Gliotransmission at central glutamatergic synapses: d-serine on stage

https://doi.org/10.1016/j.jphysparis.2005.12.011Get rights and content

Abstract

Long ignored and only considered as housekeeping cells for neurons, astroglial cells in the last decade have gained increasing attention as key players of higher functions in healthy brain, but also in diseases. This revolution in our way to think the active brain culminates in the concept of a tripartite synapse, which considers glial cells and notably astrocytes as an integral dynamic partner of synapses. Glia not only listens but also talks to neurons through the release of neuroactive substances. Recently much attention has been paid to the role played by the atypical amino acid d-serine in this signalling pathway. This molecule synthesized through racemization of l-serine fulfils most criteria as a gliotransmitter and as the endogenous ligand for the strychnine-insensitive glycine binding site of the NMDA receptors. d-Serine is considered to be a permissive factor for long-term changes in synaptic plasticity and neuronal migration through activation of NMDA receptors. It is also known that disturbance of NMDA receptors activity can cause cell death. Not surprisingly, then, d-serine has also been found to promote neurons death in experimental models of β-amyloid peptide-induced neuroinflammation and of ischaemia by overactivating the NMDA receptors. Finally, in a more recent past, studies have pointed to the molecular mechanisms leading to d-serine release into and removal from the synaptic cleft.

Introduction

Glutamate, the main excitatory neurotransmitter in the mammalian CNS, mediates slow and fast neurotransmission through three classes of ionotropic receptors (NMDA, AMPA and kainate) (Dingledine et al., 1999). The N-methyl d-aspartate (NMDA) subtype of glutamate receptors is member of a class of ionotropic receptor channels, organized as heteromeric assemblies composed of a spliced variant of NR1 subunit, combined with at least one of four NR2 (A–D) subunits (Dingledine et al., 1999, Kemp and McKernan, 2002). A third subunit, NR3, which is less common, can co-assemble with NR1/NR2 complexes. Based on the subunit composition, the pharmacology and the distribution of NMDA receptors (NMDARs) vary. Activation of NMDAR is complex. The ion-channel integral to the NMDAR is voltage-dependently blocked by magnesium. Depolarization removes this block, allowing the permeation through NMDARs of Ca2+ ions inside the depolarized cells (Dingledine et al., 1999, Kemp and McKernan, 2002). Thus the receptor acts as a coincidence detector, linking glutamate activation with the electrical state of the neuron. Accordingly, the magnitude or duration of NMDA-receptor-mediated Ca2+ influx has been shown to dictate the type and sign of plasticity induced (Perez-Otano and Ehlers, 2005). A useful simplified framework has been that small amounts of NMDA-receptor-mediated Ca2+ influx produce Long-Term Depression (LTD) whereas strong activation of NMDARs leads to Long-Term Potentiation (LTP). Furthermore, recent studies have shown that NMDARs serve a homeostatic role ensuring that plasticity is kept within a working range (away from saturation) and control the threshold shift for LTP and LTD induction (Perez-Otano and Ehlers, 2005). Thus, the activity of NMDARs is critical for normal brain function and NMDARs knock-down mice display severe brain impairment with defect in neuronal migration and in synapses stability (Lee et al., 2005, Mohn et al., 1999). On contrast, chronical and excessive overstimulation of NMDARs causes excitotoxicity (Shleper et al., 2005, Takano et al., 2001) driving the search for NMDA antagonists as neuroprotective agents. Too much NMDAR activity is harmful as too little. Thus the elucidation of the factors maintaining a balanced activity for these receptors is important.

Furthermore, in addition to glutamate, activation of NMDAR requires the binding of a coagonist on the NR1 subunit. Since the late 1980s, it has been assumed that glycine is the coagonist (Johnson and Ascher, 1987). Nevertheless, a number of reports in the last decade have led to the demonstration that d-serine is the major neuromodulator for the NMDAR at the glycine site (Miller, 2004).

Section snippets

Distribution of d-serine in the CNS

d-Serine is present in a significant amount in the brain of rodents and men where its levels (∼0.27 μmol/g weight) are up to a third of the total free (l + d) serine pool. Early HPLC analyses by Hashimoto et al., 1995a, Hashimoto et al., 1995b have revealed a heterogeneous distribution of d-serine throughout the brain with highest concentrations in the telencephalon and the developing cerebellum (Hashimoto et al., 1995a). At adult stage (8 weeks), the concentration of d-serine is higher in the

Metabolism of d-serine in the CNS

Although, d-serine may derive from diet, gastrointestinal bacteria, or from cleavage of metabolically stable proteins, highest levels of d-serine in the mammalian brain are generated by activity of the pyridoxal 5′-phosphate (PLP)-dependent serine racemase enzyme (SR). Studies have characterized the structure of the SR gene from mouse, rat and humans (De Miranda et al., 2000, Konno, 2003, Wolosker et al., 1999, Xia et al., 2004). The gene consists in seven exons, the first exon containing the

Function of d-serine in the mammalian brain

The close correlation between the anatomical distribution of d-serine and SR with the regional variation of the NMDAR suggests a functional relationship. The first functional evidence supporting the hypothesis that d-serine may interact with glutamatergic synaptic activity came from experiments measuring the impact of glutamatergic agonists on the release of d-serine from cultured astrocytes (Schell et al., 1995). These experiments revealed that activation of AMPA/kainate receptors induced a

Molecular mechanisms of glial d-serine uptake and release

Since the discovery of d-serine in the mammalian brain, research has identified the metabolic pathway and has given functions to this atypical neurotransmitter in the brain. On contrast, less attention has been paid to the molecular mechanisms controlling d-serine disposition in the synaptic cleft. How d-serine is released in and removed from the synaptic cleft has to be elucidated. These informations are important in order to unravel the functional consequences of d-serine-mediated

Conclusion

Since the pioneer work of Hashimoto’s and Snyder’s groups describing the distribution of d-serine in rat brain, expanding evidence have been accumulated from many groups showing that d-serine fulfils all criteria as a gliotransmitter and have brought d-serine on mainstream in Neuroscience.

Today, the model proposed by Snyder et al. (Schell et al., 1995) on the interdependency of glutamate and d-serine at glutamatergic synapses has been strengthened by accumulating data. According to the original

References (56)

  • A. Morikawa et al.

    Determination of free d-aspartic acid, d-serine and d-alanine in the brain of mutant mice lacking d-amino acid oxidase activity

    J. Chromatogr. B Biomed. Sci. Appl.

    (2001)
  • K.B. O’Brien et al.

    d-Serine uptake by isolated retinas is consistent with ASCT-mediated transport

    Neurosci. Lett.

    (2005)
  • I. Perez-Otano et al.

    Homeostatic plasticity and NMDA receptor trafficking

    Trends Neurosci.

    (2005)
  • C.S. Ribeiro et al.

    Glial transport of the neuromodulator d-serine

    Brain Res.

    (2002)
  • K. Strisovsky et al.

    Mouse brain serine racemase catalyzes specific elimination of l-serine to pyruvate

    FEBS Lett.

    (2003)
  • Y. Urai et al.

    Gene expression of d-amino acid oxidase in cultured rat astrocytes: regional and cell type specific expression

    Neurosci. Lett.

    (2002)
  • M. Xia et al.

    Characterization and localization of a human serine racemase

    Brain Res. Mol. Brain Res.

    (2004)
  • X. Xie et al.

    Lack of the alanine–serine–cysteine transporter 1 causes tremors, seizures, and early postnatal death in mice

    Brain. Res.

    (2005)
  • E. Yasuda et al.

    Immunohistochemical evidences for localization and production of d-serine in some neurons in the rat brain

    Neurosci. Lett.

    (2001)
  • A. Araque et al.

    SNARE protein-dependent glutamate release from astrocytes

    J. Neurosci.

    (2000)
  • A.J. Berger et al.

    Glycine uptake governs glycine site occupancy at NMDA receptors of excitatory synapses

    J. Neurophysiol.

    (1998)
  • P. Bezzi et al.

    Astrocytes contain a vesicular compartment that is competent for regulated exocytosis of glutamate

    Nat. Neurosci.

    (2004)
  • L. Cristiano et al.

    Peroxisome proliferator-activated receptors (PPARs) and peroxisomes in rat cortical and cerebellar astrocytes

    J. Neurocytol.

    (2001)
  • J. De Miranda et al.

    Cofactors of serine racemase that physiologically stimulate the synthesis of the N-methyl-d-aspartate (NMDA) receptor coagonist d-serine

    Proc. Natl. Acad. Sci. USA

    (2002)
  • R. Dingledine et al.

    The glutamate receptor ion channels

    Pharmacol. Rev.

    (1999)
  • A. Hashimoto et al.

    Anatomical distribution and postnatal changes in endogenous free d-aspartate and d-serine in rat brain and periphery

    Eur. J. Neurosci.

    (1995)
  • R. Jahn et al.

    Membrane fusion and exocytosis

    Annu. Rev. Biochem.

    (1999)
  • J.W. Johnson et al.

    Glycine potentiates the NMDA response in cultured mouse brain neurons

    Nature

    (1987)
  • Cited by (0)

    View full text