release of . Inter- ruption of the cascades of glutamate-induced cell death during ischemia may provide a way to prevent, or at least Synthesis, release and breakdown of glutamate reduce, the ischemic damage. Various therapeutic options are suggested interrupting the glutamatergic pathways, e.g., Glutamate is not synthesized in neurons nor acquired from inhibiting the glutamate synthesis or release, increasing its circulation [3], but rather synthesized in astrocytes through clearance, blocking of its receptors or preventing the rise in the intermediate metabolite . The process of intracellular calcium. Development of these strategies may glutamate synthesis and breakdown can be summarized in provide future treatment options in the management of Fig. 1. Activated cells release glutamate through hemi- ischemic stroke. channels of gap junctions [4]. The released glutamate produced by the depolarization of synaptosomes consists of Keywords Glutamate Á Excitotoxicity Á two components. A physiologically relevant Ca2?-depen- Cerebral ischemia Á Neuroprotectants dent component is produced by the exocytosis of synaptic vesicles containing glutamate. N-type Ca2? channels, a type of voltage-dependent Ca2? channels (VDCCs), are Introduction principally responsible for release of synaptic glutamate in the most of the CNS [5]. The other component, Ca2?- Glutamate is one of the major excitatory neurotransmitters independent one, can be attributed to the reversal of the in the central nervous system (CNS). It controls various glutamate transporter [6]. The released glutamate is effectively removed by five different Na?-dependent high- affinity glutamate transporters [7]: EAAT 1 (GLAST), & B. B. Kostandy ( ) EAAT2 (GLT-1), EAAT3 (EAAC1) and EAAT4 and Department of Pharmacology, Faculty of Medicine, University of Assiut, Assiut 71526, Egypt EAAT5. EAAT2 has the predominant role in clearing e-mail: [email protected] glutamate throughout the neuroaxis [8]. 123 224 Neurol Sci (2012) 33:223–237

Astrocyte Glutametergic NMDARs have been shown to be organized into multi- neuron protein signaling complexes within a specialized structure located beneath the postsynaptic membrane aligned with Glutamine Glutamine Glutamine active zones of presynaptic terminals with the CNS, known as the postsynaptic density (PSD) [15]. PSDs are involved in

Glutamine Glutaminase several functions including: cell-to-cell adhesion, regulation synthase of receptor clustering and modulation of receptor function. Glutamate Glutamate Glutamate Of the proteins involved in the PSD, PSD-95 plays a prom- inent organizational role by coupling the NR2 subunits of aminotransferase NMDARs to intracellular proteins and signaling enzymes such as neuronal nitric oxide synthase (nNOS) [16, 17]. α-ketoglutaric AMPARs are composed of four subunits GluR1–4 or A–D subunits encoded by four genes [18]. The perme- Fig. 1 Schematic representation of the process of glutamate ability to Ca2? depends on the presence or absence of a synthesis and glutamate-glutamine cycle between neurons and astrocytes (arrow release, dashed arrow enzymatic conversion, dot- GluR2 subunit. Its presence renders an AMPAR channel ted arrow uptake) impermeable to Ca2?. Activation of AMPARs after ischemia allows Na? influx which leads to the depolar- ization of the neuron and subsequent influx of Ca2? Glutamate receptors through Ca2? channels. Drugs interfering with the function of NMDAR and AMPAR are summarized in Table 1. Glutamate acts on four different postsynaptic receptors: Molecular cloning of Kainate receptors (KARs) has NMDA (N-methyl D-aspartate), AMPA (a-amino-3- identified five subtypes: GluK1-5, which co-assemble in hydroxy-5-methyl-4-isoxazole propionic acid), kainate and various combinations to form functional receptors. KAR metabotropic receptors. The three former receptors are subunits that were formally known as GluR5–7 (or linked to membrane ion channels, whereas metabotropic GLUK5–7) are now named GluK1–3 and those previously receptors are coupled with a G-protein. called KA1 and KA2 (or GLUK1 and GLUK2) are now NMDA receptors (NMDARs) are composed of seven named GluK4 and GluK5 [19]. Functional studies suggest NMDAR subunits have been identified so far: one NR1, that KARs mainly have a modulatory role in synaptic four NR2 (A–D), and two NR3 (A and B). Their functional transmission (excitatory and inhibitory) rather than being characteristics are determined by the receptors specific the major postsynaptic target for synaptically released combination of NR1 and NR2 subunits. Most native glutamate as is the case for NMDARs and AMPARs [20]. NMDARs appear to function as heterotetrameric assem- KARs regulate glutamate release. They can function as blies composed of two -binding NR1 and two glu- facilitatory autoreceptors or inhibitory autoreceptors during tamate-binding NR2 subunits [9]. NR2A containing repetitive synaptic activation and can respond to ambient receptors are characterized by faster decay times than levels of L-glutamate to provide a tonic regulation of 2? NR2B-containing ones, as evidenced by different Ca L-glutamate release. KARs contribute to excitatory synap- influx [10]. Moreover, in mature cultured neurons, NR2A- tic transmission at certain synapses. containing NMDARs promote trafficking of the AMPA Metabotropic receptors (mGluRs) are grouped to three receptors (AMPARs) subunit GluR1 while NR2B-con- families, group I (mGluR1 and mGluR5), II (mGluR2 and taining NMDARs inhibit such trafficking [11]. NR3 sub- mGluR3) and III (mGluR4–mGluR8). Group I receptors units reduce the amplitude and dramatically decrease the are linked to phospholipase C and localized in the post- Ca2? permeability of NMDAR-associated channels and it synaptic density area at excitatory synaptic sites. Group I is suggested that endogenous NR3A protects neurons [12]. mGluRs may also have effects on NMDA signaling [21]. There are also modulatory sites on the NMDAR, including They potentiate NMDA-mediated neurotoxicity, increase those for glycine, Mg2? and polyamines. arachidonic acid release and inflammation [22]. Groups II The subcellular localization of NMDARs also affects and III, primarily localized presynaptically, are negatively the nature of NMDAR signaling. Evidence suggests that linked to adenylate cyclase [23]. They are blocked by synaptic NMDAR activity is extremely important for various agents, including for example alpha-methyl- neuronal survival while the extrasynaptic NMDARs are cyclopropyl glycine (MCCG) [24] and a-cyclopropyl- coupled to cell-death pathways [13]. Extrasynaptic 4-phosphonophenylglycine (CPPG) [25], respectively. NMDARs oppose synaptic NMDARs by triggering cAMP- Activation of these receptors results in feedback inhibition responsive element-binding protein (CREB) shutoff and of glutamate release, through the inhibition of voltage- cell-death pathways [14]. gated Ca2? entry into the cell. 123 Neurol Sci (2012) 33:223–237 225

Table 1 NMDAR and AMBAR blockers NMDAR blockers AMPAR blockers Competitive Noncompetitive Glycine site Polyamine site Competitive Noncompetitive blockers blockers blockers blockers antagonists antagonists

AP-7 Cerestat ACEA 1021 NBQX GYKI 52466 CGP 37849 Liprodil PNQX SYM2206 CGP 39551 CGS 19755 HA966 CPP L-687,414 Midafotel , (CPPene) ACEA 1021 5-nitro-6,7-dichloro-1,4-dihydro-2,3-, AP-7 2-amino-7-phosphonoheptanoic acid, CGP 37849 (E)-(±)-2-amino-4- methyl-5-phosphono-3-pentenoic acid, CGP 39551 (E)-(±)-2-amino-4-methyl-5-phosphono-3-pentenoic acid ethyl ester, CPP 3-(2-carbo- xypiperazin-4-yl)propyl-1-phosphonic acid, CGS 19755 (cis-4-phosphonomethyl-2-piperidine carboxylic acid), GYKI 52466 1-(4-aminophenyl)- 4-methyl-7,8-methylenedioxy-5H-2,3-benzodiazepine, HA966 3-amino-1-hydroxypyrrolid-2-one, L-687,414 3R(?)cis-4-methyl-pyrrollid-2-one, NBQX 2,3-dihydroxy-6-nitro-7-sulfamoylbenzo[f]quinoxaline-2,3-dione, PNQX 9-methyl-amino-6-nitro-hexahydrobenzo(F)quinoxalinedione, SYM2206 4-(4-aminophenyl)-1,2-dihydro-1-methyl-2-propylcarbamoyl-6,7-methylenedioxyphthalazine

Elevation of glutamate level during ischemia substrate is enhanced by depolarization-stimulated Ca2? entry, which results in the facilitation of glutamate release Normally, neurons are exposed to small, transient impulses [36]. The rise in extracellular glutaminase, released from of glutamate due to efficient uptake removal mechanisms damaged neurons will increase the hydrolysis of glutamine, [26], but neurons are subjected to excessive glutamatergic with the formation of extracellular glutamate [37]. stimulation during ischemia due to the elevation of extra- Ischemia with subsequent poverty of ATP leads to col- cellular glutamate with overstimulation of its receptors, lapse of Na?/K? electrochemical gradient. As a result, the and this phenomenon has been recognized since the 1980s glutamate transporters operate in a reverse direction, and [27]. Ischemia-induced elevations in glutamate occur in all this process is suggested by some authors to be the main brain areas which have been investigated and in response to mechanism responsible for glutamate accumulation [38] all experimental approaches employed to provide low and the excess extracellular glutamate is the end result. The levels of cerebral blood flow [28]. Both focal and global expression of the glutamate transporters EAAT1 and cerebral ischemia cause rise in extracellular glutamate EAAT2 in astrocytes is rapidly altered following hypoxic- levels [29]. Glutamate levels return to normal shortly after ischemic injury [39], and the reduced expression of these reperfusion has been initiated [30]. transporters leading to the accumulation of their substrate The release of glutamate occur when blood supply to extracellularly [40]. neurons decrease below certain level (threshold relation- Although the glutamate physiological antidote, gamma ship). The neurotransmitter is released at a blood flow amino butyric acid (GABA), increase extracellularly in below 20 ml/100gm/min [31]. It is worth mentioning that response to ischemia, this increase is not as large as that of neuronal necrosis occur at a blood flow of 17 ml/100gm/ glutamate and does not persist as long [41]. Since gluta- min maintained for 180 min or more [32]. mate uptake via EAAC1 has been posited to provide a The increased formation and release of glutamate from source for GABA synthesis, decreases in EAAC1 may lead glutamine in intact glutamatergic neurons has been sug- to decreased GABA-ergic activity. The resultant increased gested to account for the increased extracellular glutamate extracellular glutamate and relative poverty in GABA may [33]. Reduction in adenosine triphosphate (ATP), resulting lead to overexcitation and neuronal death [39]. from anoxia, causes failure in the energy-mediated pro- cesses in the cell, e.g., Na? pumps [34], in addition to swelling-induced opening of anion channels [35], lead to Glutamate-induced cell injury accumulation of ions inside the neurons with facilitation of cellular membrane depolarization. This is followed by The concept that glutamate is a potent neurotoxin has long exocytosis and expulsion of the glutamate loaded in been recognized for more than half centaury [42]. It is intracellular vesicles. Protein kinase C (PKC)-dependent responsible for toxic neuronal death of postsynaptic neu- phosphorylation of myristoylated -rich C kinase rons. Neuronal death in cell culture occurs at doses above 123 226 Neurol Sci (2012) 33:223–237

10 lM[43]. Prolonged and excessive stimulation of influx, resulting in cytotoxic edema [48]. NMDARs play glutamatergic receptors occur when extracellular glutamate a key role in mediating at least some aspects of gluta- approaches 100 lM[44]. Even ambient glutamate con- mate neurotoxicity [49]. Many studies have referred centrations may be neurotoxic in energy-deprived cells to NMDAR-triggered, Ca2?-mediated excitotoxicity as [45]. This action has been denominated excitotoxicity and ‘‘rapidly triggered’’, emphasizing the speed at which it can occurs as a consequence of a prolonged or a strong acti- occur. In cortical cell cultures, 3–5 min of sustained vation of glutamate postsynaptic receptors [46]. The NMDAR activation is sufficient to destroy most neurons. pathophysiology of several acute or chronic neurological In contrast, AMPAR-triggered, Ca2?-mediated excitotox- disorders has been linked to excitotoxicity [1, 47]. icity typically occurs more slowly, requiring hours of The build-up of glutamate results in overstimulation of sustained receptor activation to induce lethal injury in the AMPARs, KARs and NMDARs on neurons, with conse- same cell cultures [50]. The whole story of ischemia- quent influx of Na? and Ca2? ions through the channels triggered glutamate release and excitotoxicity can be gated by these receptors. Water passively follows the ion summarized in Fig. 2.

Ischemia

Energy failure Ion pump failure Reduction of transporter s’ function and number

Reversal of Na+ Accumulation of intracellular Ca2+ exchanger ions i.e. Na+ Activation of Na+/ H+ Reduced glutamate clearance exchanger Membrane depolarization

Excessive release of glutamate

Glutamate accumulation

Excessive glutamatergic receptor activation

Metabotropic receptors I

Phospholipase C ATP NMDAR Increase in AMPAR/ KAR activation depletion and activation IP3 activation energy failure

Rise in intracellular Ca2+ Activation. of Membrane. Rise in + Activation of VDCC depolarization intracellular Na cPLA2 and cyclooxygenase Rise in Activation of mitochondrial mtNOS Activation nNOS Ca2+

Excess entry of water Mitochondrial Formation dysfunction of peroxy- Release of NO Generation of nitrite superoxide and toxic prostanoids Oxidative stress Release of Calapain Formation of Cytotoxic oedema cytochrome c activation nitrotyrosine adducts and DNA damage

Degradation of Activation of cytoskeletal and caspase-3 extracellular matrix Release of AIF proteins Neuronal cell death

DNA fragmentation

Fig. 2 Summary of role of glutamate during neuronal ischemia (mtNOS mitochondrial NOS, cPLA2 cytosolic phospholipase 2, IP3 inositol triphosphate, AIF apoptosis-inducing factor) 123 Neurol Sci (2012) 33:223–237 227

The role of elevated intracellular Ca21 bisphosphate. TRPM7 also contributes to increase in intracellular sodium content during ischemia [67]. The rise in intracellular Ca2? seems to play a major role in the pathological events following excitotoxicity [51]. NMDAR activation is the major source of Ca2? intracel- Mitochondrial dysfunction lularly in normal and ischemic cells [52]. The rise in intracellular Ca2? occurs as a consequence of direct entry Mitochondrial dysfunction is a consequent of rise in through NMDARs and VDCCs, which are activated due to intracellular Ca2? [68]. The rise in intracellular Ca2? above membrane depolarization secondary to NMDAR activation certain threshold (*0.5 lM) will increase its uptake into [53]. Of the VDCCs, the L-type mediates long-lasting Ca2? mitochondria [69]. The mitochondrial Ca2? content currents in response to depolarization in excitable cells increases from 1 to 3 nmol/mg protein to 6–9 nmol/mg [54]. These inward Ca2? currents are considered to be an protein after 24 h of reperfusion [70]. This rise causes important mediator of excitotoxicity and neuronal damage devastating effects, including opening of the mitochondrial [55]. A recent research, surprisingly, showed that certain permeability transition pore, complete loss of respiration, types of L-type VDCC in retinal neurons are subjected to and release of both NADH and cytochrome c [71]. rapid glutamate-induced internalization, serving as a pro- Impairment of mitochondrial function and collapse of tective negative feedback mechanism [56]. mGluRs can mitochondrial membrane potential lead to failing mainte- also raise intracellular Ca2? via the inositol triphosphate- nance of anti-oxidant pathways and generation of ROS (see mediated release of Ca2? from the endoplasmic reticulum below), and inhibition of Ca2? rise intracellularly will [57]. inhibit this cascade [72]. Loss of mitochondrial NADH, Other mechanisms may be responsible for the rise in which is necessary for the numerous dehydrogenases intracellular Ca2? following ischemia. The increased present within the mitochondrial matrix, occurs as a result activity of Na?/H? exchanger triggered by cellular ische- of the opening of the permeability transition pore (PTP). mia, with subsequent accumulation of Na? intracellularly, The PTP is activated by abnormally high concentrations of will secondarily activate Na?/Ca2? exchanger (NCX), that Ca2?, oxidative stress [73] and low ATP, conditions that will work in a reverse manner, leading to increased Na? are present during and immediately following hypoxic expulsion in exchange with Ca2? [58]. This reversal mode ischemia [74]. Contribution of PTP opening to ischemic of NCX seems to be responsible for the early rise in and traumatic brain injury is supported by the neuropro- intracellular Ca2?. It should be stated that Na?/H? tection observed with PTP inhibitors, e.g., cyclosporins exchanger activation seems to be subsequent to glutamate [75], which bind to cyclophilin D, the one well-established stimulation (rather than being directly activated by ische- protein associated with pore opening. Cyclophilin D mia), as toxic glutamate doses causes cellular acidification knockout mice are resistant to ischemic brain injury [76]. and H? overload, with increased Na?/H? exchange [59]. Respiratory inhibition stimulates lactate dehydrogenase This is supported by the recent evidences of the neuro- (LDH), resulting in lactic acidosis, which further inhibits protective effect of Na?/H? exchanger blockers against oxidative phosphorylation and promotes oxidative stress glutamate-induced neuronal damage [60]. Ca2? may be [77]. Mitochondrial dysfunction will result in release of also released from intracellular stores due to stimulation of cytochrome c through Bax or Bak megapores present in the ryanodine receptors [61]. outer membrane. The cytochrome c, once released, inter- The initial rise in Ca2? precedes a second wave, and so acts with deoxyadenosine triphosphate and binds to Apaf-1 the rise in the intracellular Ca2? occurs in two phases [62]. to activate caspase-9. The activated caspase-9 then acti- Cell death, however, does not depend on the initial Ca2? vates caspase-3 which leads to (assisted by the release of rise, but rather invariably follows the delayed massive apoptosis-inducing factor triggered by cytochrome c) accumulation of Ca2?, called delayed Ca2? deregulation apoptotic cell death [78] (see Fig. 2). Stabilization of (DCD), occurring a few hours after the toxic challenge, mitochondrial function is associated with causing a no-return transition into the death process [63]. [79]. Activation of transient receptor potential channels of the melastatin family (TRPM), especially TRPM2 and TRPM7, is implicated in the DCD. TRPM2 can be acti- The role of caspases and calpains vated by H2O2 [64], reactive oxygen species (ROS) [65] and intracellular Ca2? [66]. Blockade of TRPM7 channels The role of caspase-3 has been greatly supported in animal in vitro can prevent Ca2? influx and decrease cell death studies. Caspase-3 knockout mice had smaller infarct following oxygen glucose deprivation. TRPM7 can be volumes than wild-type mice [80], and administration activated by ROS as well as phosphatidylinositol of caspase enzyme inhibitors would decrease infarction 123 228 Neurol Sci (2012) 33:223–237

size [81], while mice overexpressing human caspase-3 Rise in intracellular Ca2+ exhibited increased apoptosis and larger lesion volumes following cerebral ischemia [82]. In addition, caspase-3 activation, especially if combined with mitochondrial Calpain activation dysfunction, will lead to DNA fragmentation and neuronal apoptosis [83] (see Fig. 2). Pre-incubation with caspase-3- Upregulation of Bax Degrading Calpastatin (calpain specific inhibitor z-DEVD or pan-caspase inhibitor z-VAD activity endogenous inhibitor) prior to glutamate exposure resulted in a 50% reduction in cell death [84]. Caspase-3 activation In addition to mitochondrial dysfunction the elevated 2? Ca can activate degradative proteolytic enzymes, Fig. 4 Synergistic activity of calpain and caspase including the family of cytosolic proteases, calpains [85]. Calpain I (l-calpain) and II (m-calpain) are found while caspase-3 specific cleavage generates a 120 kDa throughout the brain and are activated by Ca2? at neutral SBDP [101]. pH. Inhibitors of NMDA-mediated Ca2? influx was asso- ciated with decreased calpains activity [86]. While calpains do play a role in normal cell function [87], over activation Production of ROS of these proteases has been implicated in both necrosis [88] and apoptosis [89]. They participate in excitotoxic cell Glutamate overactivity is associated with production of death by cleaving the NCX [90] and mGluRI [91]. The various ROS. ROS cause damage and oxidization of lipids, calpain-induced degradation of NCX, in addition to inter- DNA, and proteins. Accumulated Ca2? induces the acti- nalization of plasma membrane Ca2? ATPase is suggested vation of the neutral protease, calpain, which results in the to participate in DCD by causing critical rise in Ca2? [92] conversion of xanthine dehydrogenase into xanthine oxi- (see Fig. 3), although recent studies raised some doubt dase. Xanthine oxidase catalyzes the oxidation of xanthine about such assumption [93]. and hypoxanthine into uric acid, producing superoxide as a Calpains also truncate a number of anti-apoptotic pro- by-product [102]. Excess Ca2? in the mitochondria inter- teins, such as Bcl-2, Bcl-XL and Bid, which could account rupts the electron transport chain, therefore free electrons for the role of calpains in apoptosis. Activated calpains are accumulated in the mitochondria, which react with hydrolyze peptide bonds in cytoskeletal and structural oxygen that is supplied after reperfusion, and causes the proteins, membrane proteins and other regulatory and production of superoxide. The superoxide is further pro- signaling proteins [94]. Calpains have been observed to cessed to produce the hydroxyl radical by a Fenton reaction activate calcineurin through direct cleavage [95] and by or peroxynitrite by reacting with nitric oxide. The ROS degrading its endogenous inhibitor Cain/Cabin1 [96]. They also inhibit the electron transport chain in the mitochon- also upregulate proapoptotic Bax activity [97], which dria, and amplify a generation of mitochondrial free radi- eventually leads to downstream activation of caspase-3 cals [103]. [98]. While caspase-3 may cleave calpastatin, the endog- Zn2? is stored in the presynaptic vesicles of glutama- enous inhibitor of calpains [99] and so the two proteolytic tergic neurons, released with glutamate in an activity- enzymes may act synergistically [100] (see Fig. 4). dependent manner, and translocated into adjacent neurons Calpains and caspase-3 cleave the cytoskeletal protein [104]. The Zn2? translocation was observed in degenerat- a-spectrin at specific sites, and calpain-specific cleavage ing neurons after transient forebrain ischemia [105]. Zn2? generates a 145 kDa spectrin breakdown product (SBDP) ions enter into target cells through VGCC, NMDARs that are permeable to Ca2?,Na?/Ca2? exchanger, or Zn2? 2? Rise in intracellular Ca2+ transporter [106]. The entry and accumulation of Zn into neurons result in the transient generation of ROS, by activation of cyclooxygenases (COX) and PKC that Internalization of plasma Degradation of mediate latent neuronal death [107]. membrane Ca2+ ATPase NCX

Activation of arachidonic acid pathway Activation of Calpains

2? 2? Accumulated Ca in neurons results in the translocation Fig. 3 Relationship between Ca and calpains. Being activated by 2? high intracellular Ca2? level, calpains, in turn, will contribute to the of cPLA2 into the plasma membrane. And the Ca catastrophic rise in Ca2? overload through glutamate receptors inducing the 123 Neurol Sci (2012) 33:223–237 229

activation of cPLA2 will lead to production of neurotoxic NMDAR activation metabolites, such as prostaglandins, leukotrienes, reactive oxygen species, and platelet activating factors through the metabolism of arachidonic acid and lysophospholipids. The Rise in intracellular 2+ pharmacological inhibitors of cPLA2 reduce excitotoxic Ca and hypoxic-ischemic neuronal death [108]. Furthermore, the activation of NMDAR will cause an increased Activation of NOS expression and activation of COX-2 [109]. Increased COX-2 expression in ischemic brain was found to be due to excess glutamate [110]. Absence of COX-2 or its inhibition Increase in NO attenuates NMDA-mediated excitotoxicity and hypoxic- ischemic injury [111]. Increase in Neuronal peroxynitrite Death The role of NO Fig. 5 Closed circuit is present between peroxynitrite and intracel- lular Ca2? leading finally to cell death The role of NO, generated by NMDARs activation, in neuronal damage produced by glutamate has been estab- lished in the early 1990s. PSD-95 binds to the amino ter- minal of nNOS, a Ca2?-activated form of NOS, through its Inhibition of soluble guanylate cyclase and cGMP produc- PDZ domain. Therefore, PSD-95 may concentrate nNOS tion prevents glutamate and NO neurotoxicity [122]. near the NMDARs at postsynaptic sites in neurons [112]. Primary brain cultures treated with NOS inhibitors or cultures from mice with targeted disruption of nNOS are The role of AMPAR and KAR resistant to NMDA neurotoxicity [113]. NOS activity is enhanced by the elevation of intracel- Although most of the excitotoxic glutamate-induced dam- lular Ca2? [114], by a Ca2?/calmodulin-dependent mech- age is due to NMDAR activation, the AMPA/kainate anism [115]. Mitochondrial NOS is also activated by rise in pathway is not innocent. Activation of this pathway will intramitochondrial Ca2? [116]. Activation of mGluR1 was lead to the rise in the intracellular Na content, with sec- 2? found to be associated with increased NO level [117]. ondary rise in Ca concentration due to reverse operation ? 2? NO exacerbates brain damage by reducing neuronal of the Na /Ca exchanger [123]. These events will join energy production by inhibiting glycolytic and mitochon- the downstream cascade as evoked by NMDA receptor drial enzymes, by increasing DNA damage, and through activation. Glutamate acts on AMPARs to produce mem- the conversion of superoxide to peroxynitrite, being a brane depolarization and sufficient membrane depolariza- 2? highly reactive molecule, increasing the levels of toxic free tion removes the Mg block of NMDA channels, allowing 2? radicals [94]. Increased peroxynitrite production has been the influx of extracellular Ca into the cell [124]. shown to increase cytosolic Ca2?, contributing to cell death There are evidences suggesting the formation of ROS [118] (see Fig. 5). NO together with the generation of ROS after AMPAR activation [125]. The sole rise in the intra- ? (secondary to increased Ca2? and mitochondrial dysfunc- cellular Na after AMPAR stimulation may cause neuronal tion) such as hydroxyl radicals, superoxide anions can damage due to cellular swelling [126]. cause lipid peroxidation and membrane damage [119]. Stimulation of KRs by (KA), a cyclic analog Despite the mentioned harmful effects of NO, low NO of , caused excitotoxic neurodegeneration was found neuroprotective. Low concentration of NO accompanied by the expression of various genes, including played a protective role in glutamate neurotoxicity by immediate-early genes (IEG) and stress protein genes closing the NMDA receptor-gated ion channels, suggesting [127]. the dual effect of NO [120]. KA-induced neurodegeneration was not prevented either Another mediator in NO and glutamate neurotoxicity is by MK-801 or CNQX, blockers of NMDAR and AMPA/ cGMP. The rise in intracellular (not extracellular) cGMP, KA receptors, respectively. It indicated that CNQX-resis- being stimulated by NO, plays a role in the mechanisms of tant KARs might be involved in the observed neurode- glutamate, NMDA and kainate neurotoxicity in cultured generation. Accordingly, more specific inhibitors of KARs cerebral cortical neurons [121]. There is a good correlation might be effective in preventing KA-induced neuronal between inhibition of cGMP formation and neuroprotection. [128].

123 230 Neurol Sci (2012) 33:223–237

Glutamate and preconditioning glutamate is induced by NMDAR-gated Ca2? influx [124]. Ischemia induces rapid upregulation of CREB-targeted It is known that transient, sub-lethal or non-catastrophic genes [135]. CREB phosphorylation and subsequent gene brain ischemia can induce a protected state against sub- expression may play an important role in the acquisition of sequent episodes of ischemia (ischemic tolerance); this is ischemic tolerance [136]. Cpg15 is an immediate-early called ischemic preconditioning [129]. Preconditioning is gene induced by Ca2? influx through NMDAR and L-type associated with changes in the glutamatergic system, VDCC and is regulated by multiple transcription factors mainly increase in glutamate release through the non-Ca2?- including CREB [137]. CPG15 expression is upregulated dependent mechanisms [130] associated with increased after transient global ischemia protects cortical neurons expression and activity of glutamate transporters [131] (see from apoptosis by preventing activation of caspase path- Fig. 6). Although the malfunction of glutamate transporters ways [138]. appears to be important in glutamate accumulation during ischemic injury, the increased expression of glutamate expression has been shown to be important in acquisition Drugs inhibiting glutamate-induced cell injury of ischemic tolerance by removing glutamate from the extracellular space and maintaining the glutamate below Despite the extensive and continued research the mecha- neurotoxic level in the brain. The EAAT2 inhibitor, nisms of excitotoxicity, there are currently no pharmaco- dihydrokainate, prevents preconditioning in a dose-depen- logical interventions capable of providing significant dent manner [132]. The role of ciliary neurotrophic factor neuroprotection in the clinical setting of brain ischemia or in neuronal protection may be due to enhancement of injury. As shown in this review, glutamate induces neu- glutamate transporters’ activity [133]. ronal cell damage through multiple and complex pathways, The involvement of NMDAR in ischemic tolerance has which makes the interruption of these pathways rather been suggested, but not fully explained. The different roles difficult. Therapeutic targeting of a single pathway may not of NMDAR subtypes might be largely dependent on the provide satisfactory neuroprotection, due to the presence of activation of their downstream signaling pathways. NR2B alternative neurotoxic pathways by which glutamate can subunit of NMDAR may be particularly important in bypass the inhibited one. This suggests that combination neuronal ischemic preconditioning [134]. Phosphorylation therapies may have better outcomes than monotherapy of CREB in neurons after ischemia or exposure to [139]. Due to the unsatisfactory results reported by gluta- mate receptor blockers as neuroprotectants against ische- mia, efforts have been made to attach glutamate during Transient periods of earlier stage (prereceptor level) by inhibition of synthesis ischemia or release. Although less specific, interruption of signaling pathways following interaction (post- receptor level) were also suggested, e.g., by antioxidants Release of Glutamate (see Fig. 7). Neuroprotective agents against glutamate- induced excitotoxicity are summarized in Table 2. As early as 1980s, glutamate receptor antagonists had been found to attenuate the ischemic damage observed NMDAR activation Upregulation of in animal models. Many problems still face its clinical glutamate transporters Inhibitors of Glutamate synthesis Pre receptor binding Enhancement of Glutamate clearance Phosphorylation of level CBEB Inhibitors of Glutamate release Enhancement reuptake of glutamate during further periods of ischemia Receptor binding Glutamate receptor blockers level Cpg15

Inhibition of signaling pathways Post receptor binding Protection against level apoptosis Antioxidants

Fig. 7 Mechanism and stages of action of neuroprotectants against Fig. 6 Possible role of glutamate in preconditioning glutamate-induced neurotoxicity 123 Neurol Sci (2012) 33:223–237 231

Table 2 Examples of neuroprotective agents against glutamate-induced neurotoxicity Neuroprotective agents Reference

Inhibitors of glutamate formation 6-Diazo-5-oxo-L-norleucine (DON) Decreased glutamate release from activated microglia [140] Methionine sulfoximine Inhibited the synthesis of glutamate in vivo in mice [141] Phenylsuccinate Inhibited cytosolic glutamate formation from glutamine [142] Inhibitors of glutamate release Carbenoxolone Gap junction blocker which decrease glutamate microglial release [140] Dextromethorphan Inhibited glutamate release by suppression of presynaptic VDCC [143] Enhancement of glutamate clearance Ceftriaxone Increased the glial GLT expression enhancing glutamate uptake [144] Maslinic acid Promoted glutamate clearance by NF-jB-mediated EAAT 2 upregulation [145] Rosiglitazone Increased the GLT-1 expression by acting as PPAR agonist [146] NMDAR blockers ACEA 1021 Reduced the infarction size [147] Bis(7)tacrine Showed neuroprotective effect against retinal ischemic damage [148] Cerestat Had a good preclinical profile, but low clinical efficacy [149] Dextromethorphan It reduced infarction size but with a variety of dose-related reversible adverse events [150] Dizocilpine Reduced the infarction volume, but most effective given early after ischemia with [151] neuropsychiatric problems Gavestinel Generally better tolerated without neuropsychiatric adverse effects but it had little effects [152] of on infarct volume in patients with ischemic stroke Ketamine and thiopental Showed neuroprotective effects against NMDA-induced neurotoxicity [153] Beside NMDAR blocking, it inhibited excitatory neurotransmitter release, blocked Ca2? [154] channels, dilated the cerebral vasculature Memantine Improved neurochemical and neurobehavioral functions after ischemia [155] Dizocilpine Reduced the infarction size [156] Remacemide Reduced the infarction size [157] Traxoprodil Modest neuroprotective activity in humans with stroke [158] AMPAR blockers NBQX Had neuroprotective capacities but was associated with nephrotoxicity [159] PNQX Showed neuroprotective effects against oxygen–glucose deprivation [160] KAR blockers LY37770 Selective GluK1 antagonist which is neuroprotective under various states [161] Modulation of mGluRs ACPD Activated the mGluR protecting against NMDA receptor activation [162] CHPG Acted as mGluR5 agonist reducing the infarction volume [163] LY379268 Group II mGluR agonist, stimulating astrocytes to produce neuroprotective factors and [164] reducing glutamate release PPG A novel group III mGluR agonist which is neuroprotective against hypoxic injury [165] YM-202074 mGluR1 antagonist which decreased infarction size [166] Inhibitors of Na?/H? exchanger KR-33028 The novel Na?/H? exchanger inhibitor inhibit intracellular Ca2? overload [60] Inhibition of postreceptor signaling mechanisms It inhibited glutamate-induced activation of NOS reducing infarction volume in [167] experimental animals PSD-95 inhibitors, e.g., N-alkylated Reduced infarction size even after ischemic insult and improve neurobehavioral changes [168] tetrapeptides Inhibitors of Ca2? influx 2-Cyclopropylimino-3-methyl-1,3- Decreased Ca2? influx, mitochondrial dysfunction, overproduction of NO and oxidative [169] thiazoline hydrochloride stress in cultured glial cells Lacidipine Reduced infarction size before or after ischemic injury [170]

123 232 Neurol Sci (2012) 33:223–237

Table 2 continued Neuroprotective agents Reference

Nimodipine Reduced Ca2? influx, neuroprotective effect in kainate, glutamate, and NMDA-induced [171] neurotoxicity NGP1-01 Inhibited major neuronal calcium channels; L- and N-types. It also inhibited NMDA- [172] induced Ca2? influx by 52% ACPD (1-aminocyclopentyl-1,3-dicarboylic acid), CHPG 2-chloro-5-hydroxyphenylglycine, KR-33028 4-cyano (benzobthiophene-2- carbonyl)guanidine, NGP1-01 8-benzylamino-8,11-oxapentacycloundecane, LY379268 2-oxa-4-aminobicyclohexane-4,6-dicarboxylate, PPG 4-phosphonophenylglycine, YM-202074 N-cyclohexyl-6-{[(2-methoxyethyl)(methyl)amino]methyl}-N-methylthiazolo[3,2-a]benzimidazole-2- carboxamide

usefulness. The need for very early administration and the Conclusion dose-dependent neurobehavioral toxicity in humans limit their usefulness [173]. The possibility exists that NMDAR Glutamate is not just a neurotransmitter released during antagonism, while potentially protective in focal ischemia, periods of cerebral ischemia, but it plays a keystone role in is also deleterious in terms of adversely affecting endoge- neural death and its induced excitotoxicity is considered nous NMDAR-mediated neuronal survival mechanisms one of the most important causes of nerve cell loss during [174]. AMPAR and KAR blockers are also, although less cerebrovascular stroke. Various therapeutic options are extensively, investigated with limited success due to neu- present interrupting the glutamatergic role in cerebral ronal and systemic toxicity [175, 176]. ischemic injury. But it is still early to consider many of Modulation of mGluRs represents an attractive multi- them as main line therapy. potential therapeutic strategy. The activation of mGluR5 and group II and III mGluRs has been met with improved neurosurvival after injury, and antagonism of mGluR1 has References similar effects, reflecting the multiple actions of these receptors [177]. Inhibition of downstream glutamate- 1. Wang Y, Qin ZH (2010) Molecular and cellular mechanisms of induced activation of NO synthase and Na?/H? exchanger excitotoxic neuronal death. Apoptosis 15:1382–1402 are also explored with some success [167, 178]. 2. Derouiche A (2004) The perisynaptic astrocyte process as a glial Ca2? channel blockers seem to have a direct protective compartment immunolabeling for glutamine synthetase and other glial markers. In: Hertz L (ed) Non-neuronal cells of the role against neuronal ischemic damage away from their nervous system. Function and dysfunction. Elsevier, Amster- vascular effects [179]. Inhibition of Ca2? influx provided dam, pp 147–163 neuroprotection by decreasing glutamate release [180], 3. Hertz L, Peng L, Dienel GA (2007) Energy metabolism in decreasing Ca2? overload in susceptible neurons and astrocytes, high rate of oxidative metabolism and spatiotemporal dependence on glycolysis/glycogenolysis. J Cereb Blood Flow reduction of NO production [181]. Metab 27:219–249 4. Takeuchi H, Jin S, Wang J et al (2006) Tumor necrosis factor- alpha induces neurotoxicity via glutamate release from hemi- Current status and future strategies channels of activated microglia in an autocrine manner. J Biol Chem 281:21362–21368 5. Reid CA, Bekkers JM, Clements JD (2003) Presynaptic Ca2? It is quite surprising that despite of the growing evidences channels: a functional patchwork. Trends Neurosci 26:683–687 about the negative role of glutamate in neuronal ischemic 6. Nicholls DG, Sihra TS, Sanchez-Prieto J (1987) Calcium- damage, the utilization of the previously mentioned strat- dependent and -independent release of glutamate from synap- tosomes monitored by continuous fluorometry. J Neurochem egies is still beyond clinical application. The medical 49:50–57 management of ischemic stroke is still dependent on 7. Shigeri Y, Seal RP, Shimamoto K (2004) Molecular pharma- thrombolytics, anticoagulants and nonspecific neurotonics cology of glutamate transporters, EAATs and VGLUTs. Brain [182]. Introduction of new drugs preventing the effect of Res Rev 45:250–265 8. Danbolt NC (2001) Glutamate uptake. Prog Neurobiol 65:1–105 glutamate during ischemia opens a new chapter in man- 9. Ko¨hr G (2006) NMDA receptor function: subunit composition agement of ischemic stroke which might bring substantial versus spatial distribution. Cell Tissue Res 326:439–446 10. Sobczyk A, Scheuss V, Svoboda K (2005) NMDA receptor benefits for millions of patients. The development of novel 2? glutamate receptor blockers characterized by lower toxicity subunit dependent [Ca ] signaling in individual hippocampal dendritic spines. J Neurosci 25:6037–6046 and better binding properties is a milestone for researchers 11. Kim MJ, Dunah AW, Wang YT, Sheng M (2005) Differential aiming for satisfactory neuronal protection. roles of NR2A- and NR2B-containing NMDA receptors in

123 Neurol Sci (2012) 33:223–237 233

Ras-ERK signaling and AMPA receptor trafficking. Neuron 33. Huang R, Hertz L (1994) Effect on anoxia on glutamate for- 46:745–760 mation from glutamine in cultured neurons, dependence on 12. Nakanishi N, Tu S, Shin Y et al (2009) Neuroprotection by the neuronal subtype. Brain Res 660:129–137 NR3A subunit of the NMDA receptor. J Neurosci 29:5260–5265 34. Hansen AJ (1985) Effect of anoxia on ion distribution in the 13. Papadia S, Stevenson P, Hardingham NR, Bading H, brain. Physiol Rev 65:101–148 Hardingham GE (2005) Nuclear Ca2? and the cAMP response 35. Kimelberg HK, Mongin AA (1998) Swelling-activated release element-binding protein family mediate a late phase of activity- of excitatory amino acids in the brain, relevance for patho- dependent neuroprotection. J Neurosci 25:4279–4287 physiology. Contrib Nephrol 123:240–257 14. Pokorska A, Vanhoutte P, Arnold FJ, Silvagno F, Hardingham 36. Coffey ET, Sihra TS, Nicholls DG, Pocock JM (1994) Phos- GE, Bading H (2003) Synaptic activity induces signaling to phorylation of synapsin I and MARCKS in nerve terminals is CREB without increasing global levels of cAMP in hippocampal mediated by Ca2? entry via an Aga-GI sensitive Ca2? channel neurons. J Neurochem 84:447–452 which is coupled to glutamate exocytosis. FEBS Lett 353:264– 15. Sheng M, Hoogenraad CC (2007) The postsynaptic architecture 268 of excitatory synapses. Annu Rev Biochem 76:823–847 37. Mena FV, Baab PJ, Zielke CL, Zielke HR (2000) In vivo glu- 16. Forder JP, Tymianski M (2009) Postsynaptic mechanisms of tamine hydrolysis in the formation of extracellular glutamate in excitotoxicity: involvement of postsynaptic density proteins, the injured rat brain. J Neurosci Res 60:632–641 radicals, and oxidant molecules. Neuroscience 158:293–300 38. Rossi DJ, Oshima T, Attwell D (2000) Glutamate release in 17. Cui H, Hayashi A, Sun HS et al (2007) PDZ protein interactions severe brain ischaemia is mainly by reversed uptake. Nature underlying NMDA receptor-mediated excitotoxicity and neu- 403:316–321 roprotection by PSD-95 inhibitors. J Neurosci 27:9901–9915 39. Sheldon AL, Robinson MB (2007) The role of glutamate 18. Santos SD, Carvalho AL, Caildeira MV, Duarte CB (2009) transporters in neurodegenerative diseases and potential oppor- Regulation of AMPA receptors and synaptic plasticity. Neuro- tunities for intervention. Neurochem Int 51:333–355 science 158:105–125 40. Ketheeswaranathan P, Turner NA, Spary EJ, Batten TF, McColl 19. Jane DE, Lodge D, Collingridge GL (2009) Kainate receptors: BW, Saha S (2011) Changes in glutamate transporter expression pharmacology, function and therapeutic potential. Neurophar- in mouse forebrain areas following focal ischemia. Brain Res macology 56:90–113 1418C:93–103 20. Lerma J (2003) Roles and rules of kainate receptors in synaptic 41. Globus MY, Busto R, Dietrich WD, Martinez E, Valdes I, transmission. Nat Rev Neurosci 4:481–495 Ginsberg MD (1988) Effect of ischemia on the in vivo release of 21. Benarroch EE (2008) Metabotropic glutamate receptors: syn- striatal dopamine, glutamate, and g-aminobutyric acid studied aptic modulators and therapeutic targets for neurologic disease. by intracerebral microdialysis. J Neurochem 51:1455–1464 Neurology 70:964–968 42. Lucas DR, Newhouse JP (1957) The toxic effect of sodium 22. Pacheco R, Ciruela F, Casado´ V et al (2004) Group I metabo- L-glutamate on the inner layers of the retina. AMA Arch Oph- tropic glutamate receptors mediate a dual role of glutamate in T thalmol 58:193–201 cell activation. J Biol Chem 279:33352–33358 43. Choi DW, Maulucci-Gedde M, Kriegstein AR (1987) Glutamate 23. Conn PJ, Pin JP (1997) Pharmacology and functions of meta- neurotoxicity in cortical cell culture. J Neurosci 7:357–368 botropic glutamate receptors. Annu Rev Pharmacol Toxicol 44. Benveniste H, Drejer J, Schousboe A, Diemer NH (1984) 37:205–237 Elevation of the extracellular concentrations of glutamate 24. Kno¨pfel T, Lukic S, Leonard T, Flor PJ, Kuhn R, Gasparini F and aspartate in rat hippocampus during transient cerebral (1995) Pharmacological characterization of MCCG and MAP4 ischemia monitored by intracerebral microdialysis. J Neurochem at the mGluR1b, mGluR2 and mGluR4a human metabotropic 43:1369–1374 glutamate receptor subtypes. Neuropharmacology 34:1099– 45. Novelli A, Reilly JA, Lysko PG, Henneberry RC (1988) Glu- 1102 tamate becomes neurotoxic via the N-methyl-D-aspartate 25. Toms NJ, Jane DE, Kemp MC, Bedingfield JS, Roberts PJ receptor when intracellular energy levels are reduced. Brain Res (1996) The effects of (RS)-alpha-cyclopropyl-4-phosphonophe- 451:205–212 nylglycine ((RS)-CPPG), a potent and selective metabotropic 46. Meldrum B, Evans M, Griffiths T, Simon R (1985) Ischaemic glutamate receptor antagonist. Br J Pharmacol 119:851–854 brain damage: the role of excitatory activity and of calcium 26. Siesjo¨ BK, Bengtsson F, Grampp W, Theander S (1989) Cal- entry. Br J Anaesth 57:44–46 cium, excitotoxins, and neuronal death in brain. Ann NY Acad 47. Hugon J, Vallat JM, Dumas M (1996) Role of glutamate and Sci 568:234–251 excitotoxicity in neurologic diseases. Rev Neurol 152:239–248 27. Nishizawa Y (2001) Glutamate release and neuronal damage in 48. Furukawa K, Fu W, Li Y, Witke W, Kwiatkowski DJ, Mattson MP ischemia. Life Sci 69:369–381 (1997) The actin-severing protein gelsolin modulates cal- 28. McCulloch J (1994) Glutamate receptor antagonists in cerebral cium channel and NMDA receptor activities and vulnerability ischaemia. J Neural Transm Suppl 43:71–79 to excitotoxicity in hippocampal neurons. J Neurosci 17:8178– 29. Brouns R, De Deyn PP (2009) The complexity of neurobio- 8186 logical processes in acute ischemic stroke. Clin Neurol Neuro- 49. Dobrek L, Thor P (2011) Glutamate NMDA receptors in path- surg 111:483–495 ophysiology and pharmacotherapy of selected nervous system 30. Obrenovitch TP, Richards DA (1995) Extracellular neurotrans- diseases. Postepy Hig Med Dosw 65:338–346 mitter changes in cerebral ischaemia. Cerebvasc Brain Metab 50. Simeone TA, Sanchez RM, Rho JM (2004) Molecular biology Rev 7:1–54 and ontogeny of glutamate receptors in the mammalian central 31. Shimada N, Graf R, Rosner G, Wakayama A, George CP, Heiss nervous system. J Child Neurol 19:343–360 WD (1989) Ischemic flow threshold for extracellular glutamate 51. Szydlowska K, Tymianski M (2010) Calcium, ischemia and increase in cat cortex. J Cereb Blood Flow Metab 9:603–606 excitotoxicity. Cell Calcium 47:122–129 32. Jones TH, Morawetz RB, Crowell RM, Marcoux FW, 52. Lazarawicz JW (1996) Calcium transients in brain ischemia: FitzGibbon SJ, DeGirolami U, Ojemann RG (1981) Thresholds role in neuronal injury. Acta Neurobiol Exp 56:299–311 of focal cerebral ischemia in awake monkeys. J Neurosurg 53. Cano-Abad MF, Villarroya M, Garcı´a AG, Gabilan NH, Lo´pez 54:773–782 MG (2001) Calcium entry through L-type calcium channels

123 234 Neurol Sci (2012) 33:223–237

causes mitochondrial disruption and chromaffin cell death. 73. Bernardi P, Scorrano L, Colonna R, Petronilli V, Di Lisa F J Biol Chem 276:39695–39704 (1999) Mitochondria and cell death. Mechanistic aspects and 54. Yagami T, Ueda K, Sakaeda T et al (2004) Protective effects of methodological issues. Eur J Biochem 264:687–701 a selective L-type voltage -sensitive calcium channel blocker, 74. Robertson CL, Bucci CJ, Fiskum G (2004) Mitochondrial S-312-d, on neuronal cell death. Biochem Pharmacol 67:1153– response to calcium in the developing brain. Brain Res Dev 1165 Brain Res 151:141–148 55. Rothman SM, Olney JW (1995) Excitotoxicity and the NMDA 75. Hansson MJ, Mattiasson G, Ma˚nsson R et al (2004) The receptor—still lethal after eight years. Trends Neurosci nonimmunosuppressive cyclosporin analogs NIM811 and 18:57–58 UNIL025 display nanomolar potencies on permeability transi- 56. Mizuno F, Barabas P, Krizaj D, Akopian A (2010) Glutamate- tion in brain-derived mitochondria. J Bioenerg Biomembr 36: induced internalization of Ca(v)13 L-type Ca(2?) channels 407–413 protects retinal neurons against excitotoxicity. J Physiol 588: 76. Schinzel AC, Takeuchi O, Huang Z et al (2005) Cyclophilin D is 953–966 a component of mitochondrial permeability transition and 57. Nash MS, Saunders R, Young KW, Challiss RA, Nahorski SR mediates neuronal cell death after focal cerebral ischemia. Proc (2001) Reassessment of the Ca2? sensing property of a type I Natl Acad Sci USA 102:12005–12010 metabotropic glutamate receptor by simultaneous measurement 77. Robertson CL, Scafidi S, McKenna MC, Fiskum G (2009) of inositol 1,4,5-trisphosphate and Ca2? in single cells. J Biol Mitochondrial mechanisms of cell death and neuroprotection in Chem 276:19286–19293 pediatric ischemic and traumatic brain injury. Exp Neurol 58. Trudeau LE, Parpura V, Haydon PG (1999) Activation of neu- 218:371–380 rotransmitter release in hippocampal nerve terminals during 78. Love S (2003) Apoptosis and brain ischaemia. Prog Neuro- recovery from intracellular acidification. J Neurophysiol psychopharmacol Biol Psychiatry 27:267–282 81:2627–2635 79. Li SY, Jia YH, Sun WG et al (2010) Stabilization of mito- 59. Grinstein S, Woodside M, Sardet C, Pouyssegur J, Rotin D chondrial function by tetramethylpyrazine protects against kai- (1992) Activation of the Na?/H? antiporter during cell volume nate-induced oxidative lesions in the rat hippocampus. Free regulation. J Biol Chem 267:23823–23828 Radic Biol Med 48:597–608 60. Lee BK, Lee DH, Parka S et al (2009) Effects of KR 33028, a 80. Le DA, Wu Y, Huang Z et al (2002) Caspase activation and novel Na?/H? exchanger-1 inhibitor, on glutamate-induced neuroprotection in caspase-3-deficient mice after in vivo cere- neuronal cell death and ischemia induced cerebral infarct. Brain bral ischemia and in vitro oxygen glucose deprivation. Proc Natl Res 1248:22–30 Acad Sci USA 99:15188–15193 61. Favero TG, Zable AC, Abramson JJ (1995) Hydrogen peroxide 81. Ma J, Endres M, Moskowitz MA (1998) Synergistic effects of stimulates the Ca2? release channel from skeletal muscle sar- caspase inhibitors and MK-801 in brain injury after transient coplasmic reticulum. J Biol Chem 270:25557–25563 focal cerebral ischaemia in mice. Br J Pharmacol 124:756–762 62. Celsi F, Pizzo P, Brini M et al (2009) Mitochondria, calcium and 82. Kerr LE, McGregor AL, Amet LE et al (2004) Mice over- cell death: a deadly triad in neurodegeneration. Biochim Bio- expressing human caspase 3 appear phenotypically normal but phys Acta 1787:335–344 exhibit increased apoptosis and larger lesion volumes in 63. Mattson MP (2007) Calcium and neurodegeneration. Aging Cell response to transient focal cerebral ischaemia. Cell Death Differ 6:337–350 11:1102–1111 64. Kaneko S, Kawakami S, Hara Y et al (2006) A critical role of 83. Rosell A, Cuadrado E, Alvarez-Sabin J et al (2008) Caspase-3 is TRPM2 in neuronal cell death by hydrogen peroxide. J Phar- related to infarct growth after human ischemic stroke. Neurosci macol Sci 101:66–76 Lett 430:1–6 65. Nazirog˘lu M (2007) New molecular mechanisms on the acti- 84. Zhang Y, Bhavnani BR (2005) Glutamate-induced apoptosis in vation of TRPM2 channels by oxidative stress and ADP-ribose. primary cortical neurons is inhibited by equine estrogens via Neurochem Res 32:1990–2001 down-regulation of caspase-3 and prevention of mitochondrial 66. Du J, Xie J, Yue L (2009) Intracellular calcium activates cytochrome c release. BMC Neurosci 24:6–13 TRPM2 and its alternative spliced isoforms. Proc Natl Acad Sci 85. Czogalla A, Sikorski AF (2005) Spectrin and calpain: a ‘target’ USA 106:7239–7244 and a ‘sniper’ in the pathology of neuronal cells. Cell Mol Life 67. Bae CY, Sun HS (2011) TRPM7 in cerebral ischemia and Sci 62:1913–1924 potential target for drug development in stroke. Acta Pharmacol 86. Jiang SX, Zheng RY, Zeng JQ, Li XL, Han Z, Hou ST (2010) Sin 32:725–733 Reversible inhibition of intracellular calcium influx through 68. Duchen MR (2000) Mitochondria and calcium: from cell sig- NMDA receptors by imidazoline I(2) receptor antagonists. Eur J naling to cell death. J Physiol 1:57–68 Pharmacol 629:12–19 69. Nicholls DG, Johnson-Cadwell L, Vesce S, Jekabsons M, 87. Bhatt A, Kaverina I, Otey C, Huttenlocher A (2002) Regulation Yadava N (2007) Bioenergetics of mitochondria in cultured of focal complex composition and disassembly by the calcium- neurons and their role in glutamate excitotoxicity. J Neurosci dependent protease calpain. J Cell Sci 115:3415–3425 Res 85:3206–3212 88. Gores GJ, Miyoshi H, Botla R, Aguilar HI, Bronk SF (1998) 70. Zaidan E, Sims NR (1994) The calcium content of mitochondria Induction of the mitochondrial permeability transition as a from brain subregions following short-term forebrain ischemia mechanism of liver injury during cholestasis: a potential role for and recirculation in the rat. J Neurochem 63:1812–1819 mitochondrial proteases. Biochim Biophys Acta 1366:167–175 71. Schild L, Huppelsberg J, Kahlert S, Keilhoff G, Reiser G (2003) 89. Ray SK, Wilford GG, Crosby CV, Hogan EL, Banik NL (1999) Brain mitochondria are primed by moderate Ca2? rise upon Diverse stimuli induce calpain overexpression and apoptosis in hypoxia/reoxygenation for functional breakdown and morpho- C6 glioma cells. Brain Res 829:18–27 logical disintegration. J Biol Chem 278:25454–25460 90. Hara MR, Snyder SH (2007) Cell signaling and neuronal death. 72. Li CY, Chin TY, Chueh SH (2004) Rat cerebellar granule Annu Rev Pharmacol Toxicol 47:117–141 cells are protected from glutamate-induced excitotoxicity by 91. Xu W, Wong TP, Chery N, Gaertner T, Wang YT, Baudry M S-nitrosoglutathione but not . Am J Physiol Cell (2007) Calpain-mediated mGluR1alpha truncation: a key step in Physiol 286:893–904 excitotoxicity. Neuron 53:399–412

123 Neurol Sci (2012) 33:223–237 235

92. Bano D, Munarriz E, Chen HL et al (2007) The plasma mem- acid receptor and a bivalent neuronal NO synthase PDZ domain. brane Na?/Ca2? exchanger is cleaved by distinct protease J Biol Chem 274:27467–27473 families in neuronal cell death. Ann NY Acad Sci 1099:451–455 113. Dawson VL, Dawson TM, London ED, Bredt DS, Snyder SH 93. Gerencser AA, Mark KA, Hubbard AE et al (2009) Real-time (1991) Nitric oxide mediates glutamate neurotoxicity in primary visualization of cytoplasmic calpain activation and calcium cortical cultures. Proc Natl Acad Sci USA 88:6368–6371 deregulation in acute glutamate excitotoxicity. J Neurochem 114. Bredt DS (1996) NO NMDA receptor activity. Nat Biotechnol 110:990–1004 14:944 94. Small DL, Morley P, Buchan MA (1999) Biology of ischemic 115. Guix FX, Uribesalgo I, Coma M, Munˇoz FJ (2005) The physi- cerebral cell death. Prog Cardiovasc Dis 42:185–207 ology and pathophysiology of nitric oxide in the brain. Prog 95. Wu HY, Tomizawa K, Oda Y et al (2004) Critical role of cal- Neurobiol 76:126–152 pain-mediated cleavage of calcineurin in excitotoxic neurode- 116. Haynes V, Elfering SL, Squires RJ et al (2003) Mitochondrial generation. J Biol Chem 279:4929–4940 nitric-oxide synthase: role in pathophysiology. IUBMB Life 96. Kim MJ, Jo DG, Hong GS et al (2002) Calpain-dependent 55:599–603 cleavage of cain/cabin1 activates calcineurin to mediate cal- 117. Llansola M, Felipo V (2010) Metabotropic glutamate receptor 5, cium-triggered cell death. Proc Natl Acad Sci USA 99:9870– but not 1, modulates NMDA receptor-mediated activation of 9875 neuronal nitric oxide synthase. Neurochem Int 56:535–545 97. Wood DE, Thomas A, Devi LA et al (1998) Bax cleavage is 118. Gutie´rrez-Martı´n Y, Martı´n-Romero FJ, Henao F, Gutie´rrez- mediated by calpain during drug-induced apoptosis. Oncogene Merino C (2005) Alteration of cytosolic free calcium homeo- 17:1069–1078 stasis by SIN-1: high sensitivity of L-type Ca2? channels to 98. Chan SL, Mattson MP (1999) Caspase and calpain substrates: extracellular oxidative/nitrosative stress in cerebellar granule roles in synaptic plasticity and cell death. J Neurosci Res cells. J Neurochem 92:973–989 58:167–190 119. Lovs S (1999) Oxidative stress in brain ischemia. Brain Pathol 99. Neumar RW, Xu YA, Gada H, Guttmann RP, Siman R (2003) 9:119–131 Cross-talk between calpain and caspase proteolytic systems 120. Gepdiremen A, Hacimuftuoglu A, Buyukokuroglu ME, during neuronal apoptosis. J Biol Chem 278:14162–14167 Suleyman H (2002) Nitric oxide donor sodium nitroprusside 100. Blomgren K, Zhu C, Wang X et al (2001) Synergistic activation induces neurotoxicity in cerebellar granular cell culture in rats of caspase-3 by m-calpain after neonatal hypoxia–ischemia: a by an independent mechanism from L-type or dantrolene-sen- mechanism of ‘‘pathological apoptosis’’? J Biol Chem 276: sitive calcium channels. Biol Pharm Bull 25:1295–1297 10191–10198 121. Frandsen A, Andersen CF, Schousboe A (1992) Possible role of 101. Wang KK, Posmantur R, Nath R et al (1998) Simultaneous cGMP in excitatory amino acid induced cytotoxicity in cultured degradation of aII- and bII-spectrin by caspase 3 (CPP32) in cerebral cortical neurons. Neurochem Res 17:35–43 apoptotic cells. J Biol Chem 273:22490–22497 122. Montoliu C, Llansola M, Kosenko E, Corbala´n R, Felipo V 102. McCord JM, Roy RS, Schaffer SW (1985) Free radicals and (1999) Role of cyclic GMP in glutamate neurotoxicity in pri- myocardial ischemia. The role of xanthine oxidase. Adv Myo- mary cultures of cerebellar neurons. Neuropharmacology 38: cardiol 5:183–189 1883–1891 103. Won SJ, Kim DY, Gwag BJ (2002) Cellular and molecular 123. Itoh T, Itoh A, Horiuchi K, Pleasure D (1998) AMPA receptor- pathways of ischemic neuronal death. J Biochem Mol Biol mediated excitotoxicity in human NT2-N neurons results from 35:67–86 loss of intracellular Ca2? homeostasis following marked eleva- 104. Frederickson CJ, Moncrieff DW (1994) -containing neu- tion of intracellular Na?. J Neurochem 71:112–124 rons. Biol Signals 3:127–139 124. Lin CH, Chen PS, Gean PW (2008) Glutamate preconditioning 105. Suh SW, Chen JW, Motamedi M et al (2000) Evidence that prevents neuronal death induced by combined oxygen–glucose synoptically-released zinc contributes to neuronal injury after deprivation in cultured cortical neurons. Eur J Pharmacol 589: traumatic brain injury. Brain Res 852:268–273 85–93 106. Colvin RA, Davis N, Nipper RW, Carter PA (2000) Zinc 125. Bae JH, Mun KC, Park WK et al (2002) EGCG attenuates transport in the brain: routes of zinc influx and efflux in neurons. AMPA-induced intracellular calcium increase in hippocampal J Nutr 130:1484–1487 neurons. Biochem Biophys Res Commun 290:1506–1512 107. Shuttleworth CW, Weiss JH (2011) Zinc: new clues to diverse 126. Endres M, Dirnagl U (2003) Ischemia and stroke. In: Alzheimer roles in brain ischemia. Trends Pharmacol Sci 32:480–486 C (ed) Molecular and cellular biology of neuroprotection in the 108. Rothman SM, Yamada KA, Lancaster N (1993) Nordi- CNS. Advances in experimental medicine and biology. Kluwer, hydroguaiaretic acid attenuates NMDA neurotoxicity—action New York, pp 455–473 beyond the receptor. Neuropharmacology 32:1279–1288 127. Tang Y, Lu A, Aronow BJ, Wagner KR, Sharp FR (2002) 109. Miettinen S, Fusco FR, Yrja¨nheikki J et al (1997) Spreading Genomic responses of the brain to ischemic stroke, intracerebral depression and focal brain ischemia induce cyclooxygenase-2 in hemorrhage, kainate seizures, hypoglycaemia, and hypoxia. Eur cortical neurons through N-methyl-D--receptors and J Neurosci 15:1937–1952 phospholipase A2. Proc Natl Acad Sci USA 94:6500–6505 128. Mohammadi S, Pavlik A, Krajci D, Al-Sarraf H (2009) NMDA 110. Ikeda-Matsuo Y, Hirayama Y, Ota A, Uematsu S, Akira S, preconditioning and neuroprotection in vivo: delayed onset of Sasaki Y (2010) Microsomal prostaglandin E synthase-1 and kainic acid-induced neurodegeneration and c-Fos attenuation in cyclooxygenase-2 are both required for ischaemic excitotoxic- CA3a neurons. Brain Res 1256:162–172 ity. Br J Pharmacol 159:1174–1186 129. Blanco M, Lizasoain I, Sobrino T, Vivancos J, Castillo J (2006) 111. Iadecola C, Niwa K, Nogawa S et al (2001) Reduced suscepti- Ischemic preconditioning: a novel target for neuroprotective bility to ischemic brain injury and N-methyl-D-aspartate-medi- therapy. Cerebrovasc Dis 21:38–47 ated neurotoxicity in cyclooxygenase-2-deficient mice. Proc 130. Andrade AL, Rossi DJ (2010) Simulated ischaemia induces Natl Acad Sci USA 98:1294–1299 Ca2?-independent glutamatergic vesicle release through actin 112. Christopherson KS, Hillier BJ, Lim WA, Bredt DS (1999) PSD- filament depolymerization in area CA1 of the hippocampus. 95 assembles a ternary complex with the N-methyl-D-aspartic J Physiol 588:1499–1514

123 236 Neurol Sci (2012) 33:223–237

131. Liu AJ, Hu YY, Li WB, Xu J, Zhang M (2011) Cerebral 149. Muir KW, Grosset DG, Gamzu E, Lees KR (1994) Pharmaco- ischemic pre-conditioning enhances the binding characteristics logical effects of the non-competitive NMDA antagonist CNS and glutamate uptake of glial glutamate transporter-1 in hippo- 1102 in normal volunteers. Br J Clin Pharmacol 38:33–38 campal CA1 subfield of rats. J Neurochem 119:202–209 150. Keller M, Griesmaier E, Auer M et al (2008) Dextromethorphan 132. Zhang M, Li WB, Geng JX et al (2007) The upregulation of glial is protective against sensitized N-methyl-D-aspartate receptor- glutamate transporter-1 participates in the induction of brain mediated excitotoxic brain damage in the developing mouse ischemic tolerance in rats. J Cereb Blood Flow Metab 27:1352– brain. Eur J Neurosci 27:874–883 1368 151. Olney JW (1994) Neurotoxicity of NMDA receptor antagonists: 133. Beurrier C, Faideau M, Bennouar KE et al (2010) Ciliary neu- an overview. Psychopharmacol Bull 30:533–540 rotrophic factor protects striatal neurons against excitotoxicity 152. Warach S, Kaufman D, Chiu D et al (2006) Effect of the glycine by enhancing glial glutamate uptake. PLoS One 5:8550 antagonist gavestinel on cerebral infarcts in acute stroke 134. Chen M, Lu T, Chen X et al (2008) Differential roles of NMDA patients, a randomized placebo-controlled trial: The GAIN MRI receptor subtypes in ischemic neuronal cell death and ischemic Substudy. Cerebrovasc Dis 21:106–111 tolerance. Stroke 39:3042–3048 153. Shibuta S, Varathan S, Mashimo T (2006) Ketamine and thio- 135. Barone FC, White RF, Spera PA et al (1998) Ischemic pre- pental sodium: individual and combined neuroprotective effects conditioning and brain tolerance: temporal histological and on cortical cultures exposed to NMDA or nitric oxide. Br J functional outcomes, protein synthesis requirement, and inter- Anaesth 97:517–524 leukin-1 receptor antagonist and early gene expression. Stroke 154. Ovbiagele B, Kidwell CS, Starkman S, Saver JL (2003) Neu- 29:1937–1950 roprotective agents for the treatment of acute ischemic stroke. 136. Mabuchi T, Kitagawa K, Kuwabara K (2001) Phosphorylation Curr Neurol Neurosci Rep 3:9–20 of cAMP response element-binding protein in hippocampal 155. Babu CS, Ramanathan M (2009) Pre-ischemic treatment with neurons as a protective response after exposure to glutamate in memantine reversed the neurochemical and behavioural vitro and ischemia in vivo. J Neurosci 21:9204–9213 parameters but not energy metabolites in middle cerebral artery 137. Fujino T, Lee WC, Nedivi E (2003) Regulation of CPG15 by occluded rats. Pharmacol Biochem Behav 92:424–432 signaling pathways that mediate synaptic plasticity. Mol Cell 156. Park CK, Nehls DG, Graham DI, Teasdale GM, McCulloch J Neurosci 24:538–554 (1988) Focal cerebral ischaemia in the cat: treatment with the 138. Putz U, Harwell C, Nedivi E (2005) Soluble CPG15 expressed glutamate antagonist MK-801 after induction of ischaemia. during early development rescues cortical progenitors from J Cereb Blood Flow Metab 8:757–762 apoptosis. Nat Neurosci 8:322–331 157. Bannan PE, Graham DI, Lees KR, McCulloch J (1994) Neuro- 139. Zausinger S, Scho¨ller K, Plesnila N, Schmid-Elsaesser R (2003) protective effect of remacemide hydrochloride in focal cerebral Combination drug therapy and mild hypothermia after transient ischemia in the cat. Brain Res 664:271–275 focal cerebral ischemia in rats. Stroke 34:2246–2251 158. Yurkewicz L, Weaver J, Bullock MR, Marshall LF (2005) The 140. Takeuchi H, Jin S, Suzuki H et al (2008) Blockade of microglial effect of the selective NMDA receptor antagonist traxoprodil in glutamate release protects against ischemic brain injury. Exp the treatment of traumatic brain injury. J Neurotrauma 22:1428– Neurol 214:144–146 1443 141. Ghoddoussi F, Galloway MP, Jambekar A, Bame M, Needleman 159. Gill R, Nordholm L, Lodge D (1992) The neuroprotective R, Brusilow WS (2010) Methionine sulfoximine, an inhibitor of actions of 2,3-dihydroxy-6-nitro-7-sulfamoyl-benzo(F)quinoxa- glutamine synthetase, lowers brain glutamine and glutamate in a line (NBQX) in a rat focal ischaemia model. Brain Res 580: mouse model of ALS. J Neurol Sci 290:41–47 35–43 142. Huang R, Hertz L (1995) Neuroprotective effect of phenylsuc- 160. Montero M, Nielsen M, Rønn LC, Møller A, Noraberg J, cinate, an inhibitor of cytosolic glutamate formation from glu- Zimmer J (2007) Neuroprotective effects of the AMPA antag- tamine, under anoxic conditions but not during exposure to onist PNQX in oxygen–glucose deprivation in mouse hippo- exogenous glutamate. Neurosci Lett 183:22–26 campal slice cultures and global cerebral ischemia in gerbils. 143. Lin TY, Lu CW, Wang SJ (2009) Inhibitory effect of glutamate Brain Res 1177:124–135 release from rat cerebrocortical synaptosomes by dextrometh- 161. O’Neill MJ, Bogaert L, Hicks CA et al (2000) LY377770, a orphan and its metabolite 3-hydroxymorphinan. Neurochem Int novel iGlu5 antagonist with neuroprotective 54:526–534 effects in global and focal cerebral ischaemia. Neuropharma- 144. Lai PC, Huang YT, Wu CC, Lai CJ, Wang PJ, Chiu TH (2011) cology 39:1575–1588 Ceftriaxone attenuates hypoxic-ischemic brain injury in neona- 162. Koh JY, Palmer E, Cotman CW (1991) Activation of the tal rats. J Biomed Sci 18:69 metabotropic glutamate receptor attenuates N-methyl-D-aspar- 145. Guan T, Qian Y, Tang X et al (2011) Maslinic acid, a natural tate neurotoxicity in cortical cultures. Proc Natl Acad Sci USA inhibitor of glycogen phosphorylase, reduces cerebral ischemic 88:9431–9435 injury in hyperglycemic rats by GLT-1 up-regulation. J Neurosci 163. Bao WL, Williams AJ, Faden AI, Tortella FC (2001) Selective Res 89:1829–1839 mGluR5 receptor antagonist or agonist provides neuroprotection 146. Verma R, Mishra V, Gupta K, Sasmal D, Raghubir R (2011) in a rat model of focal cerebral ischemia. Brain Res 922: Neuroprotection by rosiglitazone in transient focal cerebral 173–179 ischemia might not be mediated by glutamate transporter-1. 164. Bond A, Ragumoorthy N, Monn JA et al (1999) LY379268, a J Neurosci Res 89:1849–1858 potent and selective group II metabotropic glutamate receptor 147. Petty MA, Neumann-Haefelin C, Kalisch J, Sarhan S, Wettstein agonist, is neuroprotective in gerbil global, but not focal, cere- JG, Juretschke HP (2003) In vivo neuroprotective effects of bral ischaemia. Neurosci Lett 273:191–194 ACEA 1021 confirmed by magnetic resonance imaging in 165. Sabelhaus CF, Schro¨der UH, Breder J, Henrich-Noack P, ischemic stroke. Eur J Pharmacol 474:53–62 Reymann KG (2000) Neuroprotection against hypoxic/hypo- 148. Fang JH, Wang XH, Xu ZR, Jiang FG (2010) Neuroprotective glycaemic injury after the insult by the group III metabotropic effects of bis(7)-tacrine against glutamate-induced retinal gan- glutamate receptor agonist (R,S)-4-phosphonophenylglycine. Br glion cells damage. BMC Neurosci 11:31 J Pharmacol 131:655–658

123 Neurol Sci (2012) 33:223–237 237

166. Kohara A, Takahashi M, Yatsugi S et al (2008) Neuroprotective 174. Hoyte L, Barber PA, Buchan AM, Hill MD (2004) The rise and effects of the selective type 1 metabotropic glutamate receptor fall of NMDA antagonists for ischemic stroke. Curr Mol Med antagonist YM-202074 in rat stroke models. Brain Res 1191: 4:131–136 168–179 175. Walters MR, Kaste M, Lees KR et al (2005) The AMPA 167. Aronowski J, Strong R, Grotta JC (1996) Combined neuropro- antagonist ZK 200775 in patients with acute ischaemic stroke: a tection and reperfusion therapy for stroke. Effect of lubeluzole double-blind, multicentre, placebo-controlled safety and tolera- and diaspirin cross-linked hemoglobin in experimental focal bility study. Cerebrovasc Dis 20:304–309 ischemia. Stroke 27:1571–1576 176. Elting JW, Sulter GA, Kaste M et al (2002) AMPA antagonist 168. Bach A, Eildal JN, Stuhr-Hansen N et al (2011) Cell-permeable ZK200775 in patients with acute ischemic stroke: possible glial and plasma-stable peptidomimetic inhibitors of the postsynaptic cell toxicity detected by monitoring of S-100B serum levels. density-95/N-methyl-D-aspartate receptor interaction. J Med Stroke 33:2813–2818 Chem 54:1333–1346 177. Byrnes KR, Loane DJ, Faden AI (2009) Metabotropic glutamate 169. Duzenli S, Bakuridze K, Gepdiremen A (2005) The effects of receptors as targets for multipotential treatment of neurological ruthenium red, dantrolen and nimodipine alone or in combina- disorders. Neurotherapeutics 6:94–107 tion, in NMDA-induced neurotoxicity of cerebella granular cell 178. Matsumoto Y, Yamamoto S, Suzuki Y et al (2004) Na?/H? culture of rats. Toxicol In Vitro 19:589–594 exchanger inhibitor, SM-20220, is protective against excito- 170. Funato H, Katsuki Y, Yano T et al (1996) Effects of lacidipine, a toxicity in cultured cortical neurons. Stroke 35:185–190 new dihydropyridipine derivative, on various cerebral ischemia 179. Govoni S, Trabucchi M, Battaini F, Magnoni MS, Paoletti R (1986) models. Nippon Yakurigaku Zasshi 108:243–257 Vascular and neuronal mechanisms of calcium antagonists. Sig- 171. Choi SK, Lee GJ, Choi S, Kim YJ, Park HK, Park BJ (2011) nificance in neurological therapy. Minerva Med 77:1053–1058 Neuroprotective effects by nimodipine treatment in the experi- 180. Alps BJ (1992) Drugs acting on calcium channels: potential mental global ischemic rat model: real time estimation of glu- treatment for ischemic stroke. Br J Clin Pharmacol 34:199–206 tamate. J Korean Neurosurg Soc 49:1–7 181. Dawson VL, Kizushi VM, Huang PL, Snyder SH, Dawson TM 172. Kiewert C, Hartmann J, Stoll J, Thekkumkara TJ, Van der Schyf (1996) Resistance to neurotoxicity in cortical cultures from CJ, Klein J (2006) NGP1-01 is a brain-permeable dual blocker neuronal nitric oxide synthase-deficient mice. J Neurosci of neuronal voltage- and ligand-operated calcium channels. 16:2479–2487 Neurochem Res 31:395–399 182. Rothwell PM, Algra A, Amarenco P (2011) Medical treatment 173. Ginsberg MD (2008) Neuroprotection for ischemic stroke: past, in acute and long-term secondary prevention after transient present and future. Neuropharmacology 55:363–389 ischaemic attack and ischaemic stroke. Lancet 377:1681–1692

123