Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Proc Natl Acad Sci U S A ; 120(42): e2302780120, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37812701

ABSTRACT

Brain L-serine is critical for neurodevelopment and is thought to be synthesized solely from glucose. In contrast, we found that the influx of L-serine across the blood-brain barrier (BBB) is essential for brain development. We identified the endothelial Slc38a5, previously thought to be a glutamine transporter, as an L-serine transporter expressed at the BBB in early postnatal life. Young Slc38a5 knockout (KO) mice exhibit developmental alterations and a decrease in brain L-serine and D-serine, without changes in serum or liver amino acids. Slc38a5-KO brains exhibit accumulation of neurotoxic deoxysphingolipids, synaptic and mitochondrial abnormalities, and decreased neurogenesis at the dentate gyrus. Slc38a5-KO pups exhibit motor impairments that are affected by the administration of L-serine at concentrations that replenish the serine pool in the brain. Our results highlight a critical role of Slc38a5 in supplying L-serine via the BBB for proper brain development.


Subject(s)
Blood-Brain Barrier , Brain , Mice , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Biological Transport , Ion Transport , Serine/metabolism , Mice, Knockout
2.
Brain ; 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38662784

ABSTRACT

Mutations in the SLC1A4 transporter lead to neurodevelopmental impairments, spastic tetraplegia, thin corpus callosum, and microcephaly in children. SLC1A4 catalyzes obligatory amino acid exchange between neutral amino acids, but the physiopathology of SLC1A4 disease mutations and progressive microcephaly remain unclear. Here, we examined the phenotype and metabolic profile of three Slc1a4 mouse models, including a constitutive Slc1a4-KO mouse, a knock-in mouse with the major human Slc1a4 mutation (Slc1a4-K256E), and a selective knockout of Slc1a4 in brain endothelial cells (Slc1a4tie2-cre). We show that Slc1a4 is a bona fide L-serine transporter at the BBB and that acute inhibition or deletion of Slc1a4 leads to a decrease in serine influx into the brain. This results in microcephaly associated with decreased L-serine content in the brain, accumulation of atypical and cytotoxic 1-deoxysphingolipids in the brain, neurodegeneration, synaptic and mitochondrial abnormalities, and behavioral impairments. Prenatal and early postnatal oral administration of L-serine at levels that replenish the serine pool in the brain rescued the observed biochemical and behavioral changes. Administration of L-serine till the second postnatal week also normalized brain weight in Slc1a4-E256Ć¢Ā€Ā…K mice. Our observations suggest that the transport of "non-essential" amino acids from the blood through the BBB is at least as important as that of essential amino acids for brain metabolism and development. We proposed that SLC1A4 mutations cause a BBB aminoacidopathy with deficits in serine import across the BBB required for optimal brain growth and leads to a metabolic microcephaly, which may be amenable to treatment with L-serine.

3.
Glia ; 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39360557

ABSTRACT

Histidine dipeptides (HDs) are synthesized in brain oligodendrocytes by carnosine synthase (carns1), but their role is unknown. Using metabolomics and in vivo experiments with both constitutive and oligodendrocyte-selective carns1-KO mouse models, we found that HDs are critical for oligodendrocyte survival and protect against oxidative stress. Carns1-KO mouse models had lower numbers of mature oligodendrocytes, increased lipid peroxidation, and behavioral changes. Cuprizone administration, which increases reactive oxygen species in vivo, resulted in higher oligodendrocyte death, demyelination, axonal alterations, and oxidative damage in the corpus callosum of carns1-KO mice. Gliosis and oxidative damage by cuprizone were prevented by pretreatment with the antioxidant N-acetylcysteine. NADPH levels were increased threefold in the brains of carns1-KO mice as an antioxidant response to oxidative stress through acceleration of the pentose phosphate pathway (PPP). This was due to overexpression of glucose-6-phosphate dehydrogenase, the rate-limiting enzyme of the PPP. Likewise, expression of NAD kinase, the biosynthetic enzyme for NADP+, and NAMPT, which replenishes the NAD+ pool, was higher in carns1-KO mice brains than in controls. Our observations suggest that HDs cell-autonomously protect oligodendrocytes from oxidative stress, with implications for demyelinating diseases.

4.
Glia ; 70(6): 1133-1152, 2022 06.
Article in English | MEDLINE | ID: mdl-35195906

ABSTRACT

Synaptic damage is one of the most prevalent pathophysiological responses to traumatic CNS injury and underlies much of the associated cognitive dysfunction; however, it is poorly understood. The D-amino acid, D-serine, serves as the primary co-agonist at synaptic NMDA receptors (NDMARs) and is a critical mediator of NMDAR-dependent transmission and synaptic plasticity. In physiological conditions, D-serine is produced and released by neurons from the enzymatic conversion of L-serine by serine racemase (SRR). However, under inflammatory conditions, glial cells become a major source of D-serine. Here, we report that D-serine synthesized by reactive glia plays a critical role in synaptic damage after traumatic brain injury (TBI) and identify the therapeutic potential of inhibiting glial D-serine release though the transporter Slc1a4 (ASCT1). Furthermore, using cell-specific genetic strategies and pharmacology, we demonstrate that TBI-induced synaptic damage and memory impairment requires D-serine synthesis and release from both reactive astrocytes and microglia. Analysis of the murine cortex and acutely resected human TBI brain also show increased SRR and Slc1a4 levels. Together, these findings support a novel role for glial D-serine in acute pathological dysfunction following brain trauma, whereby these reactive cells provide the excess co-agonist levels necessary to initiate NMDAR-mediated synaptic damage.


Subject(s)
Brain Injuries , Serine , Amino Acid Transport System ASC/metabolism , Animals , Astrocytes/metabolism , Brain Injuries/drug therapy , Humans , Mice , Neuroglia/metabolism , Neuronal Plasticity/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/physiology
5.
Biochem J ; 478(5): 1175-1178, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33710333

ABSTRACT

Promiscuous catalysis is a common property of enzymes, particularly those using pyridoxal 5'-phosphate as a cofactor. In a recent issue of this journal, Katane et al. Biochem. J. 477, 4221-4241 demonstrate the synthesis and accumulation of d-glutamate in mammalian cells by promiscuous catalysis mediated by a pyridoxal 5'-phosphate enzyme, the serine/threonine dehydratase-like (SDHL). The mechanism of SDHL resembles that of serine racemase, which synthesizes d-serine, a well-established signaling molecule in the mammalian brain. d-Glutamate is present in body fluids and is degraded by the d-glutamate cyclase at the mitochondria. This study demonstrates a biochemical pathway for d-glutamate synthesis in mammalian cells and advances our knowledge on this little-studied d-amino acid in mammals. d-Amino acids may still surprise us by their unique roles in biochemistry, intercellular signaling, and as potential biomarkers of disease.


Subject(s)
Amino Acids , L-Serine Dehydratase , Animals , Glutamic Acid , Mammals , Pyridoxal Phosphate
6.
Proc Natl Acad Sci U S A ; 116(41): 20736-20742, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31548413

ABSTRACT

Astrocytes express the 3-phosphoglycerate dehydrogenase (Phgdh) enzyme required for the synthesis of l-serine from glucose. Astrocytic l-serine was proposed to regulate NMDAR activity by shuttling to neurons to sustain d-serine production, but this hypothesis remains untested. We now report that inhibition of astrocytic Phgdh suppressed the de novo synthesis of l-and d-serine and reduced the NMDAR synaptic potentials and long-term potentiation (LTP) at the Schaffer collaterals-CA1 synapse. Likewise, enzymatic removal of extracellular l-serine impaired LTP, supporting an l-serine shuttle mechanism between glia and neurons in generating the NMDAR coagonist d-serine. Moreover, deletion of serine racemase (SR) in glutamatergic neurons abrogated d-serine synthesis to the same extent as Phgdh inhibition, suggesting that neurons are the predominant source of the newly synthesized d-serine. We also found that the synaptic NMDAR activation in adult SR-knockout (KO) mice requires Phgdh-derived glycine, despite the sharp decline in the postnatal glycine levels as a result of the emergence of the glycine cleavage system. Unexpectedly, we also discovered that glycine regulates d-serine metabolism by a dual mechanism. The first consists of tonic inhibition of SR by intracellular glycine observed in vitro, primary cultures, and in vivo microdialysis. The second involves a transient glycine-induce d-serine release through the Asc-1 transporter, an effect abolished in Asc-1 KO mice and diminished by deleting SR in glutamatergic neurons. Our observations suggest that glycine is a multifaceted regulator of d-serine metabolism and implicate both d-serine and glycine in mediating NMDAR synaptic activation at the mature hippocampus through a Phgdh-dependent shuttle mechanism.


Subject(s)
Astrocytes/metabolism , Glycine/metabolism , Phosphoglycerate Dehydrogenase/metabolism , Racemases and Epimerases/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Serine/metabolism , Synapses/physiology , Animals , Astrocytes/cytology , Hippocampus/cytology , Hippocampus/metabolism , Long-Term Potentiation , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/metabolism , Phosphoglycerate Dehydrogenase/genetics , Receptors, N-Methyl-D-Aspartate/genetics
7.
J Neurosci ; 40(34): 6489-6502, 2020 08 19.
Article in English | MEDLINE | ID: mdl-32661027

ABSTRACT

D-serine is a physiologic coagonist of NMDA receptors (NMDARs) required for synaptic plasticity, but mechanisms that terminate D-serine signaling are unclear. In particular, the identity of unidirectional plasma membrane transporters that mediate D-serine reuptake has remained elusive. We report that D-serine and glutamine share the same neuronal transport system, consisting of the classic system A transporters Slc38a1 and Slc38a2. We show that these transporters are not saturated with glutamine in vivo and regulate the extracellular levels of D-serine and NMDAR activity. Glutamine increased the NMDAR-dependent long-term potentiation and the isolated NMDAR potentials at the Schaffer collateral-CA1 synapses, but without affecting basal neurotransmission in male mice. Glutamine did not increase the NMDAR potentials in slices from serine racemase knock-out mice, which are devoid of D-serine, indicating that the effect of glutamine is caused by outcompeting D-serine for a dual glutamine-D-serine transport system. Inhibition of the system A reduced the uptake of D-serine in synaptosomes and neuronal cultures of mice of either sex, while increasing the extracellular D-serine concentration in slices and in vivo by microdialysis. When compared with Slc38a2, the Slc38a1 transporter displayed more favorable kinetics toward the D-enantiomer. Biochemical experiments with synaptosomes from Slc38a1 knock-down mice of either sex further support its role as a D-serine reuptake system. Our study identifies the first concentrative and electrogenic transporters mediating D-serine reuptake in vivo In addition to their classical role in the glutamine-glutamate cycle, system A transporters regulate the synaptic turnover of D-serine and its effects on NMDAR synaptic plasticity.SIGNIFICANCE STATEMENT Despite the plethora of roles attributed to D-serine, the regulation of its synaptic turnover is poorly understood. We identified the system A transporters Slc38a1 and Slc38a2 as the main pathway for neuronal reuptake of D-serine. These transporters are not saturated with glutamine in vivo and provide an unexpected link between the serine shuttle pathway, responsible for regulating D-serine synaptic turnover, and the glutamine-glutamate cycle. Our observations suggest that Slc38a1 and Slc38a2 have a dual role in regulating neurotransmission. In addition to their classical role as the glutamine providers, the system A transporters regulate extracellular D-serine and therefore affect NMDAR-dependent synaptic plasticity. Higher glutamine export from astrocytes would increase extracellular D-serine, providing a feedforward mechanism to increase synaptic NMDAR activation.


Subject(s)
Amino Acid Transport System A/metabolism , Glutamine/metabolism , Neuronal Plasticity , Receptors, N-Methyl-D-Aspartate/metabolism , Serine/metabolism , Signal Transduction , Animals , Female , Hippocampus/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Synaptic Transmission
8.
Proc Natl Acad Sci U S A ; 115(38): 9628-9633, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30185558

ABSTRACT

d-serine is a physiologic coagonist of NMDA receptors, but little is known about the regulation of its synthesis and synaptic turnover. The amino acid exchangers ASCT1 (Slc1a4) and ASCT2 (Slc1a5) are candidates for regulating d-serine levels. Using ASCT1 and ASCT2 KO mice, we report that ASCT1, rather than ASCT2, is a physiologic regulator of d-serine metabolism. ASCT1 is a major d-serine uptake system in astrocytes and can also export l-serine via heteroexchange, supplying neurons with the substrate for d-serine synthesis. ASCT1-KO mice display lower levels of brain d-serine along with higher levels of l-alanine, l-threonine, and glycine. Deletion of ASCT1 was associated with neurodevelopmental alterations including lower hippocampal and striatal volumes and changes in the expression of neurodevelopmental-relevant genes. Furthermore, ASCT1-KO mice exhibited deficits in motor function, spatial learning, and affective behavior, along with changes in the relative contributions of d-serine vs. glycine in mediating NMDA receptor activity. In vivo microdialysis demonstrated lower levels of extracellular d-serine in ASCT1-KO mice, confirming altered d-serine metabolism. These alterations are reminiscent of some of the neurodevelopmental phenotypes exhibited by patients with ASCT1 mutations. ASCT1-KO mice provide a useful model for potential therapeutic interventions aimed at correcting the metabolic impairments in patients with ASCT1 mutations.


Subject(s)
Amino Acid Transport System ASC/metabolism , Brain/physiology , Cell Communication/physiology , Microcephaly/genetics , Serine/metabolism , Amino Acid Transport System ASC/genetics , Animals , Astrocytes/physiology , Brain/cytology , Brain/diagnostic imaging , Brain/embryology , Disease Models, Animal , Glycine/metabolism , HEK293 Cells , Humans , Long-Term Potentiation/physiology , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microcephaly/diagnostic imaging , Microcephaly/metabolism , Microcephaly/pathology , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Neurons/physiology , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/physiology
9.
Neurochem Res ; 45(6): 1344-1353, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32189130

ABSTRACT

Shape-shifting, a phenomenon wide-spread in folklore, refers to the ability to physically change from one identity to another, typically from an innocuous entity to a destructive one. The amino acid D-serine over the last 25 years has "shape-shifted" into several identities: a purported glial transmitter activating N-methyl-D-aspartate receptors (NMDARs), a co-transmitter concentrated in excitatory glutamatergic neurons, an autocrine that is released at dendritic spines to prime their post-synaptic NMDARs for an instantaneous response to glutamate and an excitotoxic moiety released from inflammatory (A1) astrocytes. This article will review evidence in support of these scenarios and the artifacts that misled investigators of the true identity of D-serine.


Subject(s)
Brain/metabolism , Excitatory Amino Acid Agonists/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/metabolism , Serine/metabolism , Animals , Brain/drug effects , Excitatory Amino Acid Agonists/pharmacology , Humans , Neurons/drug effects , Serine/pharmacology
10.
Int J Mol Sci ; 21(24)2020 Dec 11.
Article in English | MEDLINE | ID: mdl-33322577

ABSTRACT

d-serine is the major co-agonist of N-methyl-D-aspartate receptors (NMDAR) at CA3/CA1 hippocampal synapses, the activation of which drives long-term potentiation (LTP). The use of mice with targeted deletion of the serine racemase (SR) enzyme has been an important tool to uncover the physiological and pathological roles of D-serine. To date, some uncertainties remain regarding the direction of LTP changes in SR-knockout (SR-KO) mice, possibly reflecting differences in inhibitory GABAergic tone in the experimental paradigms used in the different studies. On the one hand, our extracellular recordings in hippocampal slices show that neither isolated NMDAR synaptic potentials nor LTP were altered in SR-KO mice. This was associated with a compensatory increase in hippocampal levels of glycine, another physiologic NMDAR co-agonist. SR-KO mice displayed no deficits in spatial learning, reference memory and cognitive flexibility. On the other hand, SR-KO mice showed a weaker LTP and a lower increase in NMDAR potentials compared to controls when GABAA receptors were pharmacologically blocked. Our results indicate that depletion of endogenous D-serine caused a reduced inhibitory activity in CA1 hippocampal networks, altering the excitatory/inhibitory balance, which contributes to preserve functional plasticity at synapses and to maintain related cognitive abilities.


Subject(s)
CA1 Region, Hippocampal/metabolism , Racemases and Epimerases/metabolism , Amino Acids/metabolism , Animals , Electrophysiology , Hippocampus/metabolism , Long-Term Potentiation/physiology , Male , Memory/physiology , Mice , Morris Water Maze Test , Neuronal Plasticity/physiology , Racemases and Epimerases/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
11.
Cereb Cortex ; 27(2): 1573-1587, 2017 02 01.
Article in English | MEDLINE | ID: mdl-26796213

ABSTRACT

d-Serine is a co-agonist of NMDA receptors (NMDARs) whose activity is potentially regulated by Asc-1 (SLC7A10), a transporter that displays high affinity for d-serine and glycine. Asc-1 operates as a facilitative transporter and as an antiporter, though the preferred direction of d-serine transport is uncertain. We developed a selective Asc-1 blocker, Lu AE00527, that blocks d-serine release mediated by all the transport modes of Asc-1 in primary cultures and neocortical slices. Furthermore, d-serine release is reduced in slices from Asc-1 knockout (KO) mice, indicating that d-serine efflux is the preferred direction of Asc-1. The selectivity of Lu AE00527 is assured by the lack of effect on slices from Asc-1-KO mice, and the lack of interaction with the co-agonist site of NMDARs. Moreover, in vivo injection of Lu AE00527 in P-glycoprotein-deficient mice recapitulates a hyperekplexia-like phenotype similar to that in Asc-1-KO mice. In slices, Lu AE00527 decreases the long-term potentiation at the Schaffer collateral-CA1 synapses, but does not affect the long-term depression. Lu AE00527 blocks NMDAR synaptic potentials when typical Asc-1 extracellular substrates are present, but it does not affect AMPAR transmission. Our data demonstrate that Asc-1 mediates tonic co-agonist release, which is required for optimal NMDAR activation and synaptic plasticity.


Subject(s)
Amino Acid Transport System y+/genetics , Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Prosencephalon/physiology , Synapses/physiology , Animals , Excitatory Postsynaptic Potentials/physiology , Humans , Mice, Knockout , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/physiology
12.
Proc Natl Acad Sci U S A ; 112(2): E204-13, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25550512

ABSTRACT

NMDA receptors (NMDARs) require the coagonists D-serine or glycine for their activation, but whether the identity of the coagonist could be synapse specific and developmentally regulated remains elusive. We therefore investigated the contribution of D-serine and glycine by recording NMDAR-mediated responses at hippocampal Schaffer collaterals (SC)-CA1 and medial perforant path-dentate gyrus (mPP-DG) synapses in juvenile and adult rats. Selective depletion of endogenous coagonists with enzymatic scavengers as well as pharmacological inhibition of endogenous D-amino acid oxidase activity revealed that D-serine is the preferred coagonist at SC-CA1 mature synapses, whereas, unexpectedly, glycine is mainly involved at mPP-DG synapses. Nevertheless, both coagonist functions are driven by the levels of synaptic activity as inferred by recording long-term potentiation generated at both connections. This regional compartmentalization in the coagonist identity is associated to different GluN1/GluN2A to GluN1/GluN2B subunit composition of synaptic NMDARs. During postnatal development, the replacement of GluN2B- by GluN2A-containing NMDARs at SC-CA1 synapses parallels a change in the identity of the coagonist from glycine to D-serine. In contrast, NMDARs subunit composition at mPP-DG synapses is not altered and glycine remains the main coagonist throughout postnatal development. Altogether, our observations disclose an unprecedented relationship in the identity of the coagonist not only with the GluN2 subunit composition at synaptic NMDARs but also with astrocyte activity in the developing and mature hippocampus that reconciles the complementary functions of D-serine And Glycine In Modulating Nmdars During The Maturation Of Tripartite Glutamatergic Synapses.


Subject(s)
Hippocampus/growth & development , Hippocampus/metabolism , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Animals , Animals, Newborn , Astrocytes/metabolism , CA1 Region, Hippocampal/growth & development , CA1 Region, Hippocampal/metabolism , Dentate Gyrus/growth & development , Dentate Gyrus/metabolism , Glycine/metabolism , Long-Term Potentiation , Male , Neurons/metabolism , Rats , Serine/metabolism
13.
EMBO Rep ; 16(5): 590-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25755256

ABSTRACT

Asc-1 (SLC7A10) is an amino acid transporter whose deletion causes neurological abnormalities and early postnatal death in mice. Using metabolomics and behavioral and electrophysiological methods, we demonstrate that Asc-1 knockout mice display a marked decrease in glycine levels in the brain and spinal cord along with impairment of glycinergic inhibitory transmission, and a hyperekplexia-like phenotype that is rescued by replenishing brain glycine. Asc-1 works as a glycine and L-serine transporter, and its transport activity is required for the subsequent conversion of L-serine into glycine in vivo. Asc-1 is a novel regulator of glycine metabolism and a candidate for hyperekplexia disorders.


Subject(s)
Amino Acid Transport System y+/metabolism , Brain/metabolism , Glycine/metabolism , Synaptic Transmission , Amino Acid Transport System y+/genetics , Animals , Biological Transport , Genotype , Hypoglossal Nerve/cytology , Metabolome , Metabolomics/methods , Mice , Mice, Knockout , Mutation , Neurons/metabolism , Phenotype , Receptors, Glycine/genetics , Receptors, Glycine/metabolism , Serine/metabolism , Synaptic Transmission/genetics
14.
J Biol Chem ; 290(52): 31037-50, 2015 Dec 25.
Article in English | MEDLINE | ID: mdl-26553873

ABSTRACT

D-Serine is a physiological co-agonist that activates N-methyl D-aspartate receptors (NMDARs) and is essential for neurotransmission, synaptic plasticity, and behavior. D-Serine may also trigger NMDAR-mediated neurotoxicity, and its dysregulation may play a role in neurodegeneration. D-Serine is synthesized by the enzyme serine racemase (SR), which directly converts L-serine to D-serine. However, many aspects concerning the regulation of D-serine production under physiological and pathological conditions remain to be elucidated. Here, we investigate possible mechanisms regulating the synthesis of D-serine by SR in paradigms relevant to neurotoxicity. We report that SR undergoes nucleocytoplasmic shuttling and that this process is dysregulated by several insults leading to neuronal death, typically by apoptotic stimuli. Cell death induction promotes nuclear accumulation of SR, in parallel with the nuclear translocation of GAPDH and Siah proteins at an early stage of the cell death process. Mutations in putative SR nuclear export signals (NESs) elicit SR nuclear accumulation and its depletion from the cytosol. Following apoptotic insult, SR associates with nuclear GAPDH along with other nuclear components, and this is accompanied by complete inactivation of the enzyme. As a result, extracellular D-serine concentration is reduced, even though extracellular glutamate concentration increases severalfold. Our observations imply that nuclear translocation of SR provides a fail-safe mechanism to prevent or limit secondary NMDAR-mediated toxicity in nearby synapses.


Subject(s)
Cell Nucleus/enzymology , Neurons/enzymology , Racemases and Epimerases/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Serine/biosynthesis , Synapses/metabolism , Active Transport, Cell Nucleus/genetics , Animals , Cell Nucleus/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Racemases and Epimerases/genetics , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics , Serine/genetics , Synapses/genetics
15.
Proc Natl Acad Sci U S A ; 110(27): E2518-27, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23776240

ABSTRACT

Synaptic loss is the cardinal feature linking neuropathology to cognitive decline in Alzheimer's disease (AD). However, the mechanism of synaptic damage remains incompletely understood. Here, using FRET-based glutamate sensor imaging, we show that amyloid-Ɵ peptide (AƟ) engages α7 nicotinic acetylcholine receptors to induce release of astrocytic glutamate, which in turn activates extrasynaptic NMDA receptors (eNMDARs) on neurons. In hippocampal autapses, this eNMDAR activity is followed by reduction in evoked and miniature excitatory postsynaptic currents (mEPSCs). Decreased mEPSC frequency may reflect early synaptic injury because of concurrent eNMDAR-mediated NO production, tau phosphorylation, and caspase-3 activation, each of which is implicated in spine loss. In hippocampal slices, oligomeric AƟ induces eNMDAR-mediated synaptic depression. In AD-transgenic mice compared with wild type, whole-cell recordings revealed excessive tonic eNMDAR activity accompanied by eNMDAR-sensitive loss of mEPSCs. Importantly, the improved NMDAR antagonist NitroMemantine, which selectively inhibits extrasynaptic over physiological synaptic NMDAR activity, protects synapses from AƟ-induced damage both in vitro and in vivo.


Subject(s)
Amyloid beta-Peptides/toxicity , Astrocytes/metabolism , Glutamic Acid/metabolism , Neural Inhibition/physiology , Peptide Fragments/toxicity , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Astrocytes/pathology , Coculture Techniques , Female , Fluorescence Resonance Energy Transfer , HEK293 Cells , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Transgenic , Rats , Receptors, Nicotinic/metabolism , Synapses/metabolism , alpha7 Nicotinic Acetylcholine Receptor
16.
J Biol Chem ; 289(49): 33904-15, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25336657

ABSTRACT

d-Serine is a physiological activator of NMDA receptors (NMDARs) in the nervous system that mediates several NMDAR-mediated processes ranging from normal neurotransmission to neurodegeneration. d-Serine is synthesized from l-serine by serine racemase (SR), a brain-enriched enzyme. However, little is known about the regulation of d-serine synthesis. We now demonstrate that the F-box only protein 22 (FBXO22) interacts with SR and is required for optimal d-serine synthesis in cells. Although FBXO22 is classically associated with the ubiquitin system and is recruited to the Skip1-Cul1-F-box E3 complex, SR interacts preferentially with free FBXO22 species. In vivo ubiquitination and SR half-life determination indicate that FBXO22 does not target SR to the proteasome system. FBXO22 primarily affects SR subcellular localization and seems to increase d-serine synthesis by preventing the association of SR to intracellular membranes. Our data highlight an atypical role of FBXO22 in enhancing d-serine synthesis that is unrelated to its classical effects as a component of the ubiquitin-proteasome degradation pathway.


Subject(s)
F-Box Proteins/metabolism , Neuroglia/metabolism , Neurons/metabolism , Racemases and Epimerases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Serine/biosynthesis , Cell Line, Tumor , Escherichia coli/genetics , Escherichia coli/metabolism , F-Box Proteins/antagonists & inhibitors , F-Box Proteins/genetics , Gene Expression Regulation , Half-Life , Humans , Intracellular Membranes/metabolism , Neuroglia/cytology , Neurons/cytology , Proteasome Endopeptidase Complex/metabolism , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Racemases and Epimerases/genetics , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Ubiquitination
17.
J Biol Chem ; 289(43): 29631-41, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25164819

ABSTRACT

D-Serine, an endogenous co-agonist for the glycine site of the synaptic NMDA glutamate receptor, regulates synaptic plasticity and is implicated in schizophrenia. Serine racemase (SR) is the enzyme that converts L-serine to D-serine. In this study, we demonstrate that SR interacts with the synaptic proteins, postsynaptic density protein 95 (PSD-95) and stargazin, forming a ternary complex. SR binds to the PDZ3 domain of PSD-95 through the PDZ domain ligand at its C terminus. SR also binds to the C terminus of stargazin, which facilitates the cell membrane localization of SR and inhibits its activity. AMPA receptor activation internalizes SR and disrupts its interaction with stargazin, therefore derepressing SR activity, leading to more D-serine production and potentially facilitating NMDA receptor activation. These interactions regulate the enzymatic activity as well as the intracellular localization of SR, potentially coupling the activities of NMDA and AMPA receptors. This shuttling of a neurotransmitter synthesizing enzyme between two receptors appears to be a novel mode of synaptic regulation.


Subject(s)
Calcium Channels/metabolism , Glutamic Acid/metabolism , Guanylate Kinases/metabolism , Membrane Proteins/metabolism , N-Methylaspartate/metabolism , Racemases and Epimerases/metabolism , Synaptic Transmission , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism , Animals , Biocatalysis , Cell Membrane/metabolism , Disks Large Homolog 4 Protein , Female , HEK293 Cells , Humans , Male , Mice , Models, Biological , Protein Binding , Rats , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
18.
Hum Mol Genet ; 22(10): 2083-96, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23393160

ABSTRACT

Parkin E3 ubiquitin-ligase activity and its role in mitochondria homeostasis are thought to play a role in Parkinson's disease (PD). We now report that AF-6 is a novel parkin interacting protein that modulates parkin ubiquitin-ligase activity and mitochondrial roles. Parkin interacts with the AF-6 PDZ region through its C-terminus. This leads to ubiquitination of cytosolic AF-6 and its degradation by the proteasome. On the other hand, endogenous AF-6 robustly increases parkin translocation and ubiquitin-ligase activity at the mitochondria. Mitochondrial AF-6 is not a parkin substrate, but rather co-localizes with parkin and enhances mitochondria degradation through PINK1/parkin-mediated mitophagy. On the other hand, several parkin and PINK1 juvenile disease-mutants are insensitive to AF-6 effects. AF-6 is present in Lewy bodies and its soluble levels are strikingly decreased in the caudate/putamen and substantia nigra of sporadic PD patients, suggesting that decreased AF-6 levels may contribute to the accumulation of dysfunctional mitochondria in the disease. The identification of AF-6 as a positive modulator of parkin translocation to the mitochondria sheds light on the mechanisms involved in PD and underscores AF-6 as a novel target for future therapeutics.


Subject(s)
Kinesins/metabolism , Mitochondria/metabolism , Mutation , Myosins/metabolism , Parkinson Disease/metabolism , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Caudate Nucleus/metabolism , Caudate Nucleus/pathology , HEK293 Cells , Humans , Kinesins/genetics , Mitochondria/genetics , Mitochondria/pathology , Myosins/genetics , Parkinson Disease/genetics , Parkinson Disease/pathology , Protein Kinases/genetics , Protein Transport/genetics , Proteolysis , Substantia Nigra/metabolism , Substantia Nigra/pathology , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics
19.
J Neurosci ; 33(30): 12464-9, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23884950

ABSTRACT

d-Serine is an endogenous ligand for NMDARs generated from l-serine by the enzyme serine racemase (Srr). Both neuronal and glial localizations have been reported for d-serine and Srr. 3-Phosphoglycerate dehydrogenase is an exclusively astrocytic enzyme that catalyzes the first committed step of l-serine biosynthesis. Using transgenic mice expressing enhanced green fluorescent protein under the Srr promoter and mice with targeted deletion of Srr or 3-Phosphoglycerate dehydrogenase, we demonstrate predominantly neuronal sources of d-serine dependent on astrocytic supply of l-serine. These findings clarify the cellular basis for the regulation of NMDAR neurotransmission by d-serine.


Subject(s)
Astrocytes/enzymology , Neurons/enzymology , Phosphoglycerate Dehydrogenase/genetics , Phosphoglycerate Dehydrogenase/metabolism , Serine/metabolism , Animals , Astrocytes/cytology , Cerebral Cortex/cytology , Female , Glucose/metabolism , Green Fluorescent Proteins/genetics , Male , Mice , Mice, Transgenic , Neurons/cytology , Primary Cell Culture , Promoter Regions, Genetic/physiology , Racemases and Epimerases/genetics , Racemases and Epimerases/metabolism , Serine/biosynthesis , Synaptic Transmission/physiology
20.
J Neurosci ; 33(8): 3533-44, 2013 Feb 20.
Article in English | MEDLINE | ID: mdl-23426681

ABSTRACT

D-Serine and glycine are coagonists of NMDA receptors (NMDARs), but their relative contributions for several NMDAR-dependent processes are unclear. We now report that the alanine-serine-cysteine transporter-1 (Asc-1) mediates release of both D-serine and glycine from neurons, and, in turn, this modulates NMDAR synaptic activity. Asc-1 antiporter activity is enhanced by D-isoleucine (D-Ile), which releases D-serine and glycine from Asc-1-transfected cells, primary neuronal cultures, and hippocampal slices. D-Ile has no effect on astrocytes, which do not express Asc-1. We show that D-Ile enhances the long-term potentiation (LTP) in rat and mouse hippocampal CA1 by stimulating Asc-1-mediated endogenous D-serine release. D-Ile effects on synaptic plasticity are abolished by enzymatically depleting D-serine or by using serine racemase knock-out (SR-KO) mice, confirming its specificity and supporting the notion that LTP depends mostly on D-serine release. Conversely, our data also disclose a role of glycine in activating synaptic NMDARs. Although acute enzymatic depletion of D-serine also drastically decreases the isolated NMDAR synaptic potentials, these responses are still enhanced by D-Ile. Furthermore, NMDAR synaptic potentials are preserved in SR-KO mice and are also enhanced by D-Ile, indicating that glycine overlaps with D-serine binding at synaptic NMDARs. Altogether, our results disclose a novel role of Asc-1 in regulating NMDAR-dependent synaptic activity by mediating concurrent non-vesicular release of D-serine and glycine. Our data also highlight an important role of neuron-derived D-serine and glycine, indicating that astrocytic D-serine is not solely responsible for activating synaptic NMDARs.


Subject(s)
Amino Acid Transport System y+/physiology , Glycine/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Serine/metabolism , Synapses/physiology , Animals , Animals, Newborn , Cells, Cultured , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/physiology , Rats , Rats, Sprague-Dawley , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL