11
Characterization of human DAAO variants potentially related to an increased risk of schizophrenia Laura Caldinelli a, b , Silvia Sacchi a, b , Gianluca Molla a, b , Marco Nardini c , Loredano Pollegioni a, b, a Dipartimento di Biotecnologie e Scienze della Vita, Università degli studi dell'Insubria, via J. H. Dunant 3, 21100 Varese, Italy b The Protein Factory, Politecnico di Milano, ICRM-CNR and Università degli Studi dell'Insubria, via Mancinelli 7, 20131 Milano, Italy c Dipartimento di Bioscienze, Università degli Studi di Milano, via Celoria 26, 20133 Milano, Italy abstract article info Article history: Received 2 November 2012 Received in revised form 22 November 2012 Accepted 27 November 2012 Available online 5 December 2012 Keywords: Schizophrenia D-Serine Neurotransmission D-Amino acid oxidase Enzyme variants Considering the key role of D-serine in N-methyl-D-aspartate receptor-mediated neurotransmission, it is highly relevant to dene the role that enzymes play in D-serine synthesis and degradation. In particular, the details of regulation of the D-serine catabolic human enzyme D-amino acid oxidase (hDAAO) are un- known although different lines of evidence have shown it to be involved in schizophrenia susceptibility. Here we investigated the effect of three single nucleotide polymorphisms and known mutations in hDAAO, i.e., D31H, R279A, and G331V. A very low amount of soluble G331V hDAAO is produced in E. coli cells: the re- combinant variant enzyme is fully active. Human U87 glioblastoma cells transiently transfected for G331V hDAAO show a low viability, a signicant amount of protein aggregates, and augmented apoptosis. The re- combinant D31H and R279A hDAAO variants do not show alterations in tertiary and quaternary structures, thermal stability, binding afnity for inhibitors, and the modulator pLG72, whereas the kinetic efciency and the afnity for D-serine and for FAD were higher than for the wild-type enzyme. While these effects for the substitution at position 31 cannot be structurally explained, the R279A mutation might affect the hDAAO FAD-binding afnity by altering the structurally ambivalentpeptide V47L51. In agreement with the observed increased activity, expression of D31H and R279A hDAAO variants in U87 cells produces a higher decrease in cellular D/(D +L) serine ratio than the wild-type counterpart. In vivo, these substitutions could affect cellular D-serine concentration and its release at synapsis and thus might be relevant for schizo- phrenia susceptibility. © 2012 Elsevier B.V. All rights reserved. 1. Introduction D-Amino acid oxidase (EC 1.4.3.3, DAAO) is a peroxisomal FAD-containing enzyme that catalyzes the oxidative deamination of D-amino acids to their imino acid counterparts and concomitantly re- duces FAD. The imino acid is released into solvent where it spontane- ously hydrolyzes to the corresponding α-keto acid and ammonia; the reduced avin is subsequently reoxidized by dioxygen, generating H 2 O 2 [1]. In the human brain DAAO degrades D-serine, a glutamate coagonist that binds the strychnine-insensitive glycine modulatory siteon the ionotropic N-methyl-D-aspartate subtype of glutamate receptors (NMDAr) which, together with the glutamate site, must be occupied for the receptor to operate [26]. This site of the NMDAr is not saturated under normal conditions [7]; thus, even small changes in D-serine concentration may affect ionic channel function to a signicant extent (both under physiological and patho- logical conditions). D-Serine is an atypicalneuromodulator produced through the isomerization reaction catalyzed by the PLP-dependent enzyme serine racemase (SR, EC 5.1.1.18) from L-serine and degraded by the α,β-elimination reaction catalyzed by the same enzyme and/or by the oxidative deamination due to DAAO [1,6,8,9]. This signaling molecule plays key roles in excitatory synaptic transmission and it is also involved in a number of physiological processes related to neuronal plasticity and in several pathological conditions [4,5,1013]. A recent model of D-serine dynamics the so-called serine shuttle model[9] proposes that as- trocytes synthesize and export L-serine to neurons, inducing neuronal SR to synthesize D-serine; D-serine released by neurons can be taken up by astrocytes for storage and activity-dependent release. Various studies have indicated that D-serine levels are decreased in patients affected by schizophrenia [14,15]; furthermore, D-serine great- ly ameliorated the symptoms of schizophrenia when administered Biochimica et Biophysica Acta 1832 (2013) 400410 Abbreviations: CD, circular dichroism; CPZ, chlorpromazine; DAAO, D-amino acid oxidase (EC 1.4.3.3); EYFP, enhanced yellow uorescent protein; FAD, avin adenine dinucleotide; hDAAO, human D-amino acid oxidase; NLS, N-lauroylsarcosine; NMDAr, N-methyl-D-aspartate subtype of glutamate receptor; pkDAAO, pig kidney D-amino acid oxidase; PMP70, peroxisomal membrane protein 70; SAP, structural ambivalent peptide; SNP, single nucleotide polymorphism; SR, serine racemase (EC 5.1.1.18) Corresponding author at: Dipartimento di Biotecnologie e Scienze della Vita, Università degli studi dell'Insubria, via J. H. Dunant 3, 21100 Varese, Italy. Tel.: +39 0332421506; fax: +39 0332421500. E-mail address: [email protected] (L. Pollegioni). 0925-4439/$ see front matter © 2012 Elsevier B.V. All rights reserved. http://dx.doi.org/10.1016/j.bbadis.2012.11.019 Contents lists available at SciVerse ScienceDirect Biochimica et Biophysica Acta journal homepage: www.elsevier.com/locate/bbadis

Biochimica et Biophysica Acta - COnnecting REpositories · 2017. 1. 6. · Characterization of human DAAO variants potentially related to an increased risk of schizophrenia Laura

  • Upload
    others

  • View
    7

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Biochimica et Biophysica Acta - COnnecting REpositories · 2017. 1. 6. · Characterization of human DAAO variants potentially related to an increased risk of schizophrenia Laura

Biochimica et Biophysica Acta 1832 (2013) 400–410

Contents lists available at SciVerse ScienceDirect

Biochimica et Biophysica Acta

j ourna l homepage: www.e lsev ie r .com/ locate /bbad is

Characterization of human DAAO variants potentially related to an increased riskof schizophrenia

Laura Caldinelli a,b, Silvia Sacchi a,b, Gianluca Molla a,b, Marco Nardini c, Loredano Pollegioni a,b,⁎a Dipartimento di Biotecnologie e Scienze della Vita, Università degli studi dell'Insubria, via J. H. Dunant 3, 21100 Varese, Italyb The Protein Factory, Politecnico di Milano, ICRM-CNR and Università degli Studi dell'Insubria, via Mancinelli 7, 20131 Milano, Italyc Dipartimento di Bioscienze, Università degli Studi di Milano, via Celoria 26, 20133 Milano, Italy

Abbreviations: CD, circular dichroism; CPZ, chlorproxidase (EC 1.4.3.3); EYFP, enhanced yellow fluorescendinucleotide; hDAAO, human D-amino acid oxidase; NLN-methyl-D-aspartate subtype of glutamate receptor;acid oxidase; PMP70, peroxisomal membrane protein 7peptide; SNP, single nucleotide polymorphism; SR, serin⁎ Corresponding author at: Dipartimento di Biotecnologi

degli studi dell'Insubria, via J. H. Dunant 3, 21100 Varesefax: +39 0332421500.

E-mail address: [email protected] (L

0925-4439/$ – see front matter © 2012 Elsevier B.V. Alhttp://dx.doi.org/10.1016/j.bbadis.2012.11.019

a b s t r a c t

a r t i c l e i n f o

Article history:Received 2 November 2012Received in revised form 22 November 2012Accepted 27 November 2012Available online 5 December 2012

Keywords:SchizophreniaD-SerineNeurotransmissionD-Amino acid oxidaseEnzyme variants

Considering the key role of D-serine in N-methyl-D-aspartate receptor-mediated neurotransmission, it ishighly relevant to define the role that enzymes play in D-serine synthesis and degradation. In particular,the details of regulation of the D-serine catabolic human enzyme D-amino acid oxidase (hDAAO) are un-known although different lines of evidence have shown it to be involved in schizophrenia susceptibility.Here we investigated the effect of three single nucleotide polymorphisms and known mutations in hDAAO,i.e., D31H, R279A, and G331V. A very low amount of soluble G331V hDAAO is produced in E. coli cells: the re-combinant variant enzyme is fully active. Human U87 glioblastoma cells transiently transfected for G331VhDAAO show a low viability, a significant amount of protein aggregates, and augmented apoptosis. The re-combinant D31H and R279A hDAAO variants do not show alterations in tertiary and quaternary structures,thermal stability, binding affinity for inhibitors, and the modulator pLG72, whereas the kinetic efficiencyand the affinity for D-serine and for FAD were higher than for the wild-type enzyme. While these effectsfor the substitution at position 31 cannot be structurally explained, the R279A mutation might affect thehDAAO FAD-binding affinity by altering the “structurally ambivalent” peptide V47–L51. In agreement withthe observed increased activity, expression of D31H and R279A hDAAO variants in U87 cells produces ahigher decrease in cellular D/(D+L) serine ratio than the wild-type counterpart. In vivo, these substitutionscould affect cellular D-serine concentration and its release at synapsis and thus might be relevant for schizo-phrenia susceptibility.

© 2012 Elsevier B.V. All rights reserved.

1. Introduction

D-Amino acid oxidase (EC 1.4.3.3, DAAO) is a peroxisomalFAD-containing enzyme that catalyzes the oxidative deamination ofD-amino acids to their imino acid counterparts and concomitantly re-duces FAD. The imino acid is released into solvent where it spontane-ously hydrolyzes to the corresponding α-keto acid and ammonia; thereduced flavin is subsequently reoxidized by dioxygen, generatingH2O2 [1]. In the human brain DAAO degrades D-serine, a glutamatecoagonist that binds the “strychnine-insensitive glycine modulatorysite” on the ionotropic N-methyl-D-aspartate subtype of glutamate

omazine; DAAO, D-amino acidt protein; FAD, flavin adenineS, N-lauroylsarcosine; NMDAr,pkDAAO, pig kidney D-amino0; SAP, structural ambivalente racemase (EC 5.1.1.18)e e Scienze della Vita, Università, Italy. Tel.: +39 0332421506;

. Pollegioni).

l rights reserved.

receptors (NMDAr) which, together with the glutamate site, mustbe occupied for the receptor to operate [2–6]. This site of theNMDAr is not saturated under normal conditions [7]; thus, evensmall changes in D-serine concentration may affect ionic channelfunction to a significant extent (both under physiological and patho-logical conditions).

D-Serine is an “atypical” neuromodulator produced through theisomerization reaction catalyzed by the PLP-dependent enzyme serineracemase (SR, EC 5.1.1.18) from L-serine and degraded by theα,β-elimination reaction catalyzed by the same enzyme and/or by theoxidative deamination due to DAAO [1,6,8,9]. This signaling moleculeplays key roles in excitatory synaptic transmission and it is also involvedin a number of physiological processes related to neuronal plasticity andin several pathological conditions [4,5,10–13]. A recentmodel of D-serinedynamics – the so-called “serine shuttle model” [9] – proposes that as-trocytes synthesize and export L-serine to neurons, inducing neuronalSR to synthesize D-serine; D-serine released by neurons can be takenup by astrocytes for storage and activity-dependent release.

Various studies have indicated that D-serine levels are decreased inpatients affected by schizophrenia [14,15]; furthermore, D-serine great-ly ameliorated the symptoms of schizophrenia when administered

Page 2: Biochimica et Biophysica Acta - COnnecting REpositories · 2017. 1. 6. · Characterization of human DAAO variants potentially related to an increased risk of schizophrenia Laura

401L. Caldinelli et al. / Biochimica et Biophysica Acta 1832 (2013) 400–410

together with conventional neuroleptics [16]. In recent years, severalgenes linked to schizophrenia susceptibility have been discovered[17]. Among them single nucleotide polymorphisms (SNP) and haplo-types in the genes encoding for DAAO, pLG72, and SR have been identi-fied, reviewed in [6]. The human G72 gene encodes for pLG72, a proteinpresent in primates only, which interacts with human DAAO (hDAAO)in astrocytes [18,19] and alters its stability and cellular half-life[20,21]. We showed that newly synthesized hDAAO is transitorily pres-ent in the cytoplasm, where it can interact with pLG72 located on thecytosolic side of themitochondrial external membrane before being de-livered to peroxisomes [21], and demonstrated that the cellular concen-trations of D-serine depend on expression of the active form of thisflavooxidase [19–21]. A model for correlating D-serine catabolism withschizophrenia susceptibility was proposed: a decrease in pLG72 expres-sion yields an anomalously high level of DAAO activity and induces anexcessive decrease in the local concentration of D-serine, see Scheme 1[6,19]. Importantly, a similar effect can also result from mutations thatabnormally increase flavoenzyme activity or affect its downregulation.In fact, diminished DAAO activity (by expressing the inactive G181Rvariant in mice) slightly enhanced D-serine levels in the brain and im-proved behavioral deficits associated with the negative and cognitivesymptoms of schizophrenia [22].

The hDAAO protomer (347 amino acids) shows the classicalαβ-structure of FAD-containing flavooxidases constituted by two do-mains: the FAD binding domain and the interface domain (Fig. 1A)[23,24]. hDAAO differs from other known mammalian homologues be-cause of its stable homodimeric state (also in the apoprotein form), aweak interaction with the FAD cofactor (in the absence of an active siteligand), and the specific interaction with pLG72 [1,19,21]. Various SNPsof the hDAAO gene resulting in nonsynonymous substitutions havebeen reported: themost well-known SNP (rs4262766, related to schizo-phrenia susceptibility) resulted in G331V replacement. Furthermore,

Scheme 1. Proposed role of hDAAO–pLG72 interaction in the modulation of D-serine availabacts on the postsynaptic neuron as well as on the surrounding astrocytes, activating metabcatalyzed by SR). In the synaptic cleft, D-serine binds to the glycine-binding site on the NMIn glial cells pLG72 binds to hDAAO, yielding formation of a complex constituted by twinactivated: pLG72 modulates the amount of active hDAAO acting on the stability of the hoUnder pathological conditions, the supposed lower expression level of pLG72 would reconcentration.

analysis of deposited protein sequences reveals a discrepancy at posi-tions 31 and 279 (yielding the D31H and R279A replacements) [25]: itis unclear at this stage whether these latter differences are due to poly-morphisms, sequencing, or cloning artifacts. Inspection of the 3D struc-ture of hDAAO (see Fig. 1) has shown that Arg279 is located close tothe FAD cofactor and that Asp31 and Gly331 are far from both the activesite and the interface between monomers, in a region that is likely in-volved in the protein folding process [26].

With the ultimate goal of enhancing our understanding of the cor-relation between enzymatic activity modulation in this flavoproteinand its possible role in schizophrenia susceptibility, we investigatedthe effect of D31H, R279A, and G331V replacements on enzyme func-tion by expressing and characterizing the corresponding single-pointhDAAO variants.

2. Materials and methods

2.1. Preparation and expression of hDAAO variants in E. coli

Mutagenesis reactions were performed using the QuickChangesite-directed mutagenesis kit (Stratagene) on the pET11b-HishDAAOexpression plasmid. Recombinant His-tagged hDAAO (wild-type andvariants) was expressed in E. coli cells and purified by HiTrap chelatingchromatography (GE Healthcare) as reported in [27]. The final hDAAOpreparations were equilibrated in 20 mM Tris–HCl buffer, pH 8.0,100 mM NaCl, 5% glycerol, 5 mM 2-mercaptoethanol, and 40 μM FADand diluted in plain buffer without FAD before use. Protein concentra-tion of the purified enzymes was determined using the extinction coef-ficient at 455 nm (12.2 mM−1 cm−1 for wild-type hDAAO) [27].

hDAAO apoprotein was prepared by dialysis of a 2 mg protein/mLenzyme solution vs. 50 mM sodium pyrophosphate, pH 8.3, 1 M KBr,and 1 mM EDTA for 48 h at 4 °C. The final apoprotein preparation

ility at glutamatergic synapses [6,9,19]. Glutamate released from presynaptic neuronsotropic glutamate receptors mGluR5 and synthesizing D-serine (through the reactionDAr and in conjunction with glutamate results in the opening of the receptor channel.o hDAAO homodimers and two pLG72 molecules. In this way complexed hDAAO isloenzyme, thus preventing an excessive degradation of the neuromodulator D-serine.sult in an overactivation of hDAAO and a consequent decrease in D-serine cellular

Page 3: Biochimica et Biophysica Acta - COnnecting REpositories · 2017. 1. 6. · Characterization of human DAAO variants potentially related to an increased risk of schizophrenia Laura

Fig. 1. Structure of human DAAO. A) Position of the residues mutated in this work (shown in color) on the structure of hDAAO (PDB code: 2E49) [24] dimer (left) and protomer(right). B) Detail of the H-bond interaction (dashed line) between Arg279 and Pro41 (shown in yellow). Helix α2 and the VAAGL motif are shown in cyan, with the FAD cofactorin green. An adjacent hDAAO molecule in the crystal asymmetric unit is shown in orange. Figure prepared with PyMOL (http://www.pymol.org/).

402 L. Caldinelli et al. / Biochimica et Biophysica Acta 1832 (2013) 400–410

was stored in 50 mM sodium pyrophosphate, pH 8.3, 5% glycerol, and5 mM 2-mercaptoethanol.

pLG72was prepared as stated in [28] and stored in 20 mMTris–HCl,pH 8.5, 100 mM NaCl, 5% glycerol, and 5 mM 2-mercaptoethanol andcontained 0.1% N-lauroylsarcosine (NLS).

2.2. Activity assay

The standard assay for DAAO activity was performed by employingan oxygen electrode at air saturation (0.253 mM O2) and 25 °C, using

28 mM D-alanine as substrate in 100 mM sodium pyrophosphatebuffer, pH 8.5, containing 40 μM FAD [27]. One DAAO unit is de-fined as the amount of enzyme that converts 1 μmol of D-aminoacid per minute at 25 °C. The kinetic parameters were determinedwith the same assay, employing increasing concentrations of D-ala-nine or D-serine as substrate (0.25–50 mM for D-alanine and 0.25–100 mM for D-serine). The initial reaction rates at different sub-strate concentrations were used to calculate the apparent kineticparameters (kcat,app and Km,app) using the KaleidaGraph software(Synergy Software).

Page 4: Biochimica et Biophysica Acta - COnnecting REpositories · 2017. 1. 6. · Characterization of human DAAO variants potentially related to an increased risk of schizophrenia Laura

403L. Caldinelli et al. / Biochimica et Biophysica Acta 1832 (2013) 400–410

2.3. Spectral measurements

All spectral measurements were carried out at 15 °C. The dissoci-ation constants for benzoate binding to hDAAO (0.4 mg protein/mL,≈10 μM) were assessed spectrophotometrically in 20 mM Tris–HCl,pH 8.0, 5% glycerol, and 5 mM 2-mercaptoethanol by following thechanges in absorbance at 497 nm that accompany complex formation[27,29]. Flavin and protein fluorescence were measured in a JascoFP-750 instrument (Cremello, Italy) at 0.1 mg/mL protein concentra-tion in 50 mM sodium pyrophosphate, pH 8.3, and corrected for buff-er contributions. Protein emission fluorescence spectra wererecorded between 300 and 400 nm, with excitation at 280 nm; flavinemission spectra were recorded from 475 to 600 nm, with excitationat 450 nm. FAD binding was estimated by protein fluorescencequenching at 341 nm.

Circular dichroism (CD) spectra were recorded in 20 mM Tris–HCl, pH 8.5, 150 mM sodium chloride, and 5% glycerol, using a JascoJ-815 spectropolarimeter, and analyzed by means of Jasco software.The cell path length was 1 cm for measurements above 250 nm(0.4 mg protein/mL) and 0.1 cm for measurements in the 190- to250-nm region (0.1 mg protein/mL) [20].

Temperature-ramp experiments were performed in 20 mM Tris–HCl, pH 8.0, 100 mM sodium chloride, 5% glycerol, and 5 mM2-mercaptoethanol as reported previously [30]. For this a software-driven, Peltier-equipped fluorimeter was used to measure the proteinfluorescence change at 340 nm or a CD spectropolarimeter to followthe CD signal at 220 nm in order to reproduce a temperature gradient(0.5 °C/min).

2.4. Oligomerization state and pLG72 binding

Size-exclusion chromatography was performed at room tempera-ture on a Superdex 200 column by means of an Akta chromatographicsystem (GE Healthcare), using 20 mM Tris–HCl, pH 8.5, 150 mM so-dium chloride, 5% glycerol, and 5 mM 2-mercaptoethanol as elutionbuffer. hDAAO–pLG72 complex formation and oligomeric state werealso determined by gel-permeation chromatography using the afore-mentioned buffer, to which 0.06% NLS and 40 μM FAD were added asthe elution buffer (the detergent is required because the solubilityand the oligomeric state of pLG72 strongly depends on the presenceof NLS) [19]. The area of each peak was estimated by nonlinearcurve fitting of the elution profile using PeakFit software (Systat Soft-ware, Germany). The amount of pLG72 and hDAAO present in thepeak corresponding to the protein complex (≈12.8 mL) was estimat-ed by means of the intensity of their bands following SDS-PAGE, asobtained using the program QuantityOne (BioRad Labs.).

2.5. Expression of hDAAO variants in U87 glioblastoma cells

The pEYFP-hDAAO-C3 construct [19,21] was used as a templateand the substitutions were introduced by the Quickchange system.This plasmid encodes for a chimeric hDAAO fused to the enhancedyellow fluorescent protein (EYFP) located at the N-terminus ofhDAAO. The ATCC U87 human glioblastoma cells were maintainedin DMEM supplemented with 10% fetal bovine serum, 1 mM sodiumpyruvate, 2 mM L-glutamine, 1% nonessential amino acids, 1% penicil-lin/streptomycin, and 1% amphotericin B (Euroclone) at 37 °C in a 5%CO2 incubator as reported in [19,21]. They were then transfectedusing the FuGENE HD transfection reagent (Roche) and 2 μg of thedifferent pEYFP-hDAAO constructs encoding for wild-type or variantsof hDAAO. Protein expression levels were monitored either by using afluorescence microscope (Olympus IX51) equipped with an FITC filteror by Western blot analysis. The transfected cells were used for fur-ther experiments or to select stable clones, adding 0.4 mg/mL G418to the growth medium [19,21]. The expression level of hDAAO wasassessed on stably transfected cells. These cells were suspended in

SDS-PAGE sample buffer to reach 2500 cells/μL: 20 μL of each samplewas subjected to SDS-PAGE electrophoresis and Western blot analy-ses. The immunoreactivity signals were detected by enhanced chemi-luminescence (ECL Plus, GE Healthcare) using rabbit polyclonalanti-hDAAO (1:5000, Davids Biotecnologie) or rabbit polyclonalanti-actin (1:200, Sigma) as primary antibody [19].

Immunostaining and confocal analyses were performed as statedin [19,21]. Briefly, U87 cells were seeded on round glass coverslips(2.5×105 cells each), incubated for 24 h at 37 °C and 5% CO2, andtransfected with pEYFP-hDAAO constructs encoding for wild-type orvariants of hDAAO. At 24 and 48 h after transfection, the coverslipswere extensively washed with PBS (10 mM dibasic sodium phos-phate, 2 mM monobasic potassium phosphate, 137 mM NaCl, and2.7 mM KCl, pH 7.4) and fixed with 4% p-formaldehyde and 4% su-crose in 0.1 M sodium phosphate buffer, pH 7.4, for 15 min at roomtemperature. The fixed cells were permeabilized and the unspecificbinding sites were blocked by incubation in PBS supplemented with0.2% Triton X-100 and 4% horse serum. They were subsequentlystained by using primary rabbit polyclonal anti-PMP70 (peroxisomalmembrane protein 70, 1:200, Sigma) and mouse monoclonalanti-ubiquitin antibody (F-11, 1:150, Santa Cruz Biotechnology),followed by secondary anti-rabbit Alexa 647 and anti-mouse Alexa546 antibodies (1:1000, Molecular Probes). Immunostained cover-slips were imaged using an inverted laser scanning confocal micro-scope (TCS SP5, Leica Microsystems), equipped with a 63.0×1.25NA plan apochromate oil immersion objective. Confocal image stackswere acquired using the Leica TCS software with a sequential mode toavoid interference between each channel and without saturating anypixel, as reported in [19,21].

Cellular D- and L-serine levels were determined as stated in [19,20].Briefly, 2.5×105 U87 cells stably transfected with one of the aforemen-tioned pEYFP-hDAAO expression plasmids were suspended in 1 mL ofice-cold 5% trichloroacetic acid, sonicated, and centrifuged for 45 min at16,000 g. The soluble fraction was extracted with water-saturated ether,neutralized, and derivatized with o-phthaldialdehyde/N-acetyl-L-cysteinein borate buffer. D- and L-serine were resolved by HPLC chromatographyon a 5-μm Waters C8 (4.6×250 mm) reversed-phase column; amountswere determined using a calibration curve obtained with standard D- andL-serine. The analyseswere replicatedfive times for each condition and sta-tistical analyses were performed using Kaleidagraph software (SynergySoftware). Variation between groups was evaluated by one-way ANOVA,and post-hoc significance tests were performed using Student's ttest. Significance was assessed at pb0.05.

For Western blot analyses, U87 cells seeded in a 6-well plate(2.5×105 cells/well) and transfected with the plasmids encoding forwild-type or variants of hDAAO were detached by trypsin treatment at24 and 48 h after transfection, collected by centrifugation, washed withPBS, and sonicated in lysis buffer containing 20 mM Tris–HCl, pH 8.0,150 mM NaCl, 0.5% NP40, 0.5% Triton X-100, 1 mM EDTA, 0.7 μg/mLpepstatin, 1 μg/mL leupeptin, and 0.19 mg/mL PMSF. The cell lysateswere clarified by centrifugation, separated by SDS-PAGE gel (20 μg oftotal protein/lane), and blotted on a PVDF membrane. The expressionlevel of the different EYFP-hDAAO variants was detected by using rabbitpolyclonal anti-hDAAOantibodies (1:3000, Davids Biotecnologie) and anHRP-conjugated anti-rabbit secondary antibody (1:15000, JacksonImmunoResearch). Detection of α-actin using a rabbit polyclonalanti-α-actin antibody (1:1000, Sigma) was used as loading control.Blots were developed using the ECL Prime kit (GE Healthcare). Densito-metric analysis of the immunorecognized bands was performed usingthe Quantity One software (Biorad): each hDAAO-quantified signal wasnormalized by the corresponding α-actin signal.

2.6. Caspase-Glo 3/7 assay

U87 cells were seeded in 96-well plates at a density of 104 cells/well, incubated for 24 h at 37 °C and 5% CO2, and transfected using

Page 5: Biochimica et Biophysica Acta - COnnecting REpositories · 2017. 1. 6. · Characterization of human DAAO variants potentially related to an increased risk of schizophrenia Laura

404 L. Caldinelli et al. / Biochimica et Biophysica Acta 1832 (2013) 400–410

the FuGENE HD transfection reagent (Roche) with 0.1 μg of thepEYFP-hDAAO wild-type, pEYFP-hDAAO-G331V, or pEYFP constructs.After 24 or 48 h of incubation at 37 °C and 5% CO2, caspase-3 andcaspase-7 activity in the transfected cells was measured using theCaspase-Glo 3/7 Assay kit (Promega). According to the manufacturer'sinstructions, 100 μL of the Caspase-GloW 3/7 reagent was added toeach well and the cells were incubated for 2 h at 25 °C. The light emis-sion – produced by caspase substrate cleavage, aminoluciferin release,and the subsequent luciferase reaction – was measured by a platereader (Infinite 200, Tecan). The suggested controls were analyzed inparallel.

3. Results

3.1. Expression of hDAAO variants in E. coli

The D31H and R279A variants of hDAAO were efficientlyoverexpressed as soluble proteins in BL21(DE3) Star E. coli cells(≈20 and 10 mg/L of fermentation broth, respectively) using thesame conditions set up for the wild-type flavoenzyme [27]. In con-trast, under these conditions the G331V hDAAO variant wasexpressed as inclusion bodies. This hDAAO variant was obtained insoluble form only by using the E. coli Origami (DE3) cells as host, aK-12 derivative strain with mutations in both the thioredoxin reduc-tase and glutathione reductase genes and which greatly enhances di-sulfide bond formation in the cytoplasm and expression of activeproteins. Nonetheless, G331V hDAAO expression level was too lowfor a detailed biochemical characterization (b0.1 mg/L of fermenta-tion broth). In order to increase its expression, a simplified factorialdesign approach was employed [31] by investigating the growth me-dium (LB or TB), the OD600nm at the moment of inducer addition (0.8–3.8), the time of cell collection (0, 3, 5, 20 h), and a lower temperatureof growth (25 or 30 °C) after adding IPTG. No significant increase inthe expression of soluble and active G331V hDAAO variant wasreached. Analogously, no improvement in enzyme variant recoverywas observed by adding 1 mM benzoate (a competitive hDAAO inhib-itor, see below) during the purification procedure. Indeed, Westernblot analysis of recombinant E. coli whole cells (resuspended inSDS-PAGE sample buffer) excluded proteolytic processing of theexpressed hDAAO variant (not shown). The specific activity of the par-tially purified G331V hDAAO variant obtained by HiTrap chelatingchromatography (≈25% purity) was 2.9 U/mg protein (correspondingto 11.6 U/mg protein for the pure enzyme variant vs. 12 U/mg proteinfor the wild-type one). Taken together, these results indicate thatG331V substitution significantly affects the expression of a solubleform of hDAAO but does not influence its enzymatic competence.

Recombinant D31H and R279A hDAAO variants were purified bya single chromatographic step on the HiTrap chelating column witha ≥60% recovery and a ≥90% purity, as judged by SDS-PAGE. Thespecific activity was 10.5 and 14.0 U/mg protein for R279A andD31H variants vs. 12 U/mg protein for the wild-type hDAAO, seeTable 1 [27].

Table 1Comparison of the apparent kinetic parameters of wild-type and variants of hDAAO. The ac

Enzyme Specific activitya kcat (s−1)

(U/mg protein) D-serine D-alanine

Wild-type 12.0 3.0±0.1 5.2±0.1D31H 14.0 3.0±0.2 6.0±0.2R279A 10.5 2.5±0.1 8.6±0.1G331V 2.9 (≈11.6)b n.d. n.d.

n.d.=not determined.a Specific activity was determined using the oxygen-consumption assay at 28 mM D-alanb Value estimated based on a ≈25% purity (see text for details).

3.2. Biochemical properties of expressed hDAAO variants

The visible absorption spectrum of purified D31H and R279A vari-ants in the oxidized state did not significantly differ from that of thewild-type hDAAO holoenzyme (Fig. 2A), nor did the near- andfar-UV CD spectra (not shown). These results indicate that the substi-tutions at position 31 and 279 did not alter the overall (secondary andtertiary) protein conformation of hDAAO or the microenvironmentsurrounding the flavin cofactor.

The temperature sensitivity of specific features of the holo- andapoprotein forms of wild-type, D31H, and R279A hDAAOs was com-pared using temperature-ramp experiments. By following changesin tryptophan fluorescence (taken as reporter of tertiary structuremodification) and in CD signal at 220 nm (as reporter of the changein secondary structure) midpoint transition temperatures wereobtained that were marginally higher for the D31H in the holoen-zyme form and for the apoprotein of R279A variant than for thewild-type hDAAO (Suppl. Table 1).

The apparent kinetic parameters of hDAAO variants were deter-mined using the oxygen-consumption assay at fixed 21% oxygen sat-uration and in the presence of 40 μM FAD in the assay mixture [27].The Km for the physiological substrate D-serine for D31H and R279AhDAAO variants was 2-fold lower with respect to the wild-type en-zyme, whereas the kcat value was similar (Table 1). In contrast, an in-crease in apparent kcat was evident on D-alanine while the Km wasunchanged. Interestingly, the kinetic efficiency is higher for both var-iants and on both the D-amino acids used than for wild-type hDAAO.

Concerning inhibitor binding, the D31H and R279A hDAAO variantsdid not show significant alterations in binding affinity for benzoate – asubstrate-competitive inhibitor that binds to the oxidized hDAAOholoen-zyme and yields a shoulder at 493 nm in the visible absorption spectrumof the holoenzyme, see Fig. 2B [27] – as compared to wild-type hDAAO,see Table 2. Similarly, chlorpromazine (CPZ), an FAD-competitive inhibi-tor and a drug widely employed for the treatment of schizophrenia[19,20], did not show alterations in binding affinity either.

In solution, wild-type hDAAO exists as an equilibrium of holo- andapoprotein forms because of the low affinity for the FAD cofactor(Kd=8 μM) [27]. Analogously to the wild-type enzyme, the apoproteinform of both D31H and R279A hDAAOs was obtained by dialysis vs. achaotropic salt (1 M KBr): for both variants, the apoprotein maintainedthe secondary structure of the holoenzymewhile the CD band centeredat ≈270 nm, corresponding to the tertiary structure, was lost (notshown). These properties resemble those of wild-type hDAAO [27,30].The Kd for cofactor binding to the apoprotein of the D31H and R279AhDAAO variants was determined by following the change in proteinfluorescence intensity at increasing FAD concentrations (Fig. 2C). Im-portantly, both enzyme variants showed a higher affinity for the FADcofactor than thewild-typehDAAO(up to 10-fold for the R279Avariant,Table 2). Indeed,when thewild-type and hDAAO variantswere saturat-ed by benzoate (1 mM) a ≈15- to 30-fold increase in affinity for FADwas observed (Table 2): even in the benzoate-complex form, theD31H and R279A variants again interacted more tightly with the cofac-tor than the wild-type hDAAO.

tivity was assayed at air saturation (0.25 mM oxygen), 25 °C and pH 8.5.

Km (mM) kcat/Km (s−1 mM−1)

D-serine D-alanine D-serine D-alanine

7.5±0.5 1.1±0.2 0.40 4.73.9±0.9 0.95±0.10 0.77 6.33.4±0.5 1.0±0.1 0.74 8.6n.d. n.d. n.d. n.d.

ine and in the presence of 40 μM FAD [27].

Page 6: Biochimica et Biophysica Acta - COnnecting REpositories · 2017. 1. 6. · Characterization of human DAAO variants potentially related to an increased risk of schizophrenia Laura

Table 2Comparison of binding affinity (Kd) for benzoate, CPZ and FAD of wild-type and vari-ants of hDAAO.

Benzoate (μM) CPZ (μM) FAD (μM)a

Free form Benzoate complex

Wild-type 7.0±2.0 5.0±0.1 8.0±2.0 0.3±0.1D31H 4.6±0.4 4.8±0.3 1.8±0.1 0.13±0.02R279A 8.0±0.5 6.4±0.3 0.9±0.1 0.030±0.002

Experiments were performed in 20 mM Tris–HCl, pH 8.0, containing 5% glycerol, and5 mM 2-mercaptoethanol at 15 °C.

a In 50 mM sodium pyrophosphate buffer, pH 8.3.

405L. Caldinelli et al. / Biochimica et Biophysica Acta 1832 (2013) 400–410

As observed forwild-type hDAAO, theD31H and R279A variants in ho-loenzyme form retained the dimeric oligomerization state, as judged bygel-permeation chromatography. Furthermore, the samemethodwaspre-viously used to investigate the interaction of wild-type hDAAO with itsphysiological binding partner pLG72, yielding a 200-kDa complex com-posed of two hDAAO homodimers and two pLG72 monomers [19]. Forthe hDAAO–pLG72 complex formation a Kd in the 1–5×10−7 M rangewas determined [20]. Gel-permeation analysis demonstrated that the abil-ity to interactwithpLG72wasmaintained for bothhDAAOvariants (Suppl.Fig. 1). The analysis of the saturation of the area of the peak at 12.8 mL atincreasing pLG72 concentrations (inset of Suppl. Fig. 1A, B) according tothe Stinson's method [29] estimated a Kd of≈10−7 M, thus demonstrat-ing that the D31H and R279A substitutions did not affect the strength ofhDAAO–pLG72 complex formation.

3.3. Structural considerations of expressed hDAAO variants

Asp31, the first residue of strand β2, is located far from both thesubstrate and the FAD binding sites (Fig. 1A). Therefore, the effectsof D31H substitution can be explained at a structural level, assumingonly a general, long-range interaction that affects protein conforma-tion and propagates from the mutation site to the hDAAO interior.In the case of the R279A substitution, in contrast, the absence of theguanidyl group can be predicted to have a direct role in hDAAO–FAD interaction. Arg279 is located in strand β12, which on one sidecontacts the FAD binding site at the isoalloxazine ring and on theother contributes to the interface between two dimers in thehDAAO crystals (Fig. 1B). The failure of any attempt to crystallizethe hDAAO R279A protein suggests that structural alterations mayoccur in the mutation site's surroundings, thus influencing (directlyor indirectly) both the crystal contact region and the FAD bindingpocket. Indeed, strand β12 contacts the α2 region containing the hy-drophobic stretch (residues 47–51: VAAGL), which covers the si-faceof the flavin ring, and analysis of different hDAAO structures showsthat in several cases (PDB codes 2E4A, 2E49, 3CUK, and 3G3E)Arg279 is directly involved in H-bonding of the carbonyl oxygen ofPro41 (immediately preceding theα2 helix) (Fig. 1B). This hydropho-bic stretch has been proposed to play an important role in determin-ing the affinity for FAD and to be a structurally ambivalent peptide(SAP) [24] since it can assume different conformations, dependingon the local and/or global context of the protein. In particular, this hy-drophobic stretch is structurally different in hDAAO and porcineDAAO (pkDAAO, where a structural transition occurs between the re-duced and oxidized forms), while maintaining the same sequence,also in the flanking regions (residues 43–55). Thus, the weakerFAD binding and the slower rate of flavin reduction in hDAAO, ascompared to pkDAAO, was correlated to hDAAO in the global context.Here, the VAAGL stretch is restricted to a single conformation with

Fig. 2. Spectral properties of wild-type and variants of hDAAO. A) Visible absorbance spec-trum of wild-type (———), D31H (∙∙∙∙) and R279A (− − −) hDAAOs in the oxidized state,at 15 °C. B) Effect of benzoate binding to oxidized D31H (∙∙∙∙) and R279A (− −−) variantsof hDAAOs: 0.28 mM benzoate was added to 10 μM of hDAAO at pH 8.0 and 15 °C.C) Changes in fluorescence signal at 341 nm following the titration of 1 μM apoprotein ofwild-type, D31H- or R279A-hDAAOwith increasing FAD concentrations, at pH 8.3 and15 °C.

Page 7: Biochimica et Biophysica Acta - COnnecting REpositories · 2017. 1. 6. · Characterization of human DAAO variants potentially related to an increased risk of schizophrenia Laura

Fig. 3. Time course of subcellular distribution upon transient transfection in U87 cells for wild-type, D31H, and R279A EYFP-hDAAO variants. U87 cells transiently transfected with EYFP-hDAAO, EYFP-hDAAO-D31H, or EYFP-hDAAO-R279Aencoding plasmids were immunostained at 24, 48, and 72 h after transfection. The distribution of the EYFP-hDAAO is compared with the distribution of peroxisomal marker PMP70 (red channel). Bar=5 μm.

406L.Caldinelliet

al./Biochim

icaet

BiophysicaActa

1832(2013)

400–410

Page 8: Biochimica et Biophysica Acta - COnnecting REpositories · 2017. 1. 6. · Characterization of human DAAO variants potentially related to an increased risk of schizophrenia Laura

407L. Caldinelli et al. / Biochimica et Biophysica Acta 1832 (2013) 400–410

the consequent loss of the H-bond between flavin N5 atom and thebackbone N atom of Ala49, present instead in the oxidized pkDAAO(3.0 Å) and partly weakened in the reduced form (3.3 Å) [24,32].Based on this evidence, it is tempting to propose that the R279A mu-tation alters the structure of the hDAAO SAP region (directly throughthe loss of the H-bond to Pro41, or indirectly through a generalrearrangement of the β12–α2 interface region) (Fig. 1B), and conse-quently the FAD binding affinity in the hDAAO R279A variant (Kd=0.9 μM) relative to the wild-type enzyme (Kd=8.0 μM, Table 2). Inthis context, the SAP region and the FAD binding properties ofR279A hDAAO would be more similar to those of pkDAAO (Kd=0.2 μM) where a tyrosine is present at position 279 and H-bondingdoes not form with Pro41 [33].

3.4. Subcellular localization of EYFP-hDAAO variants and their effect onD-serine level in transfected U87 cells

The physiological effect of the investigated substitutions in hDAAOwas studied on U87 human glioblastoma cells stably expressing D31Hor R279A EYFP-hDAAOvariants by assessing the subcellular localization

Fig. 4. Time course of G331V EYFP-hDAAO variant subcellular distribution upon transienttransfected with the wild-type chimeric flavoprotein or with the G331V variant (yellow)and ubiquitin signal (red) patterns. At 24 h after transfection, the wild-type EYFP-hDAAO piant chimeric protein displayed a heterogeneous distribution. In ≈40% of the G331V EYFP-htwo-thirds of them EYFP fluorescence was partially diffused in the cytoplasm and also conceIn the remaining cells, an increased intensity of ubiquitin signal and an increased signal sizoverlapped (panel 2). At 48 h from transfection, the wild-type EYFP-hDAAO (panels on thethe cells expressing G331V EYFP-hDAAO the presence of large and dense protein aggregatesphology (panel 1), even though the peroxisomal marker distribution indicated an altered orand large and compact signals corresponding to PMP70 and ubiquitin, suggesting that the cespots) was observed. EYFP=yellow channel; PMP70=blue channel; ubiquitin=red chann

of the flavoenzyme variants and bymeasuring cellular D-serine concen-trations. The subcellular distribution of D31H and R279A EYFP-hDAAOvariants in transiently transfectedU87 human glioblastoma cells closelyresembles that observed forwild-type hDAAO: these variants largely lo-calize to peroxisomes (Fig. 3). The time course of compartmentalizationfor wild-type and D31H EYFP-hDAAOs shows that the signal is mainlycytosolic (70–90%) at 24 h from transfection and largely compartmen-talized (70–85%) to peroxisomes at 48–72 h, as indicated by the overlapwith the signal for PMP70, a marker of peroxisomes; see also [21]. Asimilar behavior was observed for the R279A hDAAO variant, the onlyexception being that a comparatively higher amount of diffused signalwas apparent at 48 h (≈50% vs. 15–30% forwild-type or D31H variant).In contrast, a more complex situation was apparent for U87 cells tran-siently expressing the EYFP-hDAAO-G331V (Fig. 4). In this case proteinlocalization with respect to ubiquitin was also investigated since a pro-nounced propensity of the variant hDAAO to form dense aggregateswithin the cells, altering cell viability, was observed. At 24 h from trans-fection, when the majority of cells expressing wild-type EYFP-hDAAO(≈73%) show a largely cytosolic EYFP signal distinct from the one asso-ciated with the peroxisomal marker PMP70, ubiquitin signal pattern

transfection in U87 cells. The cellular distribution of EYFP-hDAAO signal in the cellswas monitored by confocal analyses and compared with PMP70 (peroxisomes, blue)rotein (panels on the left) showed the expected, largely cytosolic signal, while the var-DAAO transfected cells, the formation of aggregates of different sizes was observed: forntrated in small aggregates which did not colocalize with PMP70 or ubiquitin (panel 1).e for G331V EYFP-hDAAO was apparent; PMP70, ubiquitin, and EYFP signals partiallyleft) showed a partially compartmentalized pattern, while for a large part (≈80%) ofthrough the cells is evident. Half of these cells conserved a nearly normal cellular mor-ganization of this cellular compartment; the rest of the cells showed a spherical shapells were not viable (panel 2). In both cases an extensive colocalization of signals (whiteel. Bar=10 μm.

Page 9: Biochimica et Biophysica Acta - COnnecting REpositories · 2017. 1. 6. · Characterization of human DAAO variants potentially related to an increased risk of schizophrenia Laura

Fig. 5. Caspase activity detection (panel A) in U87-transfected cells expressing wild-typeand G331V EYFP-hDAAOs (panel B). A) Caspase activity was assayed at 24 and 48 h aftertransient transfection ofwild-type or G331V EYFP-hDAAO and compared to that detectedin control cells (nontransfected or EYFP-transfected U87 cells). The protease activity wasmeasured as relative light units (RLU) by lysing the cells in the presence of Caspase-Glo 3/7 substrate. A significant increase in the detected valueswas observed in the cells express-ing the G331V EYFP-hDAAO at 48 h after transfection. Black bars=wild-typeEYFP-hDAAO transfected cells; white bars=G331V EYFP-hDAAO-transfected cells; darkgray bars=EYFP-transfected cells; light gray bars=nontransfected U87 cells. The valuesare reported as the mean±standard deviation. *p=0.001. B) Western blot analysis ofsamples used in panel A; 20 μg of total proteins were loaded in each lane.

408 L. Caldinelli et al. / Biochimica et Biophysica Acta 1832 (2013) 400–410

(and intensity) resembles that observed in nontransfected, controlU87 cells. On the other hand, ≈45% of cells expressing theEYFP-hDAAO-G331V show a cytosolic, diffused fluorescent signalfor hDAAO (similar to that observed for wild-type hDAAO), 42% ofcells forming cytosolic protein aggregates and the remaining cellsseemingly nonviable. In ≈70% of cells displaying EYFP-hDAAO-G331Vaggregates, the EYFP signal does not overlapwith the signal for the per-oxisomal marker PMP70 (panel 1 at 24 h, Fig. 4), whereas in theremaining cells a partial superimposition of the signal correspondingto EYFP, PMP70, and ubiquitin is evident (white regions in mergepanel 2 at 24 h, Fig. 4). At 48 h from transfection, the cells expressingwild-type EYFP-hDAAO show a punctuate distribution pattern for theflavoenzyme largely overlapping with the signal for PMP70 (Fig. 4,panel at 48 h). In transfected cells, ubiquitin signal intensity is signifi-cantly higher than in nontransfected ones and colocalizes with that ofEYFP-hDAAO variant aggregates. On the other hand, large – 0.5 μm orgreater in diameter – protein aggregates are present in most of thecells expressing the EYFP-hDAAO-G331V. In≈50% of these transfectedcells, the aggregates partially overlap with PMP70 signal distribution(panel 1 at 48 h, Fig. 4): importantly, peroxisomes assume an altered(fused)morphology in the same cells. The remaining 50% of transfectedcells show, in addition, an abnormal cellmorphology (round shape) andalteration of peroxisome morphology: an extended colocalization ofG331V hDAAO, PMP70, and ubiquitin signals is apparent (white regionsin merge panel 2 at 48 h, Fig. 4).

The protein aggregation due to the expression of the G331V hDAAOvariant affects cell viability. To investigate whether the caspase 3/7 path-way is involved in inducing apoptosis, a Caspase-Glo 3/7 assay wasperformed on U87-transfected cells. At 24 h from transient transfection,caspase activity is similar between control (nontransfected or transfectedwith the pEYFP plasmid) cells and the ones expressing wild-type orG331V hDAAO variants, while at 48 h a statistically significant increasein caspase activity in cells expressing the G331V hDAAO is apparent(Fig. 5A). This change is all the more conspicuous considering that at48 h the level of G331V hDAAO is significantly lower (≈10-fold) thanfor the wild-type counterpart (Fig. 5B). This result is not due to differ-ences in transfection yieldswith different constructs because no variationin this parameter was observed by using the NanoLuc luciferase as a ge-netic reporter (Nano-Glo Luciferase Assay System, Promega).

U87 glioblastoma clones stably expressing EYFP-hDAAO-D31H or-R279A proteins were isolated based on the fluorescence signal ofEYFP and then confirmed by Western blot and DAAO activity analyses(Fig. 6). The expression level for D31H variant resembled that ofwild-type hDAAO, while a ≈30% higher expression was apparent forthe R279A variant. On the other hand, it was not feasible to select stableclones expressing the G331V hDAAO variant because this affected cellviability (see also above): no viable cells were found 7 days after trans-fection. The cellular concentration of D- and L-serine in U87 stablytransfected cells was assessed by HPLC chromatography [19,30].EYFP-hDAAO-D31H and -R279A protein expression was associatedwith a statistically significantly higher decrease in cellular D/(D+L)serine ratio than for the wild-type hDAAO in stably transfected U87human glioblastoma cells (Fig. 7).

4. Discussion

4.1. G331V substitution

The in vitro and cellular experiments reported in the present studydemonstrate that substitution of Gly331 with valine (SNP rs4262766)altered the solubility of hDAAOwhile the specific activity of this enzymevariant did not differ from that of the wild-type counterpart. Gly331 islocated on the C-terminal α-helix (Fig. 1): substituting it by a valinemight affect the helix structure and produce a hydrophobic surfacethat promotes protein aggregation in the folded protein or during thefolding process since C-terminal α-helix is exposed in the folding

intermediate(s) of yeast DAAO [26]. Accordingly, only a very lowlevel of soluble G331V hDAAO was obtained in E. coli cells(≤0.1 mg/L of fermentation broth), and human U87 glioblastomacells stably expressing this hDAAO variant were not maintained inculture because of a low viability, while transiently transfected U87cells showed a significant increase in aggregate-containing cells. Indeed,the G331V hDAAO variant is not efficiently targeted to peroxisomes andbecause of its ability to oxidize cytosolic D-serine – and produce hydro-gen peroxide – it might be cytotoxic. Both the low solubility and the cy-tosolic activity of G331V hDAAO could be responsible for this lowviability of U87 transfected cells, as suggested by the increase in caspaseactivity at 48 h from transfection. Importantly, even the R199W hDAAOsubstitution associated with classical adult-onset familial amyotrophiclateral sclerosis reduced cell viability: cells expressing this DAAO variantshowed abnormal shape features and the presence of ubiquitinated pro-tein aggregates [34]. It was suggested that protein aggregation (inmotorneurons) might play a main role in initiating disease in familial cases.

4.2. D31H and R279A substitutions

Thermal stability, binding affinity for inhibitors (benzoate andchlorpromazine), dimeric oligomerization, and interaction with thebinding partner pLG72 were not altered significantly in the D31H

Page 10: Biochimica et Biophysica Acta - COnnecting REpositories · 2017. 1. 6. · Characterization of human DAAO variants potentially related to an increased risk of schizophrenia Laura

Fig. 6. A) Confocal analysis of the subcellular localization of EYFP-hDAAO-D31H and -R279A variants in stably transfected U87 human glioblastoma cells. The distribution of theyellow fluorescent fusion protein (EYFP-hDAAO) is merged with the distribution of the peroxisomal marker PMP70 (red channel). Scale bars=5 μm. B) Western blot analysisperformed using rabbit anti-hDAAO antibodies confirms expression of EYFP-hDAAO wild-type, -D31H, or -R279A variants in stably transfected U87 cells (top panel). The sameamount of sample (5×104 cells) was loaded in each lane, as confirmed by using an anti-α-actin antibody as internal control (bottom panel). C) DAAO activity in U87 cells stablytransfected with EYFP-hDAAO wild-type, -D31H, or -R279A variants determined by Amplex UltraRed assay. The values are expressed as a percentage, taking as 100% the DAAOactivity in U87 cells stably transfected with EYFP-hDAAO wild-type.

Fig. 7. Comparison of D/(D+L) serine ratio (%) in U87 cells not transfected (3.86±0.54%,means±S.E.) and stably transfected with EYFP-hDAAO wild-type (1.29±0.28%), -D31H(1.01±0.27%) or -R279A variants (0.92±0.21%). The change in D/(D+L) serine ratiowith respect to theU87 cells stably transfectedwithwild-type EYFP-hDAAO is statisticallysignificant for U87-EYFP-hDAAO-D31H (*p=0.014) and for U87-EYFP-hDAAO-R279A(**p=0.0008).

409L. Caldinelli et al. / Biochimica et Biophysica Acta 1832 (2013) 400–410

and R279A hDAAO variants, whereas the kinetic efficiency and the af-finity for both the physiological substrate D-serine and the FAD cofac-tor were higher than for the wild-type enzyme (Tables 1, 2). Whilethese effects for the substitution at position 31 can be rationalizedfrom a structural point of view in terms of general, long-range inter-actions that affect protein conformation, we suggest that the R279Amutation might affect the FAD binding affinity in hDAAO by alteringthe structure of the hDAAO SAP region directly through the loss ofthe H-bond to Pro41, or indirectly through a general rearrangementof the β12–α2 interface region.

In U87-transfected cells both D31H and R279A hDAAO variantsare delivered to peroxisomes (as they colocalize with the peroxisomalmarker PMP70) in both transiently and stably transfected cells. More-over, the expression of EYFP-hDAAO-D31H and -R279A proteins pro-duced a higher decrease in the cellular D/(D+L) serine ratio than thewild-type hDAAO in the same U87 cells. These results suggest thatD31H and R279A hDAAO variants more efficiently oxidize D-serine(as compared to the wild-type counterpart) and their expressioncould affect cellular D-serine concentrations and its release at synapsis:this evidence substantiate DAAO's role in the onset of schizophrenia. Infact, the use of DAAO inhibitors as well as the expression of a hypo-functional DAAO in animal models [35,36], produced modest increases

Page 11: Biochimica et Biophysica Acta - COnnecting REpositories · 2017. 1. 6. · Characterization of human DAAO variants potentially related to an increased risk of schizophrenia Laura

410 L. Caldinelli et al. / Biochimica et Biophysica Acta 1832 (2013) 400–410

in brain D-serine levels but successfully reversed behavioral deficits in-duced by NMDAr antagonism. Our evidence supports the hypothesisthat moderate increases in hDAAO activity can affect the neurotrans-mitter level and mediate schizophrenia susceptibility.

5. Conclusions

In order to elucidate the role of D-serine in NMDAr-mediated neuro-transmission, the function of the enzymes involved in D-serine synthe-sis and degradation needs to be defined. While modulation of SRactivity by post-transcriptional modifications and binding partners iswell established [6,8], less is known about the D-serine catabolic en-zyme hDAAO. Up to now, hDAAO variants directly related to schizo-phrenia have not been identified: for this reason it is of greatimportance to biochemically characterize the hDAAO variants that arereported in protein or SNP databases. From the disease pathologicalstandpoint, it is also of utmost importance to elucidate reported substi-tutions on hDAAO function in order to validate the hypothesis that ahypofunction of NMDAr due to D-serine depletion – as the result of anexcess of hDAAO activity – enhances schizophrenia susceptibility.

Acknowledgements

This work was supported by grants from Fondo di Ateneo per laRicerca to L. Pollegioni, S. Sacchi, and G. Molla. We thank ConsorzioInteruniversitario per le Biotecnologie (CIB) and Centro di Ricerca inBiotecnologie per la Salute Umana for their support. We also aregrateful for the technical help of Alessio Barcellini and Matteo EmilioPagliani.

Appendix A. Supplementary data

Supplementary data to this article can be found online at http://dx.doi.org/10.1016/j.bbadis.2012.11.019.

References

[1] L. Pollegioni, L. Piubelli, S. Sacchi, M.S. Pilone, G. Molla, Physiological functions ofD-amino acid oxidases: from yeast to humans, Cell. Mol. Life Sci. 64 (2007)1373–1394.

[2] J.P. Mothet, A.T. Parent, H. Wolosker, R.O. Brady Jr., D.J. Linden, C.D. Ferris, M.A.Rogawski, S.H. Snyder, D-Serine is an endogenous ligand for the glycine site of theN-methyl-D-aspartate receptor, Proc. Natl. Acad. Sci. U. S. A. 97 (2000) 4926–4931.

[3] A.K. Mustafa, P.M. Kim, S.H. Snyder, D-Serine as a putative glial neurotransmitter,Neuron Glia Biol. 1 (2004) 275–281.

[4] M.J. Schell, TheN-methyl D-aspartate receptor glycine site and D-serinemetabolism: anevolutionary perspective, Philos. Trans. R. Soc. Lond. B Biol. Sci. 359 (2004) 943–964.

[5] M. Martineau, G. Baux, J.P. Mothet, D-Serine signalling in the brain: friend and foe,Trends Neurosci. 29 (2006) 481–491.

[6] L. Pollegioni, S. Sacchi, Metabolism of the neuromodulator D-serine, Cell. Mol. LifeSci. 67 (2010) 2387–2404.

[7] P. Fossat, F.R. Turpin, S. Sacchi, J. Dulong, T. Shi, J.M. Rivet, J.V. Sweedler, L.Pollegioni, M.J. Millan, S.H. Oliet, J.P. Mothet, Glial D-serine gates NMDA receptorsat excitatory synapses in prefrontal cortex, Cereb. Cortex 22 (2012) 595–606.

[8] F. Baumgart, I. Rodríguez-Crespo, D-Amino acids in the brain: the biochemistry ofbrain serine racemase, FEBS J. 275 (2008) 3538–3545.

[9] H. Wolosker, Serine racemase and the serine shuttle between neurons and astro-cytes, Biochim. Biophys. Acta 1814 (2011) 1558–1566.

[10] A. Panatier, D.T. Theodosis, J.P. Mothet, B. Touquet, L. Pollegioni, D.A. Poulain, S.H.Oliet, Glia derived D-serine controls NMDA receptor activity and synapticmemory,Cell 125 (2006) 775–784.

[11] E. Kartvelishvily, M. Shleper, L. Balan, E. Dumin, H. Wolosker, Neuron-derivedD-serine release provides a novel means to activate N-methyl-D-aspartate receptors,J. Biol. Chem. 281 (2006) 14151–14162.

[12] E.C. Gustafson, E.R. Stevens, H. Wolosker, R.F. Miller, Endogenous D-serine con-tributes to NMDA-receptor-mediated light-evoked responses in the vertebrateretina, J. Neurophysiol. 98 (2007) 122–130.

[13] S.H. Oliet, J.P. Mothet, Regulation of N-methyl-D-aspartate receptors by astrocyticD-serine, Neuroscience 158 (2009) 275–283.

[14] K. Hashimoto, T. Fukushima, E. Shimizu, N. Komatsu, H. Watanabe, N. Shinoda, M.Nakazato, C. Kumakiri, S. Okada, H. Hasegawa, K. Imai, M. Iyo, Decreased serumlevels of D-serine in patients with schizophrenia: evidence in support of theN-methyl-D-aspartate receptor hypofunction hypothesis of schizophrenia, Arch.Gen. Psychiatry 60 (2003) 572–576.

[15] I. Bendikov, C. Nadri, S. Amar, R. Panizzutti, J. De Miranda, H. Wolosker, G. Agam, ACSF and postmortem brain study of D-serinemetabolic parameters in schizophrenia,Schizophr. Res. 90 (2007) 41–51.

[16] U. Heresco-Levy, Glutamatergic neurotransmission modulators as emerging newdrugs for schizophrenia, Expert Opin. Emerg. Drugs 10 (2005) 827–844.

[17] T.R. Insel, Rethinking schizophrenia, Nature 468 (2010) 187–193.[18] I. Chumakov, M. Blumenfeld, O. Guerassimenko, L. Cavarec, M. Palicio, H.

Abderrahim, L. Bougueleret, C. Barry, H. Tanaka, P. La Rosa, A. Puech, N. Tahri, A.Cohen-Akenine, S. Delabrosse, S. Lissarrague, F.P. Picard, K. Maurice, L. Essioux,P. Millasseau, P. Grel, V. Debailleul, A.M. Simon, D. Caterina, I. Dufaure, K.Malekzadeh, M. Belova, J.J. Luan, M. Bouillot, J.L. Sambucy, G. Primas, M.Saumier, N. Boubkiri, S. Martin-Saumier, M. Nasroune, H. Peixoto, A. Delaye, V.Pinchot, M. Bastucci, S. Guillou, M. Chevillon, R. Sainz-Fuertes, S. Meguenni, J.Aurich-Costa, D. Cherif, A. Gimalac, C. Van Duijn, D. Gauvreau, G. Ouellette, I.Fortier, J. Raelson, T. Sherbatich, N. Riazanskaia, E. Rogaev, P. Raeymaekers, J.Aerssens, F. Konings, W. Luyten, F. Macciardi, P.C. Sham, R.E. Straub, D.R.Weinberger, N. Cohen, D. Cohen, Genetic and physiological data implicating thenew human gene G72 and the gene for D-amino acid oxidase in schizophrenia,Proc. Natl. Acad. Sci. U. S. A. 99 (2002) 13675–13680.

[19] S. Sacchi, M. Bernasconi, M. Martineau, J.P. Mothet, M. Ruzzene, M.S. Pilone, L.Pollegioni, G. Molla, pLG72 modulates intracellular D-serine levels through itsinteraction with D-amino acid oxidase: effect on schizophrenia susceptibility,J. Biol. Chem. 283 (2008) 22244–22256.

[20] L. Caldinelli, G. Molla, L. Bracci, B. Lelli, S. Pileri, P. Cappelletti, S. Sacchi, L.Pollegioni, Effect of ligand binding on human D-amino acid oxidase: implicationsfor the development of new drugs for schizophrenia treatment, Protein Sci. 19(2010) 1500–1512.

[21] S. Sacchi, P. Cappelletti, S. Giovannardi, L. Pollegioni, Evidence for the interactionof D-amino acid oxidase with pLG72 in a glial cell line, Mol. Cell. Neurosci. 48(2011) 20–28.

[22] V. Labrie, W. Wang, S.W. Barger, G.B. Baker, J.C. Roder, Genetic loss of D-aminoacid oxidase activity reverses schizophrenia-like phenotypes in mice, GenesBrain Behav. 9 (2010) 11–25.

[23] G.Molla, D. Porrini, V. Job, L.Motteran, C. Vegezzi, S. Campaner,M.S. Pilone, L. Pollegioni,Role of arginine 285 in the active site of Rhodotorula gracilis D-amino acid oxidase. Asite-directed mutagenesis study, J. Biol. Chem. 275 (2000) 24715–24721.

[24] T. Kawazoe, H. Tsuge, M.S. Pilone, K. Fukui, Crystal structure of human D-aminoacid oxidase: context-dependent variability of the backbone conformation ofthe VAAGL hydrophobic stretch located at the si-face of the flavin ring, ProteinSci. 15 (2006) 2708–2717.

[25] K.Momoi, K. Fukui, F.Watanabe, Y.Miyake,Molecular cloning and sequence analysis ofcDNA encoding human kidney D-amino acid oxidase, FEBS Lett. 238 (1988) 180–184.

[26] L. Caldinelli, S. Iametti, A. Barbiroli, F. Bonomi, L. Piubelli, P. Ferranti, G. Picariello,M.S. Pilone, L. Pollegioni, Unfolding intermediate in the peroxisomal flavoproteinD-amino acid oxidase, J. Biol. Chem. 279 (2004) 28426–28434.

[27] G.Molla, S. Sacchi, M. Bernasconi, M.S. Pilone, K. Fukui, L. Pollegioni, Characterizationof human D-amino acid oxidase, FEBS Lett. 580 (2006) 2358–2364.

[28] G. Molla, M. Bernasconi, S. Sacchi, M.S. Pilone, L. Pollegioni, Expression in Escherichiacoli and in vitro refolding of the human protein pLG72, Protein Expr. Purif. 46 (2006)150–155.

[29] R.A. Stinson, J.J. Holbrook, Equilibrium binding of nicotinamide nucleotides tolactate dehydrogenases, Biochem. J. 131 (1973) 719–728.

[30] L. Caldinelli, G. Molla, S. Sacchi, M.S. Pilone, L. Pollegioni, Relevance of weak flavinbinding in human D-amino acid oxidase, Protein Sci. 18 (2009) 801–810.

[31] F. Volontè, F. Marinelli, L. Gastaldo, S. Sacchi, M.S. Pilone, L. Pollegioni, G. Molla,Optimization of glutaryl-7-aminocephalosporanic acid acylase expression in E.coli, Protein Expr. Purif. 61 (2008) 131–137.

[32] F. Todone, M.A. Vanoni, A. Mozzarelli, M. Bolognesi, A. Coda, B. Curti, A.Mattevi, Active site plasticity in D-amino acid oxidase: a crystallographicanalysis, Biochemistry 36 (1997) 5853–5860.

[33] A. Mattevi, M.A. Vanoni, F. Todone, M. Rizzi, A. Teplyakov, A. Coda, M. Bolognesi,B. Curti, Crystal structure of D-amino acid oxidase: a case of active sitemirror-image convergent evolution with flavocytochrome b2, Proc. Natl. Acad.Sci. U. S. A. 93 (1996) 7496–7501.

[34] J. Mitchell, P. Paul, H.J. Chen, A.Morris, M. Payling,M. Falchi, J. Habgood, S. Panoutsou, S.Winkler, V. Tisato, A. Hajitou, B. Smith, C. Vance, C. Shaw, N.D. Mazarakis, J. deBelleroche, Familial amyotrophic lateral sclerosis is associated with a mutation inD-amino acid oxidase, Proc. Natl. Acad. Sci. U. S. A. 107 (2010) 7556–7561.

[35] S. Sacchi, E. Rosini, L. Pollegioni, G. Molla, D-Amino acid oxidase as a novel class ofdrug for schizophrenia therapy, Curr. Pharm. Des. 19 (2013), (Ahead of print).

[36] V. Labrie, A.H. Wong, J.C. Roder, Contributions of the D-serine pathway to schizo-phrenia, Neuropharmacology 62 (2012) 1484–1503.