Elsevier

Neurobiology of Aging

Volume 36, Issue 5, May 2015, Pages 1890-1902
Neurobiology of Aging

Regular article
d-Aspartate oxidase influences glutamatergic system homeostasis in mammalian brain

https://doi.org/10.1016/j.neurobiolaging.2015.02.003Get rights and content

Abstract

We have investigated the relevance of d-aspartate oxidase, the only enzyme known to selectively degrade d-aspartate (d-Asp), in modulating glutamatergic system homeostasis. Interestingly, the lack of the Ddo gene, by raising d-Asp content, induces a substantial increase in extracellular glutamate (Glu) levels in Ddo-mutant brains. Consistent with an exaggerated and persistent N-methyl-d-aspartate receptor (NMDAR) stimulation, we documented in Ddo knockouts severe age-dependent structural and functional alterations mirrored by expression of active caspases 3 and 7 along with appearance of dystrophic microglia and reactive astrocytes. In addition, prolonged elevation of d-Asp triggered in mutants alterations of NMDAR-dependent synaptic plasticity associated to reduction of hippocampal GluN1 and GluN2B subunits selectively located at synaptic sites and to increase in the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-to-N-methyl-d-aspartate ratio. These effects, all of which converged on a progressive hyporesponsiveness at NMDAR sites, functionally resulted in a greater vulnerability to phencyclidine-induced prepulse inhibition deficits in mutants. In conclusion, our results indicate that d-aspartate oxidase, by strictly regulating d-Asp levels, impacts on the homeostasis of glutamatergic system, thus preventing accelerated neurodegenerative processes.

Introduction

Of the pool of endogenous amino acids, only serine and aspartate substantially occur in free d-form in mammalian tissues (Billard, 2012, Errico et al., 2012, Mothet and Snyder, 2012, Ota et al., 2012, Wolosker and Mori, 2012). d-Serine (d-Ser) is mainly localized in forebrain structures of the central nervous system throughout embryonic development and the postnatal phase. Compelling evidence demonstrates that d-Ser acts as an endogenous co-agonist at N-methyl-d-aspartate receptors (NMDARs) (Billard, 2012, Martineau et al., 2006, Snyder and Kim, 2000), and facilitation of d-Ser transmission could represent a therapeutic option in psychiatric disorders, including schizophrenia (Coyle et al., 2002). On the other hand, much less is known about the role of free d-aspartate (d-Asp) in the mammalian brain. d-Asp binds to and activates GluN2 subunits of NMDARs (Errico et al., 2011b). In line with its pharmacological features, nonphysiological elevated levels of d-Asp in knockout mice for the d-aspartate oxidase (Ddo) gene are associated with changes in NMDAR-dependent responses, including synaptic plasticity and spatial memory (Errico et al., 2011c, Errico et al., 2008a, Errico et al., 2008b). Furthermore, we have previously reported that 1-month oral administration of d-Asp in mice enhances NMDAR-mediated currents, basal cerebral blood volume in frontohippocampal areas, and increases dendritic length and spine density in the prefrontal cortex (PFC) and hippocampus (Errico et al., 2014). In agreement with mouse studies, we also found that genetic variation predicting reduced DDO expression in the postmortem human PFC mapped to greater prefrontal gray matter and activity during working memory (Errico et al., 2014). We also have recently documented a substantial reduction of d-Asp and NMDA in the postmortem PFC and striatum of schizophrenic patients compared with healthy controls, thus suggesting a potential involvement of this molecule in schizophrenia (Errico et al., 2013). Remarkably, d-Asp displays a peculiar temporal occurrence in the brain, that is, levels are high between embryonic and early postnatal stages and rapidly decrease to trace levels after birth (Dunlop et al., 1986, Hashimoto et al., 1993, Hashimoto et al., 1995, Lee et al., 1999, Neidle and Dunlop, 1990, Sakai et al., 1998, Wolosker et al., 2000) because of the concomitant expression of DDO, the only enzyme known to degrade bicarboxylic d-amino acids (D'Aniello et al., 1993, Still et al., 1949, Van Veldhoven et al., 1991). Given the pharmacological ability of d-Asp to act as a direct NMDAR agonist and in the light of the strict control exerted by DDO on postnatal brain levels of d-Asp, we investigated the biological significance of this enzyme in regulating glutamatergic system homeostasis in the hippocampus and PFC of mice with targeted deletion of Ddo gene.

Section snippets

Animals

Knockout mice for the Ddo gene (Ddo-KO) were generated and genotyped by polymerase chain reaction as described previously (Errico et al., 2006). Animals were housed in a maximum of 5 per cage, at a constant temperature (22 ± 1 °C) and maintained on a 12-hour light-dark cycle, with food and water ad libitum. All research involving animals was carried out in accordance with the European directive 86/609/EEC governing animal welfare and protection, which is acknowledged by the Italian Legislative

d-Asp modulates Glu release in mouse brain and in synaptosomal preparation

We previously reported that d-Asp levels in the cortex and hippocampus homogenates were 15- to 20-fold higher in Ddo-KO than in Ddo-WT littermates (Errico et al., 2012). Here, we investigated the consequences of elevated d-Asp content on basal extracellular Glu levels in the brain of Ddo-KO mice. In vivo microdialysis performed in the hippocampus of freely moving mice revealed that Glu levels were significantly higher in 6-month-old mutants than in age-matched controls [F(1,12) = 14.87; p <

Discussion

Here, we document that DDO plays a role in the modulation of glutamatergic system homeostasis in the mammalian brain. Indeed, deregulation of d-Asp brain levels, through genetic deletion of the Ddo gene, triggers a consistent change in extracellular Glu levels. The ability of d-Asp to enhance Glu release is confirmed both after its acute injection in freely moving mice and by its direct application in the synaptosome preparation. Synaptosomes in superfusion are an ideal preparation for studying

Disclosure statement

The authors have no conflicts of interest to disclose.

Acknowledgements

The authors thank F. Napolitano, A. Di Maio, and V. Lucignano for their excellent technical support and Jean Ann Gilder (Scientific Communication srl, Naples, Italy) for editing the text. Alessandro Usiello represents the Mariano Scippacercola Foundation. Alessandro Usiello was supported by NARSAD Independent Investigator Grant from the Brain and Behavior Research Foundation (Grant nr: 20353). Francesco Errico was supported by grants from the Italian Ministero dell’Istruzione, dell’Università e

References (53)

  • O.S. Mabrouk et al.

    Endogenous nociceptin/orphanin FQ (N/OFQ) contributes to haloperidol-induced changes of nigral amino acid transmission and parkinsonism: a combined microdialysis and behavioral study in naive and nociceptin/orphanin FQ receptor knockout mice

    Neuroscience

    (2010)
  • M. Martineau et al.

    D-serine signalling in the brain: friend and foe

    Trends Neurosci.

    (2006)
  • M. Mellone et al.

    Modulation of NMDA receptor at the synapse: promising therapeutic interventions in disorders of the nervous system

    Eur. J. Pharmacol.

    (2013)
  • A. Neidle et al.

    Developmental changes in free D-aspartic acid in the chicken embryo and in the neonatal rat

    Life Sci.

    (1990)
  • E.S. Rosenzweig et al.

    Impact of aging on hippocampal function: plasticity, network dynamics, and cognition

    Prog. Neurobiol.

    (2003)
  • K. Sakai et al.

    Localization of D-aspartic acid in elongate spermatids in rat testis

    Arch. Biochem. Biophys.

    (1998)
  • A. Sanz-Clemente et al.

    Activated CaMKII couples GluN2B and casein kinase 2 to control synaptic NMDA receptors

    Cell Rep.

    (2013)
  • J.L. Still et al.

    Studies on the cyclophorase system; D-aspartic oxidase

    J. Biol. Chem.

    (1949)
  • P.P. Van Veldhoven et al.

    D-aspartate oxidase, a peroxisomal enzyme in liver of rat and man

    Biochim. Biophys. Acta

    (1991)
  • S. Vesce et al.

    Glutamate release from astrocytes in physiological conditions and in neurodegenerative disorders characterized by neuroinflammation

    Int. Rev. Neurobiol.

    (2007)
  • H. Wolosker et al.

    D-aspartate disposition in neuronal and endocrine tissues: ontogeny, biosynthesis and release

    Neuroscience

    (2000)
  • J. Zhang et al.

    Microglial CR3 activation triggers long-term synaptic depression in the hippocampus via NADPH oxidase

    Neuron

    (2014)
  • J.M. Billard

    D-amino acids in brain neurotransmission and synaptic plasticity

    Amino Acids

    (2012)
  • J.T. Coyle et al.

    Ionotropic glutamate receptors as therapeutic targets in schizophrenia

    Curr. Drug Targets CNS Neurol. Disord.

    (2002)
  • L. Cristino et al.

    Obesity-driven synaptic remodeling affects endocannabinoid control of orexinergic neurons

    Proc. Natl. Acad. Sci. U. S. A.

    (2013)
  • R. Dantzer et al.

    Is there a role for glutamate-mediated excitotoxicity in inflammation-induced depression?

    J. Neural Transm.

    (2014)
  • Cited by (42)

    • AQP4 mitigates chronic neuropathic pain-induced cognitive impairment in mice

      2023, Behavioural Brain Research
      Citation Excerpt :

      However, further efforts are required to elucidate the molecular mechanisms underlying the alleviation of CNP-induced cognitive impairment by AQP4 in astrocytes. Meanwhile, recent papers showed that chronic pain as well as neurodegenerative diseases can be involved in the dystrophic phenotype of microglia in mice [46]. Punzo[47] found in special gene knockouts severe age-dependent cognitive alterations mirrored by expression of active apoptosis cells with appearance of dystrophic microglia and reactive astrocytes.

    • Effects of datumetine on hippocampal NMDAR activity

      2021, Toxicology Reports
      Citation Excerpt :

      Preclinical studies have reported that NMDAR antagonist alleviates the symptoms [66] alluding to the fact that hyperactivation of NMDAR is part of the aetiology of these disorders. This result is different from the report of Cristino et al. [67], who showed that indirectly increasing NMDAR activity by knocking out gene coding for D-aspartate oxidase reduces GluN1 expression in the hippocampus with increased GluN1/AMPAR ratio. This difference may be due to the overall altered glutamate homeostasis observed in their study.

    • Dysfunctional D-aspartate metabolism in BTBR mouse model of idiopathic autism

      2020, Biochimica et Biophysica Acta - Proteins and Proteomics
      Citation Excerpt :

      In line with well-established detrimental effects associated to overstimulation of NMDARs [79], it has been reported that a long-term increase in D-Asp levels results in the precocious decay of basal glutamatergic transmission, synaptic plasticity, and hippocampal reference memory in mutants [80–82]. Such dysfunctions are mirrored by the loss of excitatory glutamatergic synapses and reduction in synaptic GluN1 and GluN2B subunits [82]. In addition, severe age-dependent neuroinflammation and cell death occur within PFC and hippocampus of Ddo knockout animals [81].

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    2

    Senior authors.

    View full text