Skip to main content
Erschienen in: Molecular Brain 1/2013

Open Access 01.12.2013 | Research

NMDA receptor subunits have different roles in NMDA-induced neurotoxicity in the retina

verfasst von: Ning Bai, Tomomi Aida, Michiko Yanagisawa, Sayaka Katou, Kenji Sakimura, Masayoshi Mishina, Kohichi Tanaka

Erschienen in: Molecular Brain | Ausgabe 1/2013

Abstract

Background

Loss of retinal ganglion cells (RGCs) is a hallmark of various retinal diseases including glaucoma, retinal ischemia, and diabetic retinopathy. N-methyl-D-aspartate (NMDA)-type glutamate receptor (NMDAR)-mediated excitotoxicity is thought to be an important contributor to RGC death in these diseases. Native NMDARs are heterotetramers that consist of GluN1 and GluN2 subunits, and GluN2 subunits (GluN2A–D) are major determinants of the pharmacological and biophysical properties of NMDARs. All NMDAR subunits are expressed in RGCs in the retina. However, the relative contribution of the different GluN2 subunits to RGC death by excitotoxicity remains unclear.

Results

GluN2B- and GluN2D-deficiency protected RGCs from NMDA-induced excitotoxic retinal cell death. Pharmacological inhibition of the GluN2B subunit attenuated RGC loss in glutamate aspartate transporter deficient mice.

Conclusions

Our data suggest that GluN2B- and GluN2D-containing NMDARs play a critical role in NMDA-induced excitotoxic retinal cell death and RGC degeneration in glutamate aspartate transporter deficient mice. Inhibition of GluN2B and GluN2D activity is a potential therapeutic strategy for the treatment of several retinal diseases.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1756-6606-6-34) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

KT, NB, and TA conceived and designed the experiments. NB, SK and MY carried out experiments and analyzed the data. KS and MM contributed reagents and materials. KT and NB wrote the paper. All authors have read and approved the manuscript for publication.
Abkürzungen
GLAST
Glutamate aspartate transporter
INL
Inner nuclear layer
IPL
inner plexiform layer
IRL
Inner retina layer
LRP
lipoprotein receptor-related protein
NMDAR
N-methyl-D-aspartate receptor
PBS
Phosphate-buffered saline
PMSF
Phenylmethylsulfonyl fluoride
RGC
Retinal ganglion cell
RT-PCR
Reverse transcriptase polymerase chain reaction
WT
Wild-type.

Background

Glutamate is the major excitatory neurotransmitter in the mammalian central nervous system. However, its accumulation in extracellular spaces kills neurons through excitotoxic mechanisms via activation of glutamate receptors [1]. Excitotoxic neuronal cell death is thought to be a final common pathway in various neurological diseases, ranging from acute ischemic stroke to chronic neurodegenerative diseases such as Alzheimer’s disease and amyotrophic lateral sclerosis [25]. Glutamate excitotoxicity has also been proposed to be an important contributor to the death of retinal ganglion cells (RGCs) in glaucoma and ischemia-related conditions such as vessel occlusion and diabetic retinopathy [68], although some investigations have failed to confirm elevated glutamate concentration both in human patients with glaucoma [9] and in animal models of glaucoma [10, 11]. The toxic effects of glutamate on RGCs are predominantly mediated by the overstimulation of N-methyl-D-aspartate (NMDA)-type glutamate receptors (NMDARs) due to their extreme permeability to calcium ions [12].
NMDARs are composed of various combinations of GluN1 and GluN2 (GluN2A–GluN2D) subunits and, in some cases, GluN3 (GluN3A and GluN3B) subunits. GluN2 subunits are major determinants of the functional properties of NMDARs, including characteristics such as agonist affinity, deactivation kinetics, single-channel conductance, Ca2+ permeability, and sensitivity to Mg2+[13]. However, the relative contribution of different GluN2 subunits to RGC death by excitotoxicity remains unclear.
We previously reported that NMDAR-mediated excitotoxicity contributed to the degeneration of RGCs in glutamate aspartate transporter (GLAST) deficient (KO) mice, the first animal model of normal tension glaucoma [14]. Furthermore, we recently reported that GluN2D deficiency partially protected against the loss of RGCs in GLAST KO mice [15]. These results suggest that other GluN2 subunits, in addition to GluN2D, may contribute to excitotoxic retinal cell death. To address this hypothesis, we examined the roles of the four different GluN2 subtypes in NMDA-induced retinal cell death using mice lacking specific GluN2 subunits. We also evaluated the neuroprotective effect of 7-hydroxy-6-methoxy-2-methyl-1-(2-(4-(trifluoromethyl)phenyl)ethyl)-1,2,3,4-tetrahydroisoquinoline hydrochloride (HON0001) [16], an specific GluN2B antagonist, on RGC degeneration due to glutamate excitotoxicity in GLAST KO mice.
In the present study, we report that GluN2B and GluN2D deficiency protect against NMDA-induced excitotoxic retinal cell death, but GluN2A and GluN2C deficiency have no protective effects. We also show that pharmacological blockade of GluN2B subunit attenuates RGC loss in GLAST KO mice.

Results

NMDA receptor subunits present in mouse RGCs

To investigate the expression of NMDAR subunits in RGCs, we used a single-cell reverse transcriptase polymerase chain reaction (RT-PCR) method. After dissociation of the retina into single cells, RGCs can no longer be identified by their morphology. We therefore used dissociated retina from B6.Cg-TgN(Thy1-CFP)23Jrs/J transgenic mice (thy1-CFP mice), which express cyan fluorescent protein (CFP) in most RGCs [17]. We first confirmed that the CFP-containing cells in the thy1-CFP mouse retina were RGCs by immunostaining with Brn3, a neurochemical marker for RGCs [18]. CFP expression colocalized with Brn3 immunoreactivity in most somata in the ganglion cell layer (GCL) (Figure 1A-C). A single CFP-expressing cell was picked with a glass capillary from the dissociation mix and transferred to the reaction tube (Figure 1D, E), and was further identified as RGC by expression of Brn3 (Figure 1F). Typical results of single-cell RT-PCR on isolated RGCs are shown in Figure 1F. GluN1 and GluN2A–D could be amplified together with an internal control (β-actin) from a single RGC, as well as from whole retina. In our samples of 4 isolated RGCs, two cells express GluN1/GluN2A/GluN2B/GluN2C/GluN2D, whereas the other two cells express GluN1/GluN2A/GluN2B/GluN2D. These results indicate the presence of GluN1 and all GluN2 subunits (GluN2A–D) in the mouse RGCs.

Retinal structure in mice lacking GluN2 subunits

We used mice lacking any one of the four GluN2 subunits to determine the distinct roles of these GluN2 subunits in NMDA-induced RGC death. Mice lacking GluN2A, GluN2C, and GluN2D are viable [1921], whereas GluN2B-deficient mice die shortly after birth [22]. We therefore generated conditional GluN2B KO mice, in which GluN2B was ablated in retinal neurons containing RGCs. For this purpose, we crossed GluN2Bf/f[23] mice with c-kit-Cre mice [24] (GluN2Bf/f/c-kit-Cre). In c-kit-Cre mice crossed with ROSA-tdTomato reporter mice [25] (ROSA-tdTomato/c-kit-Cre), tdTomato-expressing cells were localized in the GCL and inner nuclear layer (INL) and most calretinin immunoreactive cells (RGCs and amacrine cells) contained tdTomato, suggesting that Cre recombinase is expressed in RGCs and cells in the INL, including amacrine cells, in c-kit-Cre mice (Figure 2A). Immunohistochemical analysis revealed that GluN2B protein expression was eliminated in RGCs and cells in the INL in GluN2Bf/f/c-kit-Cre mice (Figure 2B). Western blot analysis showed that GluN2A, GluN2C, and GluN2D proteins were completely eliminated from mutant mice lacking GluN2A, GluN2C, and GluN2D, respectively (Figure 2C, E, F). In GluN2Bf/f/c-kit-Cre mice, GluN2B expression level in the retina was significantly lower than in control mice (Figure 2D).
We next investigated whether the absence of GluN2 subunits affects the anatomical organization of the retina by histological analyses. Hematoxylin and eosin staining revealed the retinae of GluN2A, GluN2Bf/f/c-kit-Cre, GluN2C, and GluN2D mutant mice to be normally organized, consisting of several different cell layers (Figure 3A). The thickness of the inner retinal layer (IRL) in all mutant strains was normal compared with wild-type (WT) mice (Figure 3B). As previous studies showed that ablation of GluN1 increased cell death in the developing somatosensory thalamus [26], we counted cell numbers in the GCL. The cell number in the GCL of GluN2Bf/f/c-kit-Cre mice was significantly lower than that of WT mice at 5 weeks, whereas cell number in the GCL of the other mutant strains was comparable to that of control mice at 5 weeks (Figure 3C). These results suggest that GluN2B subunit plays a survival role for RGCs during retinal development, but the other GluN2 subunits (GluN2A, GluN2C and GluN2D) are not involved in retinal development and survival in RGCs.

GluN2B and GluN2D deficiency prevents NMDA-induced-excitotoxic retinal cell death

To determine which GluN2 subtypes are involved in NMDA-induced RGC death in the retina, we examined the effect of intraocular injection of NMDA on retinal cell death in GluN2 KO and WT mice. First, to examine the acute injury of NMDA, TUNEL analysis was performed on the retinas of WT and GluN2 mutants at 1 day after NMDA treatment. A number of TUNEL-positive cells were observed in the GCL and INL in both WT and GluN2 mutant strains after NMDA injection (Figure 4A), but the percentage of TUNEL-positive cells in the GCL of GluN2Bf/f/c-kit-Cre and GluN2D−/− mice was significantly lower than that in WT mice (Figure 4B). Following NMDA injection, the number of RGCs and the thickness of IRL decreased from days 1 to 7, with no further decrease being observed from days 7 to 14 [27, 28]. To examine the chronic injury of NMDA, morphological changes were measured 7 days after NMDA or phosphate-buffered saline (PBS) injection. Intraocular administration of NMDA induced cell death in the GCL in both WT and GluN2 mutant mice (Figure 4C), but the percentage of surviving cells in the GCL was significantly larger in GluN2Bf/f/c-kit-Cre and GluN2D−/− mice than in WT mice (Figure 4D). Additionally, the thickness of IRL was significantly larger in GluN2Bf/f/c-kit-Cre mice than in WT mice (Figure 4E). Taken together, these results suggest that GluN2B and GluN2D were involved in NMDA-induced RGC death.

A specific GluN2B antagonist, HON0001, prevents RGC death in GLAST-deficient mice

We have reported that the neuroprotective role of apolipoprotein E-containing lipoproteins in glaucomatous retinal degeneration in GLAST KO mice is mediated through promoting interaction between low density lipoprotein receptor-related protein 1 (LRP-1) and the GluN2B subunit [29]. Recently, we have also demonstrated that Dock3 overexpression prevented retinal cell death in GLAST KO mice by promoting GluN2B degradation [28]. To determine whether GluN2B is involved in RGC degeneration in GLAST-deficient mice, we evaluated the effect of a specific GluN2B antagonist, HON0001, on RGC degeneration in GLAST KO mice. As shown in Figure 5, the number of cells in GLAST KO mice subjected to HON0001 (10 mg/kg) treatment (281 ± 26) was significantly greater than that in GLAST KO mice not subjected to HON0001 treatment (203 ± 10). These results suggest that GluN2B is involved in RGC loss in GLAST KO mice.

Discussion

We previously reported that GluN2D deficiency prevented loss of RGCs in GLAST KO mice [15]. These results suggest that both GluN2B and GluN2D subunits play a critical role in RGC degeneration by glutamate excitotoxicity. Therefore, an GluN2B-selective antagonist in combination with an GluN2D-selective antagonist represents an effective strategy for the management of glaucoma and various forms of retinopathy. We recently showed that Dock3 overexpression prevented excitotoxic RGC death by suppressing the surface expression of GluN2D and enhancing NMDA-mediated GluN2B degradation [15, 28]. Thus, the design of compounds capable of increasing the expression of Dock3 represents a novel strategy for the treatment of various forms of retinopathy. Previous studies also showed that calcium influx through NMDARs is modulated by LRP-1 [30, 31]. These findings may provide a novel therapeutic strategy for various forms of retinopathy that are mediated by E-containing lipoproteins through LRP-1.
The failure of GluN2C deficiency to protect RGCs from NMDA-induced excitotoxicity can be explained by the data showing that only a small number of RGCs expressed GluN2C [32]. However, almost RGCs express GluN2A [32]. The failure of GluN2A deficiency to protect RGCs from NMDA-induced excitotoxicity may be explained by the distinct functional properties conferred by GluN2 subunits on the receptors, and the different signaling pathway couplings [13, 33]. This variety is due to the large and divergent cytoplasmic C-terminal domains of GluN2 subunits [34]. A previous report showed that C-terminal domains of GluN2B subunits were more lethal than GluN2A subunits, and different coupling to PSD-95/nNOS signaling cassette may contribute to differential susceptibility of GluN2 subunits to excitotoxic injury [33]. Another possible explanation is that the location of NMDARs at synaptic or extrasynaptic sites determines the neuroprotective or neurotoxic effects of glutamate. A high level of synaptic NMDAR activity promotes neuronal survival, whereas extrasynaptic NMDAR activity promotes cell death [35]. In the retina, GluN2B is enriched at the perisynaptic site, whereas synaptic NMDARs primarily contain GluN2A [36].
The number of cells in the GCL of GluN2Bf/f/c-kit-Cre mice was significantly decreased at 5 weeks. This finding is consistent with a previous study showing that NMDAR hypofunction increased neuronal death in the developing brain [26, 37]. GluN2B is a major GluN2 subunit in the immature retina [38]; therefore, ablation of GluN2B in the developing retina can cause excessive neuronal apoptosis, resulting in a reduction in the cell number in the GCL of GluN2Bf/f/c-kit-Cre mice. Thus, loss of GluN2B can increase RGC death in the immature retina, but protect RGCs from glutamate excitotoxicity in the adult.

Conclusions

We showed that GluN2B- and GluN2D-containing NMDARs played a critical role in NMDA-induced excitotoxic retinal cell death and RGC degeneration in GLAST KO mice. Inhibition of GluN2B and GluN2D activity is a potential therapeutic strategy for the treatment of several retinal diseases, including retinal ischemia, diabetic retinopathy, and glaucoma.

Methods

Animals

B6.Cg-TgN(Thy1-CFP)23Jrs/J transgenic mice (thy1-CFP mice) and c-kit-Cre transgenic mice have been described previously [17, 24]. c-kit-Cre transgenic mice were bred with ROSA-tdTomato mice [25] to examine Cre activity. c-kit-Cre mice were bred with GluN2Bflox/flox (GluN2B f/f ) mice [23] to generate GluN2B conditional knockout mice (GluN2B f/f /c-kit-Cre). The homozygous GluN2A KO (GluN2A −/− ) [19], GluN2C KO mice (GluN2C −/− ) [20] and GluN2D KO mice (GluN2D −/− ) [21] were obtained by crossing heterozygous GluN2A+/−, GluN2C+/− and GluN2D+/− mice, respectively. GLAST KO mice have been described previously [39, 40]. In all experiments, age matched WT and GluN2Bf/f littermate controls were used. All mice were of the C57BL/6 J genetic background, and all animal procedures were approved by the Animal Committee of Tokyo Medical and Dental University (0130166C).

Isolation of single ganglion cells from mouse retina and RT-PCR

5 week old Thy1-CFP mice were deeply anesthetized by diethyl ether and retinas were dissociated by using the Papain Dissociation System (Worthington Biochemical Corporation) at 37°C for 30 min. Single-CFP-expressing cell was aspirated by glass microcapillaries and placed into the PCR-tube containing 10 μl of resuspention buffer. Single-cell RT-PCR was performed using the SuperScript III CellsDirect cDNA Synthesis System (Invitrogen). Total RNA (5 μg) from whole retina were used to synthesize first-strand cDNA by using SuperScript III First-Strand Synthesis System (Invitrogen). The retina cDNA served as positive controls. The following primers were used for cDNA detection: GluN2A FWD: 5′ GTG TGC GAC CTC ATG TCC G 3′; REV: 5′ GCC TCT TGG TCC GTA TCA TCT C 3′; GluN2B FWD: 5′ CAG CAA AGC TCG TTC CCA AAA 3′; REV: 5′ GTC AGT CTC GTT CAT GGC TAC 3′; GluN2C FWD: ATC CCC GAC GGC TGA GA 3′; REV: 5′ TTC CTA GTC CAA GCA CAA AAC G 3′; GluN2D FWD: 5′ TGT GTG GGT GAT GAT GTT CGT 3′; REV: 5′ CCA CAG GAC TGA GGT ACT CAA AGA 3′; GluN1 FWD: 5′ GCC GAT TTA AGG TGA ACA GC 3′; REV: 5′ AAT TGT GCT TCT CCA TGT GC 3′; Brn3 FWD: 5′ GCA GTC TCC ACT TGG TGC TTA CTC 3′; REV: 5′ TTC CCC CTA CAA ACA AAC CTC C 3′; β-actin FWD: 5′ ATA TCG CTG CGC TGG TCG TC 3′; REV: 5′ TCA CTT ACC TGG TGC CTA GGG 3′. The thermal cycler conditions were 5 min at 94°C and then 40 cycles of 30 s at 94°C, 30 s at 60°C, and 30 s at 72°C, followed by 7 min at 72°C.

Western blot analysis

Retinas were quickly removed and homogenized in 100 μl of cold lysis buffer (50 mM Tris–HCl, 1% Nonidet P-40, 5 mM EDTA, 150 mM NaCl, 0.5% Na-deoxycholate, 1 mM MgCl2, 1 mM DTT, 1 mM Na3VO4, 1 mM NaF, 1 mM phenylmethylsulfonyl fluoride (PMSF), and Complete Protease Inhibitor Cocktail [Roche]). Protein concentration was determined by BCA Protein Assay kit (Sigma-Aldrich). Thirty microgram of the protein was loaded per lane. Primary antibodies used were GluN2A (1:500, Covance), GluN2B (1:500) [41], GluN2C (1:100, Invitrogen), GluN2D (1:500) [42], β-actin (1:1000, Santa Cruz). They were then incubated with anti-rabbit, guinea pig or mouse IgG-HRP-conjugated secondary antibody (1:5000, Jackson ImmunoResearch). SuperSignal West Femto Maximum Sensitivity Substrate (Thermo Scientific) was used to visualize the immunoreactive proteins.

Immunohistochemistry

Sections were prepared as previously described [15]. Frozen retinal sections of 12 μm thickness were incubated using anti-Brn3 (1:50, Santa Cruz), anti-calretinin (1:500, Swant) and anti-GluN2B (1:100) antibodies. For Brn3 and calretinin detection, Cy-3-conjugated donkey anti-goat IgG (1:500, Jackson ImmunoResearch) and goat anti-rabbit IgG Alexa 488 (1:1000, Molecular Probes) were used as secondary antibodies. For GluN2B detection, peroxidase labelled polymer conjugated to goat anti-rabbit IgG (DAKO) was used as secondary antibody. Images were recorded with an LSM-510 META confocal laser microscope (Carl Zeiss).

Histology and morphometric analysis

Eyes from mice at postnatal day 35 (P35) were enucleated and fixed in Davidson’s solution fixative [43], then embedded in paraffin wax. In some experiments, HON0001 (10 mg/kg, a gift from T. Honda at Hoshi University) [16] or saline was injected orally (p.o.) into GLAST KO mice daily from P21 to P35. These mice were sacrificed on P35 and processed for RGC count. Paraffin sections (7 μm thick) were cut though the optic nerve and stained with hematoxylin and eosin (H&E). The number of neurons in the GCL was counted as previously described [15]. The thickness of the IRL (from GCL to INL) was measured at a distance of 0.5 to 1.0 mm from optic disc.

Animal models of NMDA-induecd retinal neuronal death and morphometric analysis

Intravitreal injection of NMDA (Sigma) was conducted as previously described [15]. Briefly, a single 2-μl injection of 20 mM NMDA in 0.1 M PBS (pH 7.4) was administered intravitreally into the right eye of each mouse, the same volume of PBS was administered to the contralateral (left) eye as control. The animals were sacrificed at 1 day or 7 days after injection, and eyes were enucleated for morphometric and TUNEL analysis. Paraffin sections (5 μm thick) were cut though the optic nerve and stained with H&E. The extent of NMDA-induced retinal cell death after 7 days was quantified by counts of neurons in the GCL and the thickness of the IRL. The changes of the number of ganglion cells and thickness of IRL after NMDA injection were expressed as percentages of the control eyes.

TUNEL staining

At 1 day after the NMDA or PBS injection, TUNEL staining was performed with paraffin sections (5 μm thick) according to the manufacturer’s instructions (Promega). Fluorescence detection was carried out using Alexa-fluor-568-conjugated streptavidin (Molecular Probes). TUNEL-positive cells in the GCL were counted and expressed as percentages of total DAPI stained cells in the GCL.

Statistics

Statistical analyses were conducted using Student’s t-test for comparison between two samples, or one-way ANOVA followed by Bonferroni’s test for multiple comparisons, using the SPSS 17.0 software package. Data are expressed as mean ± S.E.M. P values < 0.05 were considered statistically significant.

Acknowledgements

We thank M. Watanabe and T. Honda for GluN2B and GluN2D antibody and for HON0001, respectively. We also thank Y. Hiraoka for technical support. This study was supported by “Understanding of molecular and environmental bases for brain health” executed under the Strategic Research Program for Brain Sciences from the Ministry of Education, Culture, Sports, Science and Technology, Japan (KT) and by the Ministry of health, Labor and Welfare of Japan (KT).
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

KT, NB, and TA conceived and designed the experiments. NB, SK and MY carried out experiments and analyzed the data. KS and MM contributed reagents and materials. KT and NB wrote the paper. All authors have read and approved the manuscript for publication.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

e.Med Neurologie

Kombi-Abonnement

Mit e.Med Neurologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes, den Premium-Inhalten der neurologischen Fachzeitschriften, inklusive einer gedruckten Neurologie-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Choi DW: Glutamate neurotoxicity and diseases of the nervous system. Neuron. 1988, 1: 623-634. 10.1016/0896-6273(88)90162-6.CrossRefPubMed Choi DW: Glutamate neurotoxicity and diseases of the nervous system. Neuron. 1988, 1: 623-634. 10.1016/0896-6273(88)90162-6.CrossRefPubMed
2.
Zurück zum Zitat Choi DW, Rothman SM: The role of glutamate neurotoxicity in hypoxic-ischemic neuronal death. Annu Rev Neurosci. 1990, 13: 171-182. 10.1146/annurev.ne.13.030190.001131.CrossRefPubMed Choi DW, Rothman SM: The role of glutamate neurotoxicity in hypoxic-ischemic neuronal death. Annu Rev Neurosci. 1990, 13: 171-182. 10.1146/annurev.ne.13.030190.001131.CrossRefPubMed
3.
Zurück zum Zitat Chapman AG: Glutamate and epilepsy. J Nutr. 2000, 130 (4S Suppl): 1043S-1045S.PubMed Chapman AG: Glutamate and epilepsy. J Nutr. 2000, 130 (4S Suppl): 1043S-1045S.PubMed
4.
Zurück zum Zitat Meldrum B: Amino acids as dietary excitotoxins: a contribution to understanding neurodegenerative disorders. Brain Res Brain Res Rev. 1993, 18: 293-314. 10.1016/0165-0173(93)90014-Q.CrossRefPubMed Meldrum B: Amino acids as dietary excitotoxins: a contribution to understanding neurodegenerative disorders. Brain Res Brain Res Rev. 1993, 18: 293-314. 10.1016/0165-0173(93)90014-Q.CrossRefPubMed
5.
Zurück zum Zitat Kalia LV, Kalia SK, Salter MW: NMDA receptors in clinical neurology: excitatory times ahead. Lancet Neurol. 2008, 7: 742-755. 10.1016/S1474-4422(08)70165-0.PubMedCentralCrossRefPubMed Kalia LV, Kalia SK, Salter MW: NMDA receptors in clinical neurology: excitatory times ahead. Lancet Neurol. 2008, 7: 742-755. 10.1016/S1474-4422(08)70165-0.PubMedCentralCrossRefPubMed
6.
Zurück zum Zitat Casson RJ: Possible role of excitotoxicity in the pathogenesis of glaucoma. Clin Experiment Ophthalmol. 2006, 34: 54-63. 10.1111/j.1442-9071.2006.01146.x.CrossRefPubMed Casson RJ: Possible role of excitotoxicity in the pathogenesis of glaucoma. Clin Experiment Ophthalmol. 2006, 34: 54-63. 10.1111/j.1442-9071.2006.01146.x.CrossRefPubMed
8.
Zurück zum Zitat Hernández C, Simó R: Neuroprotection in diabetic retinopathy. Curr Diab Rep. 2012, 12: 329-337. 10.1007/s11892-012-0284-5.CrossRefPubMed Hernández C, Simó R: Neuroprotection in diabetic retinopathy. Curr Diab Rep. 2012, 12: 329-337. 10.1007/s11892-012-0284-5.CrossRefPubMed
9.
Zurück zum Zitat Honkanen RA, Baruah S, Zimmerman MB, Khanna CL, Weaver YK, Narkiewicz J, Waziri R, Gehrs KM, Weingeist TA, Boldt HC, et al: Vitreous amino acid concentrations in patients with glaucoma undergoing vitrectomy. Arch Ophthalmol. 2003, 121: 183-188. 10.1001/archopht.121.2.183.CrossRefPubMed Honkanen RA, Baruah S, Zimmerman MB, Khanna CL, Weaver YK, Narkiewicz J, Waziri R, Gehrs KM, Weingeist TA, Boldt HC, et al: Vitreous amino acid concentrations in patients with glaucoma undergoing vitrectomy. Arch Ophthalmol. 2003, 121: 183-188. 10.1001/archopht.121.2.183.CrossRefPubMed
10.
Zurück zum Zitat Levkovitch-Verbin H, Martin KR, Quigley HA, Baumrind LA, Pease ME, Valenta D: Measurement of amino acid levels in the vitreous humor of rats after chronic intraocular pressure elevation or optic nerve transection. J Glaucoma. 2002, 11: 396-405. 10.1097/00061198-200210000-00005.CrossRefPubMed Levkovitch-Verbin H, Martin KR, Quigley HA, Baumrind LA, Pease ME, Valenta D: Measurement of amino acid levels in the vitreous humor of rats after chronic intraocular pressure elevation or optic nerve transection. J Glaucoma. 2002, 11: 396-405. 10.1097/00061198-200210000-00005.CrossRefPubMed
11.
Zurück zum Zitat Carter-Dawson L, Crawford ML, Harwerth RS, Smith EL, Feldman R, Shen FF, Mitchell CK, Whitetree A: Vitreal glutamate concentration in monkeys with experimental glaucoma. Invest Ophthalmol Vis Sci. 2002, 43: 2633-2637.PubMed Carter-Dawson L, Crawford ML, Harwerth RS, Smith EL, Feldman R, Shen FF, Mitchell CK, Whitetree A: Vitreal glutamate concentration in monkeys with experimental glaucoma. Invest Ophthalmol Vis Sci. 2002, 43: 2633-2637.PubMed
12.
Zurück zum Zitat Ferreira IL, Duarte CB, Carvalho AP: Ca2+ influx through glutamate receptor-associated channels in retina cells correlates with neuronal cell death. Eur J Pharmacol. 1996, 302: 153-162. 10.1016/0014-2999(96)00044-1.CrossRefPubMed Ferreira IL, Duarte CB, Carvalho AP: Ca2+ influx through glutamate receptor-associated channels in retina cells correlates with neuronal cell death. Eur J Pharmacol. 1996, 302: 153-162. 10.1016/0014-2999(96)00044-1.CrossRefPubMed
13.
Zurück zum Zitat Cull-Candy S, Brickley S, Farrant M: NMDA receptor subunits: diversity, development and disease. Curr Opin Neurobiol. 2001, 11: 327-335. 10.1016/S0959-4388(00)00215-4.CrossRefPubMed Cull-Candy S, Brickley S, Farrant M: NMDA receptor subunits: diversity, development and disease. Curr Opin Neurobiol. 2001, 11: 327-335. 10.1016/S0959-4388(00)00215-4.CrossRefPubMed
14.
Zurück zum Zitat Harada T, Harada C, Nakamura K, Quah HM, Okumura A, Namekata K, Saeki T, Aihara M, Yoshida H, Mitani A, et al: The potential role of glutamate transporters in the pathogenesis of normal tension glaucoma. J Clin Invest. 2007, 117: 1763-1770. 10.1172/JCI30178.PubMedCentralCrossRefPubMed Harada T, Harada C, Nakamura K, Quah HM, Okumura A, Namekata K, Saeki T, Aihara M, Yoshida H, Mitani A, et al: The potential role of glutamate transporters in the pathogenesis of normal tension glaucoma. J Clin Invest. 2007, 117: 1763-1770. 10.1172/JCI30178.PubMedCentralCrossRefPubMed
15.
Zurück zum Zitat Bai N, Hayashi H, Aida T, Namekata K, Harada T, Mishina M, Tanaka K: Dock3 interaction with a glutamate-receptor NR2D subunit protects neurons from excitotoxicity. Mol Brain. 2013, 6: 22-10.1186/1756-6606-6-22.PubMedCentralCrossRefPubMed Bai N, Hayashi H, Aida T, Namekata K, Harada T, Mishina M, Tanaka K: Dock3 interaction with a glutamate-receptor NR2D subunit protects neurons from excitotoxicity. Mol Brain. 2013, 6: 22-10.1186/1756-6606-6-22.PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Suetake-Koga S, Shimazaki T, Takamori K, Chaki S, Kanuma K, Sekiguchi Y, Suzuki T, Kikuchi T, Matsui Y, Honda T: In vitro and antinociceptive profile of HON0001, an orally active NMDA receptor NR2B subunit antagonist. Pharmacol Biochem Behav. 2006, 84: 134-141. 10.1016/j.pbb.2006.04.018.CrossRefPubMed Suetake-Koga S, Shimazaki T, Takamori K, Chaki S, Kanuma K, Sekiguchi Y, Suzuki T, Kikuchi T, Matsui Y, Honda T: In vitro and antinociceptive profile of HON0001, an orally active NMDA receptor NR2B subunit antagonist. Pharmacol Biochem Behav. 2006, 84: 134-141. 10.1016/j.pbb.2006.04.018.CrossRefPubMed
17.
Zurück zum Zitat Feng G, Mellor RH, Bernstein M, Keller-Peck C, Nguyen QT, Wallace M, Nerbonne JM, Lichtman JW, Sanes JR: Imaging neuronal subsets in transgenic mice expressing multiple spectral variants of GFP. Neuron. 2000, 28: 41-51. 10.1016/S0896-6273(00)00084-2.CrossRefPubMed Feng G, Mellor RH, Bernstein M, Keller-Peck C, Nguyen QT, Wallace M, Nerbonne JM, Lichtman JW, Sanes JR: Imaging neuronal subsets in transgenic mice expressing multiple spectral variants of GFP. Neuron. 2000, 28: 41-51. 10.1016/S0896-6273(00)00084-2.CrossRefPubMed
18.
Zurück zum Zitat Xiang M, Zhou L, Peng YW, Eddy RL, Shows TB, Nathans J: Brn-3b: a POU domain gene expressed in a subset of retinal ganglion cells. Neuron. 1993, 11: 689-701. 10.1016/0896-6273(93)90079-7.CrossRefPubMed Xiang M, Zhou L, Peng YW, Eddy RL, Shows TB, Nathans J: Brn-3b: a POU domain gene expressed in a subset of retinal ganglion cells. Neuron. 1993, 11: 689-701. 10.1016/0896-6273(93)90079-7.CrossRefPubMed
19.
Zurück zum Zitat Sakimura K, Kutsuwada T, Ito I, Manabe T, Takayama C, Kushiya E, Yagi T, Aizawa S, Inoue Y, Sugiyama H: Reduced hippocampal LTP and spatial learning in mice lacking NMDA receptor epsilon 1 subunit. Nature. 1995, 373: 151-155. 10.1038/373151a0.CrossRefPubMed Sakimura K, Kutsuwada T, Ito I, Manabe T, Takayama C, Kushiya E, Yagi T, Aizawa S, Inoue Y, Sugiyama H: Reduced hippocampal LTP and spatial learning in mice lacking NMDA receptor epsilon 1 subunit. Nature. 1995, 373: 151-155. 10.1038/373151a0.CrossRefPubMed
20.
Zurück zum Zitat Kadotani H, Hirano T, Masugi M, Nakamura K, Nakao K, Katsuki M, Nakanishi S: Motor discoordination results from combined gene disruption of the NMDA receptor NR2A and NR2C subunits, but not from single disruption of the NR2A or NR2C subunit. J Neurosci. 1996, 16: 7859-7867.PubMed Kadotani H, Hirano T, Masugi M, Nakamura K, Nakao K, Katsuki M, Nakanishi S: Motor discoordination results from combined gene disruption of the NMDA receptor NR2A and NR2C subunits, but not from single disruption of the NR2A or NR2C subunit. J Neurosci. 1996, 16: 7859-7867.PubMed
21.
Zurück zum Zitat Ikeda K, Araki K, Takayama C, Inoue Y, Yagi T, Aizawa S, Mishina M: Reduced spontaneous activity of mice defective in the epsilon 4 subunit of the NMDA receptor channel. Brain Res Mol Brain Res. 1995, 33: 61-71. 10.1016/0169-328X(95)00107-4.CrossRefPubMed Ikeda K, Araki K, Takayama C, Inoue Y, Yagi T, Aizawa S, Mishina M: Reduced spontaneous activity of mice defective in the epsilon 4 subunit of the NMDA receptor channel. Brain Res Mol Brain Res. 1995, 33: 61-71. 10.1016/0169-328X(95)00107-4.CrossRefPubMed
22.
Zurück zum Zitat Kutsuwada T, Sakimura K, Manabe T, Takayama C, Katakura N, Kushiya E, Natsume R, Watanabe M, Inoue Y, Yagi T, et al: Impairment of suckling response, trigeminal neuronal pattern formation, and hippocampal LTD in NMDA receptor epsilon 2 subunit mutant mice. Neuron. 1996, 16: 333-344. 10.1016/S0896-6273(00)80051-3.CrossRefPubMed Kutsuwada T, Sakimura K, Manabe T, Takayama C, Katakura N, Kushiya E, Natsume R, Watanabe M, Inoue Y, Yagi T, et al: Impairment of suckling response, trigeminal neuronal pattern formation, and hippocampal LTD in NMDA receptor epsilon 2 subunit mutant mice. Neuron. 1996, 16: 333-344. 10.1016/S0896-6273(00)80051-3.CrossRefPubMed
23.
Zurück zum Zitat Akashi K, Kakizaki T, Kamiya H, Fukaya M, Yamasaki M, Abe M, Natsume R, Watanabe M, Sakimura K: NMDA receptor GluN2B (GluR epsilon 2/NR2B) subunit is crucial for channel function, postsynaptic macromolecular organization, and actin cytoskeleton at hippocampal CA3 synapses. J Neurosci. 2009, 29: 10869-10882. 10.1523/JNEUROSCI.5531-08.2009.CrossRefPubMed Akashi K, Kakizaki T, Kamiya H, Fukaya M, Yamasaki M, Abe M, Natsume R, Watanabe M, Sakimura K: NMDA receptor GluN2B (GluR epsilon 2/NR2B) subunit is crucial for channel function, postsynaptic macromolecular organization, and actin cytoskeleton at hippocampal CA3 synapses. J Neurosci. 2009, 29: 10869-10882. 10.1523/JNEUROSCI.5531-08.2009.CrossRefPubMed
24.
Zurück zum Zitat Eriksson B, Bergqvist I, Eriksson M, Holmberg D: Functional expression of Cre recombinase in sub-regions of mouse CNS and retina. FEBS Lett. 2000, 479: 106-110. 10.1016/S0014-5793(00)01893-7.CrossRefPubMed Eriksson B, Bergqvist I, Eriksson M, Holmberg D: Functional expression of Cre recombinase in sub-regions of mouse CNS and retina. FEBS Lett. 2000, 479: 106-110. 10.1016/S0014-5793(00)01893-7.CrossRefPubMed
25.
Zurück zum Zitat Madisen L, Zwingman TA, Sunkin SM, Oh SW, Zariwala HA, Gu H, Ng LL, Palmiter RD, Hawrylycz MJ, Jones AR, et al: A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat Neurosci. 2010, 13: 133-140. 10.1038/nn.2467.PubMedCentralCrossRefPubMed Madisen L, Zwingman TA, Sunkin SM, Oh SW, Zariwala HA, Gu H, Ng LL, Palmiter RD, Hawrylycz MJ, Jones AR, et al: A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat Neurosci. 2010, 13: 133-140. 10.1038/nn.2467.PubMedCentralCrossRefPubMed
26.
Zurück zum Zitat Adams SM, de Rivero Vaccari JC, Corriveau RA: Pronounced cell death in the absence of NMDA receptors in the developing somatosensory thalamus. J Neurosci. 2004, 24: 9441-9450. 10.1523/JNEUROSCI.3290-04.2004.CrossRefPubMed Adams SM, de Rivero Vaccari JC, Corriveau RA: Pronounced cell death in the absence of NMDA receptors in the developing somatosensory thalamus. J Neurosci. 2004, 24: 9441-9450. 10.1523/JNEUROSCI.3290-04.2004.CrossRefPubMed
27.
Zurück zum Zitat Endo K, Nakamachi T, Seki T, Kagami N, Wada Y, Nakamura K, Kishimoto K, Hori M, Tsuchikawa D, Shinntani N, et al: Neuroprotective effect of PACAP against NMDA-induced retinal damage in the mouse. J Mol Neurosci. 2011, 43: 22-29. 10.1007/s12031-010-9434-x.CrossRefPubMed Endo K, Nakamachi T, Seki T, Kagami N, Wada Y, Nakamura K, Kishimoto K, Hori M, Tsuchikawa D, Shinntani N, et al: Neuroprotective effect of PACAP against NMDA-induced retinal damage in the mouse. J Mol Neurosci. 2011, 43: 22-29. 10.1007/s12031-010-9434-x.CrossRefPubMed
28.
Zurück zum Zitat Namekata K, Kimura A, Kawamura K, Guo X, Harada C, Tanaka K, Harada T: Dock3 attenuates neural cell death due to NMDA neurotoxicity and oxidative stress in a mouse model of normal tension glaucoma. Cell Death Differ. 2013, 10.1038/cdd.2013.91. Namekata K, Kimura A, Kawamura K, Guo X, Harada C, Tanaka K, Harada T: Dock3 attenuates neural cell death due to NMDA neurotoxicity and oxidative stress in a mouse model of normal tension glaucoma. Cell Death Differ. 2013, 10.1038/cdd.2013.91.
29.
Zurück zum Zitat Hayashi H, Eguchi Y, Fukuchi-Nakaishi Y, Takeya M, Nakagata N, Tanaka K, Vance JE, Tanihara H: A potential neuroprotective role of apolipoprotein E-containing lipoproteins through low density lipoprotein receptor-related protein 1 in normal tension glaucoma. J Biol Chem. 2012, 287: 25395-25406. 10.1074/jbc.M112.370130.PubMedCentralCrossRefPubMed Hayashi H, Eguchi Y, Fukuchi-Nakaishi Y, Takeya M, Nakagata N, Tanaka K, Vance JE, Tanihara H: A potential neuroprotective role of apolipoprotein E-containing lipoproteins through low density lipoprotein receptor-related protein 1 in normal tension glaucoma. J Biol Chem. 2012, 287: 25395-25406. 10.1074/jbc.M112.370130.PubMedCentralCrossRefPubMed
30.
Zurück zum Zitat Qiu Z, Strickland DK, Hyman BT, Rebeck GW: alpha 2-Macroglobulin exposure reduces calcium responses to N-methyl-D-aspartate via low density lipoprotein receptor-related protein in cultured hippocampal neurons. J Biol Chem. 2002, 277: 14458-14466. 10.1074/jbc.M112066200.CrossRefPubMed Qiu Z, Strickland DK, Hyman BT, Rebeck GW: alpha 2-Macroglobulin exposure reduces calcium responses to N-methyl-D-aspartate via low density lipoprotein receptor-related protein in cultured hippocampal neurons. J Biol Chem. 2002, 277: 14458-14466. 10.1074/jbc.M112066200.CrossRefPubMed
31.
Zurück zum Zitat Martin AM, Kuhlmann C, Trossbach S, Jaeger S, Waldron E, Roebroek A, Luhmann HJ, Laatsch A, Weggen S, Lessmann V, et al: The functional role of the second NPXY motif of the LRP1 beta-chain in tissue-type plasminogen activator-mediated activation of N-methyl-D-aspartate receptors. J Biol Chem. 2008, 283: 12004-12013. 10.1074/jbc.M707607200.CrossRefPubMed Martin AM, Kuhlmann C, Trossbach S, Jaeger S, Waldron E, Roebroek A, Luhmann HJ, Laatsch A, Weggen S, Lessmann V, et al: The functional role of the second NPXY motif of the LRP1 beta-chain in tissue-type plasminogen activator-mediated activation of N-methyl-D-aspartate receptors. J Biol Chem. 2008, 283: 12004-12013. 10.1074/jbc.M707607200.CrossRefPubMed
32.
Zurück zum Zitat Jakobs TC, Ben Y, Masland RH: Expression of mRNA for glutamate receptor subunits distinguishes the major classes of retinal neurons, but is less specific for individual cell types. Mol Vis. 2007, 13: 933-948.PubMedCentralPubMed Jakobs TC, Ben Y, Masland RH: Expression of mRNA for glutamate receptor subunits distinguishes the major classes of retinal neurons, but is less specific for individual cell types. Mol Vis. 2007, 13: 933-948.PubMedCentralPubMed
33.
Zurück zum Zitat Martel MA, Ryan TJ, Bell KF, Fowler JH, McMahon A, Al-Mubarak B, Komiyama NH, Horsburgh K, Kind PC, Grant SG, et al: The subtype of GluN2 C-terminal domain determines the response to excitotoxic insults. Neuron. 2012, 74: 543-556. 10.1016/j.neuron.2012.03.021.PubMedCentralCrossRefPubMed Martel MA, Ryan TJ, Bell KF, Fowler JH, McMahon A, Al-Mubarak B, Komiyama NH, Horsburgh K, Kind PC, Grant SG, et al: The subtype of GluN2 C-terminal domain determines the response to excitotoxic insults. Neuron. 2012, 74: 543-556. 10.1016/j.neuron.2012.03.021.PubMedCentralCrossRefPubMed
34.
Zurück zum Zitat Ryan TJ, Emes RD, Grant SG, Komiyama NH: Evolution of NMDA receptor cytoplasmic interaction domains: implications for organisation of synaptic signalling complexes. BMC Neurosci. 2008, 9: 6-10.1186/1471-2202-9-6.PubMedCentralCrossRefPubMed Ryan TJ, Emes RD, Grant SG, Komiyama NH: Evolution of NMDA receptor cytoplasmic interaction domains: implications for organisation of synaptic signalling complexes. BMC Neurosci. 2008, 9: 6-10.1186/1471-2202-9-6.PubMedCentralCrossRefPubMed
35.
Zurück zum Zitat Hardingham GE, Bading H: Synaptic versus extrasynaptic NMDA receptor signalling: implications for neurodegenerative disorders. Nat Rev Neurosci. 2010, 11: 682-696. 10.1038/nrn2911.PubMedCentralCrossRefPubMed Hardingham GE, Bading H: Synaptic versus extrasynaptic NMDA receptor signalling: implications for neurodegenerative disorders. Nat Rev Neurosci. 2010, 11: 682-696. 10.1038/nrn2911.PubMedCentralCrossRefPubMed
36.
Zurück zum Zitat Zhang J, Diamond JS: Subunit- and pathway-specific localization of NMDA receptors and scaffolding proteins at ganglion cell synapses in rat retina. J Neurosci. 2009, 29: 4274-4286. 10.1523/JNEUROSCI.5602-08.2009.PubMedCentralCrossRefPubMed Zhang J, Diamond JS: Subunit- and pathway-specific localization of NMDA receptors and scaffolding proteins at ganglion cell synapses in rat retina. J Neurosci. 2009, 29: 4274-4286. 10.1523/JNEUROSCI.5602-08.2009.PubMedCentralCrossRefPubMed
37.
Zurück zum Zitat Ikonomidou C, Bosch F, Miksa M, Bittigau P, Vöckler J, Dikranian K, Tenkova TI, Stefovska V, Turski L, Olney JW: Blockade of NMDA receptors and apoptotic neurodegeneration in the developing brain. Science. 1999, 283: 70-74. 10.1126/science.283.5398.70.CrossRefPubMed Ikonomidou C, Bosch F, Miksa M, Bittigau P, Vöckler J, Dikranian K, Tenkova TI, Stefovska V, Turski L, Olney JW: Blockade of NMDA receptors and apoptotic neurodegeneration in the developing brain. Science. 1999, 283: 70-74. 10.1126/science.283.5398.70.CrossRefPubMed
38.
Zurück zum Zitat Watanabe M, Mishina M, Inoue Y: Differential distributions of the NMDA receptor channel subunit mRNAs in the mouse retina. Brain Res. 1994, 634: 328-332. 10.1016/0006-8993(94)91938-0.CrossRefPubMed Watanabe M, Mishina M, Inoue Y: Differential distributions of the NMDA receptor channel subunit mRNAs in the mouse retina. Brain Res. 1994, 634: 328-332. 10.1016/0006-8993(94)91938-0.CrossRefPubMed
39.
Zurück zum Zitat Harada T, Harada C, Watanabe M, Inoue Y, Sakagawa T, Nakayama N, Sasaki S, Okuyama S, Watase K, Wada K, et al: Functions of the two glutamate transporters GLAST and GLT-1 in the retina. Proc Natl Acad Sci USA. 1998, 95: 4663-4666. 10.1073/pnas.95.8.4663.PubMedCentralCrossRefPubMed Harada T, Harada C, Watanabe M, Inoue Y, Sakagawa T, Nakayama N, Sasaki S, Okuyama S, Watase K, Wada K, et al: Functions of the two glutamate transporters GLAST and GLT-1 in the retina. Proc Natl Acad Sci USA. 1998, 95: 4663-4666. 10.1073/pnas.95.8.4663.PubMedCentralCrossRefPubMed
40.
Zurück zum Zitat Watase K, Hashimoto K, Kano M, Yamada K, Watanabe M, Inoue Y, Okuyama S, Sakagawa T, Ogawa S, Kawashima N, et al: Motor discoordination and increased susceptibility to cerebellar injury in GLAST mutant mice. Eur J Neurosci. 1998, 10: 976-988. 10.1046/j.1460-9568.1998.00108.x.CrossRefPubMed Watase K, Hashimoto K, Kano M, Yamada K, Watanabe M, Inoue Y, Okuyama S, Sakagawa T, Ogawa S, Kawashima N, et al: Motor discoordination and increased susceptibility to cerebellar injury in GLAST mutant mice. Eur J Neurosci. 1998, 10: 976-988. 10.1046/j.1460-9568.1998.00108.x.CrossRefPubMed
41.
Zurück zum Zitat Watanabe M, Fukaya M, Sakimura K, Manabe T, Mishina M, Inoue Y: Selective scarcity of NMDA receptor channel subunits in the stratum lucidum (mossy fibre-recipient layer) of the mouse hippocampal CA3 subfield. Eur J Neurosci. 1998, 10: 478-487. 10.1046/j.1460-9568.1998.00063.x.CrossRefPubMed Watanabe M, Fukaya M, Sakimura K, Manabe T, Mishina M, Inoue Y: Selective scarcity of NMDA receptor channel subunits in the stratum lucidum (mossy fibre-recipient layer) of the mouse hippocampal CA3 subfield. Eur J Neurosci. 1998, 10: 478-487. 10.1046/j.1460-9568.1998.00063.x.CrossRefPubMed
42.
Zurück zum Zitat Wu Y, Kawakami R, Shinohara Y, Fukaya M, Sakimura K, Mishina M, Watanabe M, Ito I, Shigemoto R: Target-cell-specific left-right asymmetry of NMDA receptor content in schaffer collateral synapses in epsilon1/NR2A knock-out mice. J Neurosci. 2005, 25: 9213-9226. 10.1523/JNEUROSCI.2134-05.2005.CrossRefPubMed Wu Y, Kawakami R, Shinohara Y, Fukaya M, Sakimura K, Mishina M, Watanabe M, Ito I, Shigemoto R: Target-cell-specific left-right asymmetry of NMDA receptor content in schaffer collateral synapses in epsilon1/NR2A knock-out mice. J Neurosci. 2005, 25: 9213-9226. 10.1523/JNEUROSCI.2134-05.2005.CrossRefPubMed
43.
Zurück zum Zitat Chi ZL, Akahori M, Obazawa M, Minami M, Noda T, Nakaya N, Tomarev S, Kawase K, Yamamoto T, Noda S, et al: Overexpression of optineurin E50K disrupts Rab8 interaction and leads to a progressive retinal degeneration in mice. Hum Mol Genet. 2010, 19: 2606-2615. 10.1093/hmg/ddq146.PubMedCentralCrossRefPubMed Chi ZL, Akahori M, Obazawa M, Minami M, Noda T, Nakaya N, Tomarev S, Kawase K, Yamamoto T, Noda S, et al: Overexpression of optineurin E50K disrupts Rab8 interaction and leads to a progressive retinal degeneration in mice. Hum Mol Genet. 2010, 19: 2606-2615. 10.1093/hmg/ddq146.PubMedCentralCrossRefPubMed
Metadaten
Titel
NMDA receptor subunits have different roles in NMDA-induced neurotoxicity in the retina
verfasst von
Ning Bai
Tomomi Aida
Michiko Yanagisawa
Sayaka Katou
Kenji Sakimura
Masayoshi Mishina
Kohichi Tanaka
Publikationsdatum
01.12.2013
Verlag
BioMed Central
Erschienen in
Molecular Brain / Ausgabe 1/2013
Elektronische ISSN: 1756-6606
DOI
https://doi.org/10.1186/1756-6606-6-34

Weitere Artikel der Ausgabe 1/2013

Molecular Brain 1/2013 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Thrombektomie auch bei großen Infarkten von Vorteil

16.05.2024 Ischämischer Schlaganfall Nachrichten

Auch ein sehr ausgedehnter ischämischer Schlaganfall scheint an sich kein Grund zu sein, von einer mechanischen Thrombektomie abzusehen. Dafür spricht die LASTE-Studie, an der Patienten und Patientinnen mit einem ASPECTS von maximal 5 beteiligt waren.

Schwindelursache: Massagepistole lässt Otholiten tanzen

14.05.2024 Benigner Lagerungsschwindel Nachrichten

Wenn jüngere Menschen über ständig rezidivierenden Lagerungsschwindel klagen, könnte eine Massagepistole der Auslöser sein. In JAMA Otolaryngology warnt ein Team vor der Anwendung hochpotenter Geräte im Bereich des Nackens.

Schützt Olivenöl vor dem Tod durch Demenz?

10.05.2024 Morbus Alzheimer Nachrichten

Konsumieren Menschen täglich 7 Gramm Olivenöl, ist ihr Risiko, an einer Demenz zu sterben, um mehr als ein Viertel reduziert – und dies weitgehend unabhängig von ihrer sonstigen Ernährung. Dafür sprechen Auswertungen zweier großer US-Studien.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.