Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2004

Open Access 01.12.2004 | Research

Induction of serine racemase expression and D-serine release from microglia by amyloid β-peptide

verfasst von: Sheng-Zhou Wu, Angela M Bodles, Mandy M Porter, W Sue T Griffin, Anthony S Basile, Steven W Barger

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2004

Abstract

Background

Roles for excitotoxicity and inflammation in Alzheimer's disease have been hypothesized. Proinflammatory stimuli, including amyloid β-peptide (Aβ), elicit a release of glutamate from microglia. We tested the possibility that a coagonist at the NMDA class of glutamate receptors, D-serine, could respond similarly.

Methods

Cultured microglial cells were exposed to Aβ. The culture medium was assayed for levels of D-serine by HPLC and for effects on calcium and survival on primary cultures of rat hippocampal neurons. Microglial cell lysates were examined for the levels of mRNA and protein for serine racemase, the enzyme that forms D-serine from L-serine. The racemase mRNA was also assayed in Alzheimer hippocampus and age-matched controls. A microglial cell line was transfected with a luciferase reporter construct driven by the putative regulatory region of human serine racemase.

Results

Conditioned medium from Aβ-treated microglia contained elevated levels of D-serine. Bioassays of hippocampal neurons with the microglia-conditioned medium indicated that Aβ elevated a NMDA receptor agonist that was sensitive to an antagonist of the D-serine/glycine site (5,7-dicholorokynurenic acid; DCKA) and to enzymatic degradation of D-amino acids by D-amino acid oxidase (DAAOx). In the microglia, Aβ elevated steady-state levels of dimeric serine racemase, the apparent active form of the enzyme. Promoter-reporter and mRNA analyses suggest that serine racemase is transcriptionally induced by Aβ. Finally, the levels of serine racemase mRNA were elevated in Alzheimer's disease hippocampus, relative to age-matched controls.

Conclusions

These data suggest that Aβ could contribute to neurodegeneration through stimulating microglia to release cooperative excitatory amino acids, including D-serine.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1742-2094-1-2) contains supplementary material, which is available to authorized users.

Competing interests

None declared.

Authors' contributions

Author 1 (S-Z.W.) performed the calcium measurements, neuronal survival experiments, DAAOx controls; participated in RT-PCR; cloned the racemase promoter and performed the luciferase assays; and composed the first draft of the manuscript. Author 2 (A.M.B.) produced the primary microglial cultures, performed western blot analyses and participated in the neuronal survival experiments. Author 3 (M.M.P.) was primarily responsible for RT-PCR. Author 4 (W.S.T.G.) provided the RNA samples from characterized human cases and controls. Author 5 (A.S.B.) performed the HPLC measurements and participated in the design of the study. Author 6 (S.W.B.) conceived of the study, participated in its design and coordination, performed feasibility studies for the calcium measurements and neuronal survival assays, and wrote the final draft of the manuscript. All authors read and approved the final manuscript.
Abkürzungen
amyloid β-peptide
DAAOx
D-amino acid oxidase
DCKA
5,7-dicholorokynurenate
HPLC
high pressure liquid chromatography
LDH
lactate dehydrogenase
LPS
lipopolysaccharide
MTT
methyltetrazolium
NMDA
N-methyl D-aspartate
RT-PCR
reverse-transcriptase polymerase chain reaction
sAPP
secreted amyloid precursor protein
TTX
tetrodotoxin.
Alzheimer's disease (AD) involves neuronal cell loss and reductions of synaptic density in specific brain regions. Some of the pathological signatures of AD implicate the process of excitotoxicity. For instance, glutamate receptors are altered in the AD brain [1], which also shows evidence of activation of the calcium-triggered protease calpain [2]. A glutamate receptor antagonist can reverse deficiencies in synaptic transmission in a mouse model of AD [3]. Elevations in glutamatergic stimulation may also contribute to several other neurodegenerative conditions [4].
Most excitotoxic paradigms involve NMDA receptors, complex ligand-gated calcium/sodium channels. In addition to glutamate, the NMDA receptors require a co-agonist at a second site. Glycine has been the most extensively studied ligand for this site. However, D-serine shows an approximately three-fold greater potency than glycine at this site [57]. D-serine satisfies several criteria for a neurotransmitter or -modulator at NMDA receptors: selective localization, controlled release, and physiological effect. Inactivation of D-serine by D-amino acid oxidase (DAAOx) markedly reduces NMDA neurotransmission as monitored by NO synthase activity and electrophysiology in ex vivo cerebellar and hippocampal preparations [8]. Furthermore, injection of D-serine can modulate NMDA receptor function in vivo [9, 10]. D-serine is generated from the more prevalent L-serine by serine racemase (EC 5-1-1). Regulation of expression of serine racemase has not been characterized, but under normal conditions the enzyme is localized to the D-serine-containing protoplasmic astrocytes in areas of the brain rich in NMDA receptors [11].
We recently reported that some of the derivatives of the β-amyloid precursor protein (βAPP), including amyloid β-peptide (Aβ), can stimulate glutamate release from microglial cells [12]. Aβ has been reported by many laboratories to activate an inflammatory phenotype in microglia, including the elevation of phagocytic activity, cytokine expression, and production of NO and reactive oxygen species [1315]. Because of the ability of D-serine to cooperate with glutamate in physiological and pathological stimulation of the NMDA receptor, we tested whether proinflammatory stimuli could influence the synthesis and/or release of D-serine in microglia as well.

Materials and methods

Materials

1–42 was purchased from Anaspec (San Jose CA). Lyophilized peptide was dissolved in anhydrous dimethyl sulfoxide at 2 mM, diluted with minimal essential medium (Earle's salts) (MEM) to 150 μM and incubated 16–24 h at 37°C. Recombinant sAPPα was produced and purified as described previously [12]. Lipopolysaccharide (LPS) and 5,7-dicholorokynurenic acid (DCKA) were from Sigma (St. Louis MO). D-amino acid oxidase (DAAOx) was from Worthington Biochemicals (Lakewood NJ); for heat-inactivation controls, DAAOx was incubated for 15 min at 80°C. The antibody against serine racemase was from Becton-Dickinson/Transduction Laboratories (Mississauga ON).

Cell culture

Primary microglia were obtained from mixed glial cultures generated from neonatal Sprague-Dawley rats as described previously [12]. Briefly, cortical tissue was dissociated and plated in MEM supplemented to 10% with fetal bovine serum (FBS), 0.5 mM L-glutamine, and 10 μg/mL gentamycin. After 10–14 days, microglia were removed by vigorous lavage and plated into secondary culture. A second lavage 30 min after secondary plating removed the astrocytes and oligodendrocytes; resulting secondary cultures were >95% microglia as determined by staining with Griffonia simplicifolia isolectin B4 and glial fibrillary acid protein (GFAP; exclusionary).
For RNA or protein harvest, cells were plated at 4 × 105/dish in 35-mm dishes. For collection of conditioned medium, cells were plated at 2 × 105/well in 24-well plates. Cultures were changed to serum-free MEM before stimulation.
Primary cultures of hippocampal neurons were established from E18 Sprague-Dawley rats as described previously [12]. Cultures were maintained in Neurobasal/B27 (Invitrogen) for 8–10 days before use in experiments. The N9 mouse microglial cell line (courtesy of P. Ricciardi-Castagnoli, Milan) and the HAPI rat microglial cell line (courtesy of J. R. Connor, Penn State U.) were maintained in MEM/10% FBS.

Measurement of D-serine

Reverse-phase HPLC was used to separate and detect D-serine in samples of conditioned medium, similar to the methods of Hashimoto et al. [16]. For these experiments, microglia were switched to MEM in which the concentration of L-glutamine had been reduced to 10 μM; otherwise, the glutamine elution peak obscured that of D-serine. Samples were derivatized by a 3:7 mixture of solution A (30 mg/mL t-BOC-L-cysteine, 30 mg/mL o-phthaldialdehyde in methanol):solution B (100 mM sodium tetraborate solution, pH 9.4). Resolution was achieved on two consecutive 4-μ NOVA-PAK C18 columns (Waters), 100 and 300 × 3.9 mm, respectively. A linear gradient was established from 100% buffer A (0.1 M sodium acetate buffer, pH 6; 7% acetonitrile; 3% tetrahydrofuran) to 100% buffer B (0.1 M sodium acetate buffer, pH 6; 47% acetonitrile; 3% tetrahydrofuran) over 120 min at 0.8 mL/min. Fluorescence was monitored with 344 nm excitation and 443 nm emission.

RT-PCR

Total RNA was isolated from microglial cultures and 1 μg was reverse-transcribed using the "Advantage RT-for-PCR" kit (Clontech); 1 μL of this product was used in PCR reactions with Clontech reagents. Serine racemase mRNA levels were often so low as to require a two-step PCR to avoid nonlinear effects of reagent depletion; the first PCR was 10 cycles and the second utilized 10% of this product in a 25-cycle reaction. Primers were designed to span an intron/exon junction. Mouse racemase; forward: 5'-GTT ACT CAC AGC AGC GGA AAC C; reverse: 5'-GAG GGC TCA GCA GCG TAT ACC (annealing at 61°C). Rat racemase; forward: 5'-TAG CGG GAC AAG GGA CAA TT; reverse: 5'-TGC ATA CTT GAT TTC ATC TTC CGT G (annealing at 61°C). Human racemase; forward: 5'-CTA TCC ACC TCA CAC CAG TGC TAA C; reverse: 5'-ACA ATT GAC GCT CCG TAG GCT (annealing temperature: 71°C). Equivalency of input was confirmed by RT-PCR for GAPDH as described previously [17].

Human subjects

Total mRNA was obtained from hippocampus of twelve persons (38% males, ages 60–92) diagnosed with Alzheimer's disease by CERAD criteria. Nine (9) age-matched controls (AMC) (87% male, ages 59–97) were free of other neurological conditions and heart disease.

Western blot analysis

Cell culture lysates were analyzed for serine racemase by immunoblotting techniques described previously [18], with the primary antibody diluted to 1:200. Blots were digitized on a conventional scanner.

Luciferase reporter assay

The serine racemase sequence representing nucleotides 1511 upstream of the start of translation was cloned from the T98G human cell line; fidelity was confirmed by sequencing. This sequence was fused to the coding region of firefly luciferase in pGL3-basic (Promega) to create pGL3-RaceProm. The pRL-TK vector (Promega) was used as a cotransfected control for transfection efficiency and cell survival. For each 2-cm2 transfection well, 2 μL Lipofectamine 2000 (Invitrogen) were mixed in MEM with 300 ng pGL3-RaceProm, 10 ng pRL-TK, and 690 ng miscellaneous DNA, and this mixture was incubated at room temperature for 20 min. The mixture was applied to HAPI cell cultures for 2 h, then removed by a medium change to fresh MEM with or without agonists as indicated. After an additional 24 h, a lysate of each culture was assayed sequentially for firefly luciferase and Renilla luciferase activity with a commercial kit (Promega).

Calcium measurements

Primary neurons were assayed for intracellular ionic calcium concentration ([Ca2+]i) as described previously [12]. Unless otherwise indicated, 800 nM tetrodotoxin was present during measurements. For DAAOx pretreatment of conditioned media samples, the enzyme was dissolved in the same buffer used during imaging, added to conditioned media at a final concentration of 100 μg/mL control, and the mixture was incubated for 7 min at 37°C. Control incubations were performed with media diluted with an equal volume of the imaging buffer.

Neuronal survival assay

Neurotoxicity was determined by measuring lactate dehydrogenase (LDH) released into the culture medium using a commercial kit (Sigma). Primary cultures from rat hippocampus were plated in 24-well plates, and glia were restricted by a two-day exposure to 1 μM cytosine arabinoside (AraC). Eight days after plating, neurons were treated with pharmacological agents and microglial conditioned medium. Aliquots of culture medium were assayed for LDH 24–48 h later. A survival index was generated wherein the lowest LDH reading from untreated conditioned medium was assigned a value of 100 (% survival) and the highest LDH reading from maximally lysed neurons was assigned a value of 0 (% survival). MTT assays were performed as described previously [19]. For tests of the effect of DAAOx, microglia-conditioned medium was incubated as described above for calcium measurements.

Results

As a first test of the role D-serine might play in Aβ-stimulated microglial neurotoxicity, we measured D-serine levels in microglia-conditioned medium. Reverse-phase HPLC was performed on media samples, and conditions were determined under which D-serine could be quantified. Treatment of primary microglia with Aβ1–42 for 20–24 h resulted in a large increase in D-serine in the medium (Fig. 1). The maximal D-serine concentration varied between experiments, ranging from 115 to 660 μM. LPS also evoked an increase in D-serine levels. Neither Aβ nor LPS caused an elevation of glycine levels, which typically approximated the resting levels of D-serine (e.g., Fig. 1B). MTT assays were also performed, excluding any artifacts of cell number or lysis (not shown).
D-serine is produced primarily by conversion from L-serine by serine racemase. This racemase is known to be expressed in protoplasmic astrocytes in vivo. To confirm its expression in microglia, semi-quantitative RT-PCR was performed on mRNA isolated from several culture types. Serine racemase expression was detected in cultures of primary rat microglia and appeared to increase after activation with Aβ (Fig. 2A). We also surveyed microglial cell lines to exclude possible astrocyte contamination. The HAPI rat microglial line contained serine racemase mRNA after treatment with Aβ (Fig. 2B), as did the N9 mouse microglial line in the presence two other proinflammatory stimuli: sAPPα and LPS (Fig. 2C). The mouse sequence was subcloned and sequenced to confirm identity.
The presence of serine racemase mRNA in activated microglia raised the possibility that increases in expression of this enzyme were responsible for the apparent elevations of D-serine release by Aβ, so western blot analysis was performed on cell lysates from primary microglia. In both cell lysates and positive control samples, the serine racemase antibody detected monomeric protein (~37 kD) and an apparent dimer (~74 kD) (Fig. 3); specificity of the detection was confirmed by a preabsorption control (Fig. 3A). Such oligomers of the enzyme have been described recently and appear to include its soluble, active forms [20]; as reported in that study, we found the serine racemase dimer to be insensitive to reducing agents. Exposure of primary microglia to Aβ had little or no effect on monomeric serine racemase but resulted in significantly higher levels of the apparent dimer (299% of control) (Fig. 3B). Similar inductions were observed in the HAPI microglial cell line.
To address the possibility of a transcriptional induction of serine racemase, a 1.5 kb sequence 5' to the luciferase coding region was cloned from human genomic DNA. This sequence was placed in the pGL3-basic plasmid for luciferase reporter assays. HAPI microglial cells were transfected with this construct and treated with either Aβ or LPS. After one day of treatment, luciferase levels indicated an induction of the presumptive serine racemase promoter by both stimuli (Fig. 4).
Previous experiments demonstrated a release of glutamate by microglia activated with sAPP and Aβ. Useful in those studies were bioassays in which hippocampal neurons were monitored for intracellular ionic calcium concentration ([Ca2+]i) during application of conditioned medium collected from control or activated microglia [12]. As an initial step to determine if proinflammatory activation of microglia might evoke release of NMDA-R agonists other than glutamate, we sought conditions suitable for detecting ligands of the glycine/D-serine site of the NMDA receptor. With no other manipulations, application of glutamate to hippocampal neurons elevated [Ca2+]i to levels that were partially inhibited by a glycine/D-serine site antagonist, 5,7-dicholorokynurenic (DCKA), suggesting synaptic release of endogenous glycine. To circumvent this effect in bioassays of conditioned medium, tetrodotoxin (TTX) was employed. This intervention resulted in a [Ca2+]i response to glutamate that was smaller and insensitive to DCKA (data not shown). Therefore, 800 nM TTX was included in subsequent bioassays of microglia conditioned medium. Under these conditions, graded responses to D-serine could be detected at concentrations from 3–100 μM (data not shown).
Bioassays were performed on conditioned medium from primary microglia activated with either Aβ1–42 or LPS. Hippocampal neurons responded to such media with a rapid increase in [Ca2+]i (Fig. 5). Conditioned medium from Aβ-treated microglia evoked a modest response at a dilution of 1:100 into the imaging buffer; a 1:18 dilution elevated [Ca2+]i dramatically. Medium from LPS-treated microglia had a similar effect (Fig. 5B). By contrast, the conditioned medium from unactivated sister cultures showed no effect on neuronal [Ca2+]i at ratios up to 1:18 (Fig. 5A) and evoked only a modest increase at 1:10 (Fig. 5B). Acute treatment of neurons with equivalent amounts of Aβ or LPS had no significant effect on [Ca2+]i. DCKA (100 μM) reversed the [Ca2+]i response to microglia-conditioned medium. The elevation was also sensitive to more general antagonists of the NMDA receptor (data not shown). As an independent test of the role of D-serine in the calcium responses evoked by microglia-conditioned medium, samples of media were incubated with D-amino acid oxidase (DAAOx) to remove D-serine; catalase was also added to the imaging buffer to obviate effects of H2O2 produced by the DAAOx. Treatment with DAAOx dramatically lowered the ability of microglia-conditioned medium to evoke a [Ca2+]i response (Fig. 5C). Similar elevations of a DAAOx-sensitive NMDA agonist were observed in medium conditioned by the HAPI microglial cell line. Several controls for the specificity of the DAAOx treatments were performed (data not shown): i. exogenous glycine was able to overcome the effect of DAAOx, confirming independence from hydrogen peroxide or similar artifacts of the DAAOx treatment; ii. when samples of conditioned media were treated with DAAOx that had been heat-inactivated, there was little difference from untreated media samples; iii. DAAOx was shown to be specific for D-serine under the conditions of incubation by tests in which the enzyme was incubated with [14C]glycine or [3H]D-serine, subsequently analyzed by thin-layer chromatography.
To explore the ramifications of D-serine release, we tested the influence of D-serine on neuronal health. Primary hippocampal neurons were exposed to 1 or 3 μM D-serine, and effects on metabolic activity were monitored by MTT assay the following day. Treatment with 1 μM D-serine lowered MTT values to 71.1% of control (±8.08), and 3 μM D-serine resulted in a value that was 11.54% of control (±7.50). D-serine also generally potentiated the toxicity of low levels of glutamate (data not shown).
We next tested whether D-serine played a requisite role in the neurotoxicity exhibited by Aβ-treated microglia. Primary microglia were left untreated or were exposed to Aβ overnight. Conditioned medium from these cells was applied to cultures of primary hippocampal neurons (Fig. 6). A higher rate of LDH release was observed in the presence of conditioned medium from Aβ-treated microglia compared to that obtained from untreated microglia. As a control for the potential neurotoxicity of residual Aβ carried over with the conditioned medium, an aliquot of Aβ was diluted into culture medium in a cell culture dish lacking cells and incubated under identical conditions; treatment of neurons with medium thus prepared showed no significant toxicity under the conditions of our assay (not shown). The neurotoxicity resulting from Aβ-activated microglia was partially reversed by inclusion of 1 or 10 μM DCKA (Fig. 6). Furthermore, pretreatment of the conditioned medium from Aβ-treated microglia with DAAOx also partially reversed its neurotoxicity. Because DAAOx can generate hydrogen peroxide, a separate treatment was tested utilizing catalase, but this did not alter the effect of DAAOx (data not shown).
Based on the inductions by Aβ and other proinflammatory stimuli, the levels of expression of serine racemase in AD brain tissue were examined. RT-PCR of mRNA isolated from hippocampus of AD indicated a significant elevation of serine racemase expression compared to age-matched controls (AMC) (Fig. 7). Within the AD group alone, the female subjects showed a nonsignificant trend towards higher levels than the male subjects. The AD pool contained a higher percentage of females, creating the possibility that gender contributed to the difference observed. However, the difference between AD and AMC was significant within males alone (ratios of racemase:GAPDH signals, AD: 1.62 ± 0.342; AMC: 0.447 ± 0.024; p < 0.05).

Discussion

The studies presented here document the capacity of activated microglia to express serine racemase and release D-serine, thereby implicating D-serine as a contributor to the neurotoxicity exhibited by inflammatory situations in the CNS. Microglia stimulated with Aβ or LPS released D-serine, a potent NMDA receptor coagonist. The conditioned medium from such microglia elevated [Ca2+]i in cultured hippocampal neurons in a manner that was largely reversed by D-serine/glycine-site antagonists, as well as more general antagonists of NMDA receptors. Pretreatment of the conditioned medium with DAAOx also blocked the effects on neuronal [Ca2+]i. Aβ treatment elevated the steady-state levels of serine racemase mRNA and protein, suggesting that increased synthesis may be involved in the release of D-serine observed under these conditions. Finally, a potential role for D-serine in Alzheimer's disease was further implicated by the observation that serine racemase mRNA is elevated in Alzheimer's brain tissue.
D-serine has gained increased scrutiny as a NMDA receptor agonist that may be more important than glycine in vivo, at least in specific regions or developmental stages. However, the potential contribution of D-serine in excitotoxic pathologies has not received much attention. Damage resulting from intracortical infusion of NMDA is attenuated by an inhibitor of poly-ADP ribose polymerase (PARP), and this neuroprotection was associated with a depression in the levels of D-serine but not glycine [21]. Conventional wisdom held that D-serine/glycine sites on NMDA receptors are typically saturated in vivo, making elevations in their agonists irrelevant. However, this idea has been refuted for over a decade now by observations of responsiveness to infused glycine [22]. Similarly, applications of D-serine have shown dramatic physiological effects [9, 10, 23, 24]. One set of results indicates that much of the biological action of exogenous D-serine may come from stimulation of extrasynaptic NMDA receptors [8]. To the extent that the actions of D-serine on NMDA receptors replicate those of glycine (perhaps, even more potently), the vast literature on exogenous glycine in excitotoxicity paradigms can be translated to D-serine. But issues of production, release, uptake, and catabolism appear distinct for these two glutamate co-agonists, making studies of D-serine a distinct priority.
Release of D-serine from astrocytes can be stimulated by non-NMDA, ionotropic glutamate receptor agonists [25]. This fact has led to the hypothesis that the synaptic elements of astrocytes may contribute to synaptic efficacy by participating in a positive-feedback loop whereby neuronal release of glutamate stimulates astrocytes to release D-serine and further amplify NMDA receptor activation [26]. Recently, data were published consistent with the possibility that serine racemase is activated by direct binding of calcium [20]; notably, AMPA/kainate receptor activation elevates intracellular calcium levels in astrocytes [27]. Therefore, a global release of glutamate – or, in fact, any strong calcium agonist – may lead to extrasynaptic release of D-serine, from both astrocytes and microglia. Similar to its effects on neurons, Aβ can elevate [Ca2+]i in microglia [28], as can LPS [29]. To wit, the degree of elevation in D-serine released into medium was surprisingly high given the changes in serine racemase protein levels, suggesting that some of the Aβ-evoked increase in D-serine release may have come from stimulation of enzyme activity, in addition to expression levels. Detailed time-course analyses of protein levels and D-serine release may provide some insight into this question.
In the experimental paradigms applied here, the presumptive actions of D-serine in microglia-conditioned medium were attenuated by DAAOx. Under the normal conditions of neurotransmission, glutamate concentrations at the synapse are reduced primarily by astrocyte uptake [30]. The mechanisms controlling D-serine concentrations are less clear; the relative contributions of degradation (e.g., by DAAOx), glial uptake, or diffusion out of the synaptic cleft are topics of ongoing research. There appears to be a transporter for D-serine at the synapse [31], but it is incompletely characterized. Neuronal uptake, perhaps to replenish presynaptic stores, would be consistent with the finding that some pyramidal neurons in the cerebral cortex and neurons in the nucleus of the trapezoid body contain D-serine [32]. Degradation of D-serine by DAAOx produces hydrogen peroxide, creating potential for additional harm. However, the concentrations of peroxide predicted from this reaction would be make a relatively minor contribution to neuropathology compared to the potent synergistic activation of NMDA receptors by D-serine and glutamate.
A role for excitotoxicity in CNS inflammation is becoming well established. One of the first analyses of the relative roles of various neurotoxins released by activated microglia found that NMDA receptor antagonists were the most efficacious neuroprotectants [33]. Subsequently, Giulian et al. [34] described an excitotoxin released from microglia exposed to amyloid plaques. Excitotoxicity appears to contribute to neuronal damage in more general models of inflammation as well, such as intracerebroventricular LPS infusion [35]. Several studies have concluded that nitric oxide (NO) mediates microglial neurotoxicity because inducible NO synthase (iNOS) responds to proinflammatory stimuli and general NOS inhibitors can be protective in micoglia-neuron cocultures [36]. However, most such experiments cannot distinguish between NO generated by microglia versus that generated by the neurons themselves through classic excitotoxic mechanisms [37]. When corrected for Ki, inhibitors selective for neuronal NOS are more potent protective agents than are iNOS-selective compounds [12], suggesting that the primary neurotoxic agents microglia produce are excitotoxins that activate nNOS to produce NO within the neurons themselves.
Previously, Li et al. [38] showed that microglial cells synthesize and release IL-1 in response to conditioned media obtained from glutamate-stressed neurons. The neurons respond with an increase in expression and processing of βAPP. Secreted APP and Aβ can stimulate proinflammatory activation in microglia [13, 18], including the release glutamate [12, 39]. These data are consistent with the plethora of evidence linking inflammatory mechanisms to AD pathogenesis [40]. Together with the potential for such stimuli to also trigger release of D-serine, these findings suggest that a vicious circle of inflammation and excitotoxicity may be important in AD pathogenesis. Excitotoxic events are a common aspect of many forms of neurodegeneration, even when they occur secondarily to ischemia or trauma, and considerable evidence suggests that excessive stimulation of glutamate receptors occurs in AD [13].
Free D-serine concentrations are reported to be unaltered in the Alzheimer brain [41, 42]. However, one study found an elevation of overall serine levels in AD CSF per unit volume, but when normalized to protein concentration, the serine levels were similar between AD and controls [43], suggesting that elevated protein levels in AD CSF could confound analyses and interpretations. Our initial analysis here indicates that there is an elevated steady-state level of serine racemase mRNA in AD hippocampus versus age-matched controls. Nevertheless, the elevation of D-serine itself might be expected to occur early in the disease progression; thus, any elevation might be difficult to detect after the disease has progressed to its final stages.
Our semi-quantitative analysis showed that the mRNA for serine racemase was increased in AD brain nearly three-fold relative to age-matched controls. It is possible that a portion of this difference can be accounted for by the hypothetical increase in numbers of astrocytes in AD. However, a similar analysis of GFAP mRNA in AD reported levels to be only 57% higher in AD than in controls [44], and this effect includes an augmented expression per cell [45]. A recent microarray analysis of AD concluded that GFAP mRNA could not be compared to controls reliably due to variability across post-mortem interval, agonal state, etc [46].
In conclusion, Aβ and other AD-relevant proinflammatory stimuli are capable of stimulating release of D-serine from microglia. Together with the release of glutamate evoked by similar conditions, a cooperative activation of NMDA receptors could be anticipated. In addition to delineating details of the mechanisms by which CNS inflammation harms neuronal elements, this line of evidence may be relevant to the development of therapies. If approaches targeting the general inflammatory system or glutamatergic neurotransmission are accompanied by unacceptable contraindications, a more specific interference with D-serine production or release may be more useful in AD and other neurodegenerative conditions. For this reason and others, it will be important to elucidate the mechanisms controlling D-serine synthesis, degradation, and transport under normal and pathological situations.

Acknowledgements

Supported by the National Institute of Aging (R01AG17498 & P01AG12411). We greatly appreciate Dr. Camilo Rojas (Guilford Pharmaceuticals), who generously provided recombinant serine racemase for preabsorption antibody controls, as well as helpful discussions. Richard Jones also provided technical assistance.

Competing interests

None declared.

Authors' contributions

Author 1 (S-Z.W.) performed the calcium measurements, neuronal survival experiments, DAAOx controls; participated in RT-PCR; cloned the racemase promoter and performed the luciferase assays; and composed the first draft of the manuscript. Author 2 (A.M.B.) produced the primary microglial cultures, performed western blot analyses and participated in the neuronal survival experiments. Author 3 (M.M.P.) was primarily responsible for RT-PCR. Author 4 (W.S.T.G.) provided the RNA samples from characterized human cases and controls. Author 5 (A.S.B.) performed the HPLC measurements and participated in the design of the study. Author 6 (S.W.B.) conceived of the study, participated in its design and coordination, performed feasibility studies for the calcium measurements and neuronal survival assays, and wrote the final draft of the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Jansen KL, Faull RL, Dragunow M, Synek BL: Alzheimer's disease: changes in hippocampal N-methyl-D-aspartate, quisqualate, neurotensin, adenosine, benzodiazepine, serotonin and opioid receptors--an autoradiographic study. Neuroscience. 1990, 39: 613-627. 10.1016/0306-4522(90)90246-Z.CrossRefPubMed Jansen KL, Faull RL, Dragunow M, Synek BL: Alzheimer's disease: changes in hippocampal N-methyl-D-aspartate, quisqualate, neurotensin, adenosine, benzodiazepine, serotonin and opioid receptors--an autoradiographic study. Neuroscience. 1990, 39: 613-627. 10.1016/0306-4522(90)90246-Z.CrossRefPubMed
2.
Zurück zum Zitat Saito K, Elce JS, Hamos JE, Nixon RA: Widespread activation of calcium-activated neutral proteinase (calpain) in the brain in Alzheimer disease: a potential molecular basis for neuronal degeneration. Proc. Natl. Acad. Sci. (USA). 1993, 90: 2628-2632.CrossRef Saito K, Elce JS, Hamos JE, Nixon RA: Widespread activation of calcium-activated neutral proteinase (calpain) in the brain in Alzheimer disease: a potential molecular basis for neuronal degeneration. Proc. Natl. Acad. Sci. (USA). 1993, 90: 2628-2632.CrossRef
3.
Zurück zum Zitat Fitzjohn SM, Morton RA, Kuenzi F, Rosahl TW, Shearman M, Lewis H, Smith D, Reynolds DS, Davies CH, Collingridge GL, Seabrook GR: Age-related impairment of synaptic transmission but normal long-term potentiation in transgenic mice that overexpress the human APP695SWE mutant form of amyloid precursor protein. J Neurosci. 2001, 21: 4691-4698.PubMed Fitzjohn SM, Morton RA, Kuenzi F, Rosahl TW, Shearman M, Lewis H, Smith D, Reynolds DS, Davies CH, Collingridge GL, Seabrook GR: Age-related impairment of synaptic transmission but normal long-term potentiation in transgenic mice that overexpress the human APP695SWE mutant form of amyloid precursor protein. J Neurosci. 2001, 21: 4691-4698.PubMed
4.
Zurück zum Zitat Doble A: The role of excitotoxicity in neurodegenerative disease: implications for therapy. Pharmacol Ther. 1999, 81: 163-221. 10.1016/S0163-7258(98)00042-4.CrossRefPubMed Doble A: The role of excitotoxicity in neurodegenerative disease: implications for therapy. Pharmacol Ther. 1999, 81: 163-221. 10.1016/S0163-7258(98)00042-4.CrossRefPubMed
5.
Zurück zum Zitat Fadda E, Danysz W, Wroblewski JT, Costa E: Glycine and D-serine increase the affinity of N-methyl-D-aspartate sensitive glutamate binding sites in rat brain synaptic membranes. Neuropharmacology. 1988, 27: 1183-1185. 10.1016/0028-3908(88)90015-9.CrossRefPubMed Fadda E, Danysz W, Wroblewski JT, Costa E: Glycine and D-serine increase the affinity of N-methyl-D-aspartate sensitive glutamate binding sites in rat brain synaptic membranes. Neuropharmacology. 1988, 27: 1183-1185. 10.1016/0028-3908(88)90015-9.CrossRefPubMed
6.
Zurück zum Zitat Matsui T, M Sekiguchi, Hashimoto A, Tomita U, Nishikawa T, Wada K: Functional comparison of D-serine and glycine in rodents: the effect on cloned NMDA receptors and the extracellular concentration. J. Neurochem. 1995, 65: 454-458.CrossRefPubMed Matsui T, M Sekiguchi, Hashimoto A, Tomita U, Nishikawa T, Wada K: Functional comparison of D-serine and glycine in rodents: the effect on cloned NMDA receptors and the extracellular concentration. J. Neurochem. 1995, 65: 454-458.CrossRefPubMed
7.
Zurück zum Zitat Priestley T, Laughton P, Myers J, Le Bourdelles B, Kerby J, Whiting PJ: Pharmacological properties of recombinant human N-methyl-D-aspartate receptors comprising NR1a/NR2A and NR1a/NR2B subunit assemblies expressed in permanently transfected mouse fibroblast cells. Mol. Pharmacol. 1995, 48: 841-848.PubMed Priestley T, Laughton P, Myers J, Le Bourdelles B, Kerby J, Whiting PJ: Pharmacological properties of recombinant human N-methyl-D-aspartate receptors comprising NR1a/NR2A and NR1a/NR2B subunit assemblies expressed in permanently transfected mouse fibroblast cells. Mol. Pharmacol. 1995, 48: 841-848.PubMed
8.
Zurück zum Zitat Mothet JP, Parent AT, Wolosker H, Brady R. O., Jr., Linden DJ, Ferris CD, Rogawski MA, Snyder SH: D-serine is an endogenous ligand for the glycine site of the N-methyl-D- aspartate receptor. Proc Natl Acad Sci U S A. 2000, 97: 4926-4931. 10.1073/pnas.97.9.4926.PubMedCentralCrossRefPubMed Mothet JP, Parent AT, Wolosker H, Brady R. O., Jr., Linden DJ, Ferris CD, Rogawski MA, Snyder SH: D-serine is an endogenous ligand for the glycine site of the N-methyl-D- aspartate receptor. Proc Natl Acad Sci U S A. 2000, 97: 4926-4931. 10.1073/pnas.97.9.4926.PubMedCentralCrossRefPubMed
9.
Zurück zum Zitat Wood PL, Emmett MR, Rao TS, Mick S, Cler J, Iyengar S: In vivo modulation of the N-methyl-D-aspartate receptor complex by D- serine: potentiation of ongoing neuronal activity as evidenced by increased cerebellar cyclic GMP. J Neurochem. 1989, 53: 979-981.CrossRefPubMed Wood PL, Emmett MR, Rao TS, Mick S, Cler J, Iyengar S: In vivo modulation of the N-methyl-D-aspartate receptor complex by D- serine: potentiation of ongoing neuronal activity as evidenced by increased cerebellar cyclic GMP. J Neurochem. 1989, 53: 979-981.CrossRefPubMed
10.
Zurück zum Zitat Wroblewski JT, Fadda E, Mazzetta J, Lazarewicz JW, Costa E: Glycine and D-serine act as positive modulators of signal transduction at N-methyl-D-aspartate sensitive glutamate receptors in cultured cerebellar granule cells. Neuropharmacology. 1989, 28: 447-452. 10.1016/0028-3908(89)90077-4.CrossRefPubMed Wroblewski JT, Fadda E, Mazzetta J, Lazarewicz JW, Costa E: Glycine and D-serine act as positive modulators of signal transduction at N-methyl-D-aspartate sensitive glutamate receptors in cultured cerebellar granule cells. Neuropharmacology. 1989, 28: 447-452. 10.1016/0028-3908(89)90077-4.CrossRefPubMed
11.
Zurück zum Zitat Wolosker H, Blackshaw S, Snyder SH: Serine racemase: a glial enzyme synthesizing D-serine to regulate glutamate-N-methyl-D-aspartate neurotransmission. Proc. Natl. Acad. Sci. U.S.A. 1999, 96: 13409-13414. 10.1073/pnas.96.23.13409.PubMedCentralCrossRefPubMed Wolosker H, Blackshaw S, Snyder SH: Serine racemase: a glial enzyme synthesizing D-serine to regulate glutamate-N-methyl-D-aspartate neurotransmission. Proc. Natl. Acad. Sci. U.S.A. 1999, 96: 13409-13414. 10.1073/pnas.96.23.13409.PubMedCentralCrossRefPubMed
12.
Zurück zum Zitat Barger SW, Basile AS: Activation of microglia by secreted amyloid precursor protein evokes release of glutamate by cystine exchange and attenuates synaptic function. J. Neurochem. 2001, 76: 846-854. 10.1046/j.1471-4159.2001.00075.x.CrossRefPubMed Barger SW, Basile AS: Activation of microglia by secreted amyloid precursor protein evokes release of glutamate by cystine exchange and attenuates synaptic function. J. Neurochem. 2001, 76: 846-854. 10.1046/j.1471-4159.2001.00075.x.CrossRefPubMed
13.
Zurück zum Zitat Meda L, Cassatella MA, Szendrei GI, Otvos L., Jr., Baron P, Villalba M, Ferrari D, Rossi F: Activation of microglial cells by β-amyloid protein and interferon-γ. Nature. 1995, 374: 647-650. 10.1038/374647a0.CrossRefPubMed Meda L, Cassatella MA, Szendrei GI, Otvos L., Jr., Baron P, Villalba M, Ferrari D, Rossi F: Activation of microglial cells by β-amyloid protein and interferon-γ. Nature. 1995, 374: 647-650. 10.1038/374647a0.CrossRefPubMed
14.
Zurück zum Zitat Giulian D, Haverkamp LJ, Yu JH, Karshin W, Tom D, Li J, Kirkpatrick J, Kuo LM, Roher AE: Specific domains of beta-amyloid from Alzheimer plaque elicit neuron killing in human microglia. J. Neurosci. 1996, 16: 6021-6037.PubMed Giulian D, Haverkamp LJ, Yu JH, Karshin W, Tom D, Li J, Kirkpatrick J, Kuo LM, Roher AE: Specific domains of beta-amyloid from Alzheimer plaque elicit neuron killing in human microglia. J. Neurosci. 1996, 16: 6021-6037.PubMed
15.
Zurück zum Zitat McDonald DR, Brunden KR, Landreth GE: Amyloid fibrils activate tyrosine kinase-dependent signaling and superoxide production in microglia. J Neurosci. 1997, 17: 2284-2294.PubMed McDonald DR, Brunden KR, Landreth GE: Amyloid fibrils activate tyrosine kinase-dependent signaling and superoxide production in microglia. J Neurosci. 1997, 17: 2284-2294.PubMed
16.
Zurück zum Zitat Hashimoto A, Nishikawa T, Oka T, Takahashi K, Hayashi T: Determination of free amino acid enantiomers in rat brain and serum by high-performance liquid chromatography after derivatization with N-tert-butyloxycarbonyl-L-cysteine and o-phthaldialdehyde. J Chromatogr. 1992, 582: 41-48. 10.1016/0378-4347(92)80300-F.CrossRefPubMed Hashimoto A, Nishikawa T, Oka T, Takahashi K, Hayashi T: Determination of free amino acid enantiomers in rat brain and serum by high-performance liquid chromatography after derivatization with N-tert-butyloxycarbonyl-L-cysteine and o-phthaldialdehyde. J Chromatogr. 1992, 582: 41-48. 10.1016/0378-4347(92)80300-F.CrossRefPubMed
17.
Zurück zum Zitat Li Y, Liu L, Kang J, Sheng JG, Barger SW, Mrak RE, Griffin WST: Neuronal-glial interactions mediated by interleukin-1 enhance neuronal acetylcholinesterase activity and mRNA expression. J. Neurosci. 2000, 20: 149-155.PubMed Li Y, Liu L, Kang J, Sheng JG, Barger SW, Mrak RE, Griffin WST: Neuronal-glial interactions mediated by interleukin-1 enhance neuronal acetylcholinesterase activity and mRNA expression. J. Neurosci. 2000, 20: 149-155.PubMed
18.
Zurück zum Zitat Barger SW, Harmon AD: Microglial activation by secreted Alzheimer amyloid precursor protein and modulation by apolipoprotein E. Nature. 1997, 388: 878-881. 10.1038/42257.CrossRefPubMed Barger SW, Harmon AD: Microglial activation by secreted Alzheimer amyloid precursor protein and modulation by apolipoprotein E. Nature. 1997, 388: 878-881. 10.1038/42257.CrossRefPubMed
19.
Zurück zum Zitat Mao X, Barger SW: Neuroprotection by dehydroepiandrosterone-sulfate: involvement of an NFκB-like factor. NeuroReport. 1998, 9: 759-763.CrossRefPubMed Mao X, Barger SW: Neuroprotection by dehydroepiandrosterone-sulfate: involvement of an NFκB-like factor. NeuroReport. 1998, 9: 759-763.CrossRefPubMed
20.
Zurück zum Zitat Cook SP, Galve-Roperh I, Martinez Del Pozo A, Rodriguez-Crespo I: Direct calcium binding results in activation of brain Serine Racemase. J Biol Chem. 2002, 20: 27782-27792. 10.1074/jbc.M111814200.CrossRef Cook SP, Galve-Roperh I, Martinez Del Pozo A, Rodriguez-Crespo I: Direct calcium binding results in activation of brain Serine Racemase. J Biol Chem. 2002, 20: 27782-27792. 10.1074/jbc.M111814200.CrossRef
21.
Zurück zum Zitat Lo EH, Bosque-Hamilton P, Meng W: Inhibition of poly(ADP-ribose) polymerase: reduction of ischemic injury and attenuation of N-methyl-D-aspartate-induced neurotransmitter dysregulation. Stroke. 1998, 29: 830-836.CrossRefPubMed Lo EH, Bosque-Hamilton P, Meng W: Inhibition of poly(ADP-ribose) polymerase: reduction of ischemic injury and attenuation of N-methyl-D-aspartate-induced neurotransmitter dysregulation. Stroke. 1998, 29: 830-836.CrossRefPubMed
22.
Zurück zum Zitat Thomson AM, Walker VE, Flynn DM: Glycine enhances NMDA-receptor mediated synaptic potentials in neocortical slices. Nature. 1989, 338: 422-424. 10.1038/338422a0.CrossRefPubMed Thomson AM, Walker VE, Flynn DM: Glycine enhances NMDA-receptor mediated synaptic potentials in neocortical slices. Nature. 1989, 338: 422-424. 10.1038/338422a0.CrossRefPubMed
23.
Zurück zum Zitat Rao TS, Cler JA, Compton RP, Emmett MR, Mick S, Sun ET, Iyengar S, Wood PL: Neuropharmacological characterization of 1-aminocyclopropane-1- carboxylate and 1-aminocyclobutane-1-carboxylate, ligands of the N- methyl-D-aspartate-associated glycine receptor. Neuropharmacology. 1990, 29: 305-309. 10.1016/0028-3908(90)90018-M.CrossRefPubMed Rao TS, Cler JA, Compton RP, Emmett MR, Mick S, Sun ET, Iyengar S, Wood PL: Neuropharmacological characterization of 1-aminocyclopropane-1- carboxylate and 1-aminocyclobutane-1-carboxylate, ligands of the N- methyl-D-aspartate-associated glycine receptor. Neuropharmacology. 1990, 29: 305-309. 10.1016/0028-3908(90)90018-M.CrossRefPubMed
24.
Zurück zum Zitat Singh L, Oles RJ, Tricklebank MD: Modulation of seizure susceptibility in the mouse by the strychnine- insensitive glycine recognition site of the NMDA receptor/ion channel complex. Br J Pharmacol. 1990, 99: 285-288.PubMedCentralCrossRefPubMed Singh L, Oles RJ, Tricklebank MD: Modulation of seizure susceptibility in the mouse by the strychnine- insensitive glycine recognition site of the NMDA receptor/ion channel complex. Br J Pharmacol. 1990, 99: 285-288.PubMedCentralCrossRefPubMed
25.
Zurück zum Zitat Schell MJ, Molliver ME, Snyder SH: D-serine, an endogenous synaptic modulator: localization to astrocytes and glutamate-stimulated release. Proc Natl Acad Sci U S A. 1995, 92: 3948-3952.PubMedCentralCrossRefPubMed Schell MJ, Molliver ME, Snyder SH: D-serine, an endogenous synaptic modulator: localization to astrocytes and glutamate-stimulated release. Proc Natl Acad Sci U S A. 1995, 92: 3948-3952.PubMedCentralCrossRefPubMed
26.
Zurück zum Zitat Baranano DE, Ferris CD, Snyder SH: Atypical neural messengers. Trends Neurosci. 2001, 24: 99-106. 10.1016/S0166-2236(00)01716-1.CrossRefPubMed Baranano DE, Ferris CD, Snyder SH: Atypical neural messengers. Trends Neurosci. 2001, 24: 99-106. 10.1016/S0166-2236(00)01716-1.CrossRefPubMed
27.
Zurück zum Zitat Jensen AM, Chiu SY: Fluorescence measurement of changes in intracellular calcium induced by excitatory amino acids in cultured cortical astrocytes. J Neurosci. 1990, 10: 1165-1175.PubMed Jensen AM, Chiu SY: Fluorescence measurement of changes in intracellular calcium induced by excitatory amino acids in cultured cortical astrocytes. J Neurosci. 1990, 10: 1165-1175.PubMed
28.
Zurück zum Zitat Korotzer AR, Whittemore ER, Cotman CW: Differential regulation by β-amyloid peptides of intracellular free Ca2+ concentration in cultured rat microglia. Eur. J. Pharmacol. 1995, 288: 125-130. 10.1016/0922-4106(95)90006-3.CrossRefPubMed Korotzer AR, Whittemore ER, Cotman CW: Differential regulation by β-amyloid peptides of intracellular free Ca2+ concentration in cultured rat microglia. Eur. J. Pharmacol. 1995, 288: 125-130. 10.1016/0922-4106(95)90006-3.CrossRefPubMed
29.
Zurück zum Zitat Bader MF, Taupenot L, Ulrich G, Aunis D, Ciesielski-Treska J: Bacterial endotoxin induces [Ca2+]i transients and changes the organization of actin in microglia. Glia. 1994, 11: 336-344.CrossRefPubMed Bader MF, Taupenot L, Ulrich G, Aunis D, Ciesielski-Treska J: Bacterial endotoxin induces [Ca2+]i transients and changes the organization of actin in microglia. Glia. 1994, 11: 336-344.CrossRefPubMed
30.
Zurück zum Zitat Rothstein JD, Dykes-Hoberg M, Pardo CA, Bristol LA, Jin L, Kuncl RW, Kanai Y, Hediger MA, Wang Y, Schielke JP, Welty DF: Knockout of glutamate transporters reveals a major role for astroglial transport in excitotoxicity and clearance of glutamate. Neuron. 1996, 16: 675-686. 10.1016/S0896-6273(00)80086-0.CrossRefPubMed Rothstein JD, Dykes-Hoberg M, Pardo CA, Bristol LA, Jin L, Kuncl RW, Kanai Y, Hediger MA, Wang Y, Schielke JP, Welty DF: Knockout of glutamate transporters reveals a major role for astroglial transport in excitotoxicity and clearance of glutamate. Neuron. 1996, 16: 675-686. 10.1016/S0896-6273(00)80086-0.CrossRefPubMed
31.
Zurück zum Zitat Javitt DC, Balla A, Sershen H: A novel alanine-insensitive D-serine transporter in rat brain synaptosomal membranes. Brain Res. 2002, 941: 146-149. 10.1016/S0006-8993(02)02557-X.CrossRefPubMed Javitt DC, Balla A, Sershen H: A novel alanine-insensitive D-serine transporter in rat brain synaptosomal membranes. Brain Res. 2002, 941: 146-149. 10.1016/S0006-8993(02)02557-X.CrossRefPubMed
32.
Zurück zum Zitat Yasuda E, Ma N, Semba R: Immunohistochemical evidences for localization and production of D- serine in some neurons in the rat brain. Neurosci Lett. 2001, 299: 162-164. 10.1016/S0304-3940(01)01502-6.CrossRefPubMed Yasuda E, Ma N, Semba R: Immunohistochemical evidences for localization and production of D- serine in some neurons in the rat brain. Neurosci Lett. 2001, 299: 162-164. 10.1016/S0304-3940(01)01502-6.CrossRefPubMed
33.
Zurück zum Zitat Piani D, Spranger M, Frei K, Schaffner A, Fontana A: Macrophage-induced cytotoxicity of N-methyl-D-aspartate receptor positive neurons involves excitatory amino acids rather than reactive oxygen intermediates and cytokines. Eur. J. Immunol. 1992, 22: 2429-2436.CrossRefPubMed Piani D, Spranger M, Frei K, Schaffner A, Fontana A: Macrophage-induced cytotoxicity of N-methyl-D-aspartate receptor positive neurons involves excitatory amino acids rather than reactive oxygen intermediates and cytokines. Eur. J. Immunol. 1992, 22: 2429-2436.CrossRefPubMed
34.
Zurück zum Zitat Giulian D, Haverkamp LJ, Li J, Karshin WL, Yu J, Tom D, Li X, Kirkpatrick JB: Senile plaques stimulate microglia to release a neurotoxin found in Alzheimer brain. Neurochem. Int. 1995, 27: 119-137. 10.1016/0197-0186(95)00067-I.CrossRefPubMed Giulian D, Haverkamp LJ, Li J, Karshin WL, Yu J, Tom D, Li X, Kirkpatrick JB: Senile plaques stimulate microglia to release a neurotoxin found in Alzheimer brain. Neurochem. Int. 1995, 27: 119-137. 10.1016/0197-0186(95)00067-I.CrossRefPubMed
35.
Zurück zum Zitat Willard LB, Hauss-Wegrzyniak B, Danysz W, Wenk GL: The cytotoxicity of chronic neuroinflammation upon basal forebrain cholinergic neurons of rats can be attenuated by glutamatergic antagonism or cyclooxygenase-2 inhibition. Exp Brain Res. 2000, 134: 58-65. 10.1007/s002210000446.CrossRefPubMed Willard LB, Hauss-Wegrzyniak B, Danysz W, Wenk GL: The cytotoxicity of chronic neuroinflammation upon basal forebrain cholinergic neurons of rats can be attenuated by glutamatergic antagonism or cyclooxygenase-2 inhibition. Exp Brain Res. 2000, 134: 58-65. 10.1007/s002210000446.CrossRefPubMed
36.
Zurück zum Zitat Xie Z, Wei M, Morgan TE, Fabrizio P, Han D, Finch CE, Longo VD: Peroxynitrite mediates neurotoxicity of amyloid β-peptide1-42- and lipopolysaccharide-activated microglia. J. Neurosci. 2002, 22: 3484-3492.PubMed Xie Z, Wei M, Morgan TE, Fabrizio P, Han D, Finch CE, Longo VD: Peroxynitrite mediates neurotoxicity of amyloid β-peptide1-42- and lipopolysaccharide-activated microglia. J. Neurosci. 2002, 22: 3484-3492.PubMed
37.
Zurück zum Zitat Dawson VL, Dawson TM, London ED, Bredt DS, Snyder SH: Nitric oxide mediates glutamate neurotoxicity in primary cortical cultures. Proc. Natl. Acad. Sci. (USA). 1991, 88: 6368-6371.CrossRef Dawson VL, Dawson TM, London ED, Bredt DS, Snyder SH: Nitric oxide mediates glutamate neurotoxicity in primary cortical cultures. Proc. Natl. Acad. Sci. (USA). 1991, 88: 6368-6371.CrossRef
38.
Zurück zum Zitat Li Y, Liu L, Barger SW, Griffin WS: Interleukin-1 mediates pathological effects of microglia on tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway. J Neurosci. 2003, 23: 1605-1611.PubMedCentralPubMed Li Y, Liu L, Barger SW, Griffin WS: Interleukin-1 mediates pathological effects of microglia on tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway. J Neurosci. 2003, 23: 1605-1611.PubMedCentralPubMed
39.
Zurück zum Zitat Klegeris A, McGeer PL: β-amyloid protein enhances macrophage production of oxygen free radicals and glutamate. J. Neurosci. Res. 1997, 49: 229-235. 10.1002/(SICI)1097-4547(19970715)49:2<229::AID-JNR11>3.0.CO;2-W.CrossRefPubMed Klegeris A, McGeer PL: β-amyloid protein enhances macrophage production of oxygen free radicals and glutamate. J. Neurosci. Res. 1997, 49: 229-235. 10.1002/(SICI)1097-4547(19970715)49:2<229::AID-JNR11>3.0.CO;2-W.CrossRefPubMed
40.
Zurück zum Zitat Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM, Cooper NR, Eikelenboom P, Emmerling M, Fiebich BL, Finch CE, Frautschy S, Griffin WS, Hampel H, Hull M, Landreth G, Lue L, Mrak R, Mackenzie IR, McGeer PL, O'Banion MK, Pachter J, Pasinetti G, Plata-Salaman C, Rogers J, Rydel R, Shen Y, Streit W, Strohmeyer R, Tooyoma I, Van Muiswinkel FL, Veerhuis R, Walker D, Webster S, Wegrzyniak B, Wenk G, Wyss-Coray T: Inflammation and Alzheimer's disease. Neurobiol Aging. 2000, 21: 383-421. 10.1016/S0197-4580(00)00124-X.PubMedCentralCrossRefPubMed Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM, Cooper NR, Eikelenboom P, Emmerling M, Fiebich BL, Finch CE, Frautschy S, Griffin WS, Hampel H, Hull M, Landreth G, Lue L, Mrak R, Mackenzie IR, McGeer PL, O'Banion MK, Pachter J, Pasinetti G, Plata-Salaman C, Rogers J, Rydel R, Shen Y, Streit W, Strohmeyer R, Tooyoma I, Van Muiswinkel FL, Veerhuis R, Walker D, Webster S, Wegrzyniak B, Wenk G, Wyss-Coray T: Inflammation and Alzheimer's disease. Neurobiol Aging. 2000, 21: 383-421. 10.1016/S0197-4580(00)00124-X.PubMedCentralCrossRefPubMed
41.
Zurück zum Zitat Chouinard ML, Gaitan D, Wood PL: Presence of the N-methyl-D-aspartate-associated glycine receptor agonist, D-serine, in human temporal cortex: comparison of normal, Parkinson, and Alzheimer tissues. J Neurochem. 1993, 61: 1561-1564.CrossRefPubMed Chouinard ML, Gaitan D, Wood PL: Presence of the N-methyl-D-aspartate-associated glycine receptor agonist, D-serine, in human temporal cortex: comparison of normal, Parkinson, and Alzheimer tissues. J Neurochem. 1993, 61: 1561-1564.CrossRefPubMed
42.
Zurück zum Zitat Kumashiro S, Hashimoto A, Nishikawa T: Free D-serine in post-mortem brains and spinal cords of individuals with and without neuropsychiatric diseases. Brain Res. 1995, 681: 117-125. 10.1016/0006-8993(95)00307-C.CrossRefPubMed Kumashiro S, Hashimoto A, Nishikawa T: Free D-serine in post-mortem brains and spinal cords of individuals with and without neuropsychiatric diseases. Brain Res. 1995, 681: 117-125. 10.1016/0006-8993(95)00307-C.CrossRefPubMed
43.
Zurück zum Zitat Molina JA, Jimenez-Jimenez FJ, Vargas C, Gomez P, de Bustos F, Orti-Pareja M, Tallon-Barranco A, Benito-Leon J, Arenas J, Enriquez-de-Salamanca R: Cerebrospinal fluid levels of non-neurotransmitter amino acids in patients with Alzheimer's disease. J Neural Transm. 1998, 105: 279-286. 10.1007/s007020050057.CrossRefPubMed Molina JA, Jimenez-Jimenez FJ, Vargas C, Gomez P, de Bustos F, Orti-Pareja M, Tallon-Barranco A, Benito-Leon J, Arenas J, Enriquez-de-Salamanca R: Cerebrospinal fluid levels of non-neurotransmitter amino acids in patients with Alzheimer's disease. J Neural Transm. 1998, 105: 279-286. 10.1007/s007020050057.CrossRefPubMed
44.
Zurück zum Zitat Yamada T, Kondo A, Takamatsu J, Tateishi J, Goto I: Apolipoprotein E mRNA in the brains of patients with Alzheimer's disease. J Neurol Sci. 1995, 129: 56-61. 10.1016/0022-510X(94)00249-N.CrossRefPubMed Yamada T, Kondo A, Takamatsu J, Tateishi J, Goto I: Apolipoprotein E mRNA in the brains of patients with Alzheimer's disease. J Neurol Sci. 1995, 129: 56-61. 10.1016/0022-510X(94)00249-N.CrossRefPubMed
45.
Zurück zum Zitat Delacourte A: General and dramatic glial reaction in Alzheimer brains. Neurology. 1990, 40: 33-37.CrossRefPubMed Delacourte A: General and dramatic glial reaction in Alzheimer brains. Neurology. 1990, 40: 33-37.CrossRefPubMed
46.
Zurück zum Zitat Loring JF, Wen X, Lee JM, Seilhamer J, Somogyi R: A gene expression profile of Alzheimer's disease. DNA Cell Biol. 2001, 20: 683-695. 10.1089/10445490152717541.CrossRefPubMed Loring JF, Wen X, Lee JM, Seilhamer J, Somogyi R: A gene expression profile of Alzheimer's disease. DNA Cell Biol. 2001, 20: 683-695. 10.1089/10445490152717541.CrossRefPubMed
Metadaten
Titel
Induction of serine racemase expression and D-serine release from microglia by amyloid β-peptide
verfasst von
Sheng-Zhou Wu
Angela M Bodles
Mandy M Porter
W Sue T Griffin
Anthony S Basile
Steven W Barger
Publikationsdatum
01.12.2004
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2004
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/1742-2094-1-2

Weitere Artikel der Ausgabe 1/2004

Journal of Neuroinflammation 1/2004 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.