Alterasi Pada Metabolisme, Peran Protein Dan Fungsi Sinapt

18
Proteomic analysis of parkin knockout mice: alterations in energy metabolism, protein handling and synaptic function Magali Periquet, Olga Corti, Sandrine Jacquier and Alexis Brice INSERM U679, Ho ˆpital de la Salpe ˆtrie `re, AP-HP, 75013 Paris, France Abstract Parkin knockout (KO) mice show behavioural and biochemical changes that reproduce some of the presymptomatic aspects of Parkinson’s disease, in the absence of neuronal degeneration. To provide insight into the pathogenic mechanisms underlying the preclinical stages of parkin-related parkinsonism, we sear- ched for possible changes in the brain proteome of parkin KO mice by means of fluorescence two-dimensional difference gel electrophoresis and mass spectrometry. We identified 87 pro- teins that differed in abundance between wild-type and parkin KO mice by at least 45%. A high proportion of these proteins were related to energy metabolism. The levels of several pro- teins involved in detoxification, stress-related chaperones and components of the ubiquitin–proteasome pathway were also altered. These differences might reflect adaptive mechanisms aimed at compensating for the presence of reactive oxygen species and the accumulation of damaged proteins in parkin KO mice. Furthermore, the up-regulation of several members of the membrane-associated guanylate kinase family of synaptic scaffold proteins and several septins, including the Parkin substrate cell division control related protein 1 (CDCRel-1), may contribute to the abnormalities in neurotransmitter release previously observed in parkin KO mice. This study provides clues into possible compensatory mechanisms that protect dopaminergic neurones from death in parkin KO mice and may help us understand the preclinical deficits observed in parkin- related parkinsonism. Keywords: knockout mice, parkin, proteomic, two-dimen- sional fluorescence difference gel electrophoresis. J. Neurochem. (2005) 95, 1259–1276. Parkinson’s disease (PD) is a common neurodegenerative disorder clinically characterized by resting tremor, rigidity and bradykinesia. These severe neurological symptoms are caused by the selective, progressive degeneration of dopaminergic neurones in the substantia nigra pars compacta. Lewy bodies – ubiquitylated neuronal cytoplasmic inclusions – are a patho- logical hallmark of PD (Forno 1987). Seven genes involved in rare monogenic forms of PD have been discovered in the past 8 years (Dekker et al. 2003). Missense mutations in the a-synuclein gene, multiplications of a genomic region inclu- ding this gene (Singleton et al. 2003), as well as missense mutations in the recently discovered dardarin gene (Paisan- Ruiz et al. 2004; Zimprich et al. 2004) cause autosomal dominant forms of parkinsonism. The ubiquitin carboxyter- minal hydrolase L1 (UCH-L1) and Nurr-1 genes are also potentially involved in autosomal dominant parkinsonian syndromes. However, their role in the pathogenesis of the disease remains uncertain, because the corresponding muta- tions have been found in only a small number of families. In addition, the parkin, DJ-1 and PTEN-induced kinase 1 (PINK1) genes are responsible for autosomal recessive forms of the disease (Dekker et al. 2003; Valente et al. 2004). In 1998, the parkin gene was shown to be responsible for a distinct clinical and genetic entity in Japan, defined as autosomal recessive juvenile parkinsonism (Kitada et al. 1998). A series of parkin exon rearrangements and point mutations have since been identified in almost 50% of patients with familial autosomal recessive early-onset parkinsonism from different populations, and in 15% of non-familial cases (Lu ¨ cking et al. 2000; Lohmann et al. 2003; Periquet et al. Received March 1, 2005; revised manuscript received May 10, 2005; accepted July 18, 2005. Address correspndence and reprint requests to Alexis Brice, INSERM U679, Ho ˆpital de la Salpe ˆtrie `re, 47 Boulevard de l’Ho ˆpital, 75651 Paris Cedex 13, France. E-mail: [email protected] Abbreviations used: AcCN, acetonitrile; CDCRel-1, cell division control related protein 1; 2D DIGE, two-dimensional difference gel electrophoresis; DTT, dithiothreitol; GTP2, glutathione S-transferase P2; IPG, immobilized pH gradient; KO, knockout; MAGUK, membrane- associated guanylate kinase; MALDI–TOF, matrix-assisted laser desorption/ionization–time of flight; MS, mass spectrometry; NSF, N-ethylmaleimide sensitive fusion protein; OTUB1, OTU-domain uba1- binding protein; PD, Parkinson’s disease; PINK1, PTEN-induced kinase 1; UCH-L1, ubiquitin carboxyterminal hydrolase L1; WT, wild type. Journal of Neurochemistry , 2005, 95, 1259–1276 doi:10.1111/j.1471-4159.2005.03442.x ȑ 2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 1259–1276 1259

description

SIIP

Transcript of Alterasi Pada Metabolisme, Peran Protein Dan Fungsi Sinapt

  • Proteomic analysis of parkin knockout mice: alterations inenergy metabolism, protein handling and synaptic function

    Magali Periquet, Olga Corti, Sandrine Jacquier and Alexis Brice

    INSERM U679, Hopital de la Salpetrie`re, AP-HP, 75013 Paris, France

    Abstract

    Parkin knockout (KO) mice show behavioural and biochemical

    changes that reproduce some of the presymptomatic aspects of

    Parkinsons disease, in the absence of neuronal degeneration.

    To provide insight into the pathogenic mechanisms underlying

    the preclinical stages of parkin-related parkinsonism, we sear-

    ched for possible changes in the brain proteome of parkin KO

    mice by means of fluorescence two-dimensional difference gel

    electrophoresis and mass spectrometry. We identified 87 pro-

    teins that differed in abundance between wild-type and parkin

    KO mice by at least 45%. A high proportion of these proteins

    were related to energy metabolism. The levels of several pro-

    teins involved in detoxification, stress-related chaperones and

    components of the ubiquitinproteasome pathway were also

    altered. These differences might reflect adaptive mechanisms

    aimed at compensating for the presence of reactive oxygen

    species and the accumulation of damaged proteins inparkinKO

    mice. Furthermore, the up-regulation of several members of the

    membrane-associated guanylate kinase family of synaptic

    scaffold proteins and several septins, including the Parkin

    substrate cell division control related protein 1 (CDCRel-1), may

    contribute to the abnormalities in neurotransmitter release

    previously observed in parkin KO mice. This study provides

    clues into possible compensatory mechanisms that protect

    dopaminergic neurones from death in parkin KO mice and may

    help us understand the preclinical deficits observed in parkin-

    related parkinsonism.

    Keywords: knockout mice, parkin, proteomic, two-dimen-

    sional fluorescence difference gel electrophoresis.

    J. Neurochem. (2005) 95, 12591276.

    Parkinsons disease (PD) is a common neurodegenerativedisorder clinically characterized by resting tremor, rigidity andbradykinesia. These severe neurological symptoms are causedby the selective, progressive degeneration of dopaminergicneurones in the substantia nigra pars compacta. Lewy bodies ubiquitylated neuronal cytoplasmic inclusions are a patho-logical hallmark of PD (Forno 1987). Seven genes involved inrare monogenic forms of PD have been discovered in the past8 years (Dekker et al. 2003). Missense mutations in thea-synuclein gene, multiplications of a genomic region inclu-ding this gene (Singleton et al. 2003), as well as missensemutations in the recently discovered dardarin gene (Paisan-Ruiz et al. 2004; Zimprich et al. 2004) cause autosomaldominant forms of parkinsonism. The ubiquitin carboxyter-minal hydrolase L1 (UCH-L1) and Nurr-1 genes are alsopotentially involved in autosomal dominant parkinsoniansyndromes. However, their role in the pathogenesis of thedisease remains uncertain, because the corresponding muta-tions have been found in only a small number of families. Inaddition, the parkin, DJ-1 and PTEN-induced kinase 1(PINK1) genes are responsible for autosomal recessive formsof the disease (Dekker et al. 2003; Valente et al. 2004).

    In 1998, the parkin gene was shown to be responsible for adistinct clinical and genetic entity in Japan, dened asautosomal recessive juvenile parkinsonism (Kitada et al.1998). A series of parkin exon rearrangements and pointmutations have since been identied in almost 50% of patientswith familial autosomal recessive early-onset parkinsonismfrom different populations, and in 15% of non-familial cases(Lucking et al. 2000; Lohmann et al. 2003; Periquet et al.

    Received March 1, 2005; revised manuscript received May 10, 2005;accepted July 18, 2005.Address correspndence and reprint requests to Alexis Brice, INSERM

    U679, Hopital de la Salpetrie`re, 47 Boulevard de lHopital, 75651 ParisCedex 13, France. E-mail: [email protected] used: AcCN, acetonitrile; CDCRel-1, cell division

    control related protein 1; 2D DIGE, two-dimensional difference gelelectrophoresis; DTT, dithiothreitol; GTP2, glutathione S-transferase P2;IPG, immobilized pH gradient; KO, knockout; MAGUK, membrane-associated guanylate kinase; MALDITOF, matrix-assisted laserdesorption/ionizationtime of ight; MS, mass spectrometry; NSF,N-ethylmaleimide sensitive fusion protein; OTUB1, OTU-domain uba1-binding protein; PD, Parkinsons disease;PINK1, PTEN-induced kinase 1;UCH-L1, ubiquitin carboxyterminal hydrolase L1; WT, wild type.

    Journal of Neurochemistry, 2005, 95, 12591276 doi:10.1111/j.1471-4159.2005.03442.x

    2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 12591276 1259

  • 2003). These mutations are associated with a wide range ofages at onset and a broad phenotypic spectrum, includingcases clinically indistinguishable from idiopathic PD (Kleinet al. 2000; Lucking et al. 2000). Neuropathological descrip-tions of brains from patients with homozygous parkindeletions have reported specic dopaminergic neuronaldegeneration in the substantia nigra pars compacta in theabsence of Lewybodies (Mori et al. 1998; Hayashi et al.2000; van de Warrenburg et al. 2001; Gouider-Khouja et al.2003). However, Lewy body pathology and/or tau depositshave been described in a few individuals with compoundheterozygous mutations (Mori et al. 1998; Farrer et al. 2001).Parkin, a 465-amino acid protein with a ubiquitin-like

    domain at its N-terminus and a C-terminal cysteine-richRING-IBR-RING motif, has E3 ubiquitinprotein ligaseactivity that promotes the ubiquitylation and proteasomaldegradation of specic protein substrates (Imai et al. 2000;Shimura et al. 2000; Zhang et al. 2000; Dev et al. 2003).Loss of Parkin function is thought to result in the progressiveaccumulation of non-ubiquitylated, potentially toxic sub-strates, leading to neurodegeneration. At least 10 Parkinsubstrates have been identied so far with roles in cellprocesses as diverse as cell signalling (Pael-R), cell cyclecontrol (cyclin E), protein biosynthesis (the p38 scaffoldsubunit of the multi-aminoacyl-tRNA synthetase compo-nent), cytoskeletal dynamics (a/b tubulin), and vesicular andsynaptic functions (CDCRel-1 and CDCRel-2, synaptotag-min IX, O-glycosylated a-synuclein, synphilin, the dopaminetransporter) (Hattori and Mizuno 2004; Jiang et al. 2004).However, the specic and potentially synergistic pathologicalroles of these substrates are unclear.We and others have recently generated a parkin knockout

    (KO) mouse model to facilitate investigation of thepathogenic mechanisms underlying parkinsonism due toparkin gene mutations (Goldberg et al. 2003; Itier et al.2003; Von Coelln et al. 2004; Perez and Palmiter 2005).There was no evidence for a loss of nigrostriatal dopam-inergic neurones in these mice, but a number of behaviouraland biochemical changes were observed, including decitsin dopamine handling, reproducing some of the presymp-tomatic aspects of PD (Itier et al. 2003). This model istherefore of value for investigation of the functionalconsequences of parkin gene inactivation, including poten-tial compensatory mechanisms preventing the onset of aparkinsonian phenotype.To shed line on the molecular pathways affected in parkin

    KO mice and identify potential Parkin substrates predicted toaccumulate in these mice, we performed a differential analysisof parkin KO and wild-type (WT) brain proteomes. Two-dimensional uorescence difference gel electrophoresis (2DDIGE)was chosen for this purpose, because it has proven to bevaluable for a number of biological applications, includingstudies related to neurodegenerative disorders such as schizo-phrenia and Huntingtons disease (Zabel et al. 2002; Swatton

    et al. 2004). This technique involves the pre-electrophoreticlabelling of the protein samples to be compared withuorescent dyes, such as Cy2 and Cy5, which allowsstatistically valid quantication of a dynamic range of proteinconcentrations with high sensitivity (Patton 2000); thelabelled protein samples are pooled and mixed with aninternal standard prelabelled with Cy3 to normalize the resultsand reduce gel-to-gel variability. When used with the dedica-ted DeCyder analysis software (Amersham Bioscience Inc.,Amersham, UK), this technique permits the sensitive, massspectrometry (MS)-compatible and reproducible identicationof statistically signicant differences in the protein expressionproles of multiple samples examined simultaneously (Tongeet al. 2001; Gharbi et al. 2002; Yan et al. 2002).We combined this approach with sensitive matrix-assis-

    ted laser desorption/ionizationtime of ight (MALDITOF) MS and tandem MS, to screen six WT and six parkinKO mice at 2 and 12 months, in two brain structures(cortex and striatum). Eighty-seven unique proteins wereidentied that differed in abundance between the brains ofparkin KO and WT mice. These differences provide newinformation concerning the molecular pathways that mightbe involved in the preclinical stages of parkin-relatedparkinsonism.

    Experimental procedures

    Animals and brain tissues

    Studies were carried out on 2- and 12-month-old parkin KO (Itieret al. 2003) and WT mice with a pure 129SV background. Animalswere anaesthetized with sodium pentobarbital (60 mg/kg i.p.) and

    perfused intercardially with saline. Brains were removed, and the

    cortex and striatum tissues were dissected out and rapidly frozen.

    Brain tissue was lyophilized for 48 h.

    Preparation of protein samples

    Lyophilized tissues (10 mg) were resuspended in 80 lL 0.032 MTris-HCl/Tris base containing 1.2% (v/v) Triton X-100. The

    preparations were heated at 100C for 5 min, then homogenized,and 7.5 lL Dnase I and Rnase in 100 mM Pefabloc/100 mM EDTAwas gradually added. The mixture was incubated for 10 min, then

    the proteins were solubilized by adding 105 mg urea, 38 mg

    thiourea and 47 lL 21% CHAPS. Protein samples were kept atroom temperature (25C) for 10 min, gently vortexed, and thencentrifuged for 5 min at 14 000 g, followed by 45 min at100 000 g. Protein extracts were aliquoted and stored at )80C orimmediately run on rst-dimension gels.

    Labelling of protein samples (DIGE)

    Fluorescent dyes were conjugated to solubilized proteins via

    N-hydroxysuccinimidyl linkages, such that 10% of each proteinwas labelled. Typically, 50 lg parkin KO or WT mouse proteinextract was labelled with 400 pmol cyanin dye Cy5 for parkin-KOand Cy2 for WT (Amersham Bioscience Inc.). As 2D-DIGE

    analysis requires large amounts of protein, we used a pool of six

    1260 M. Periquet et al.

    2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 12591276

  • mice to study the striatum proteome. We also pooled cortex samples,

    to make it possible to compare the results obtained in the striatum

    and cortex. A pool of all samples was also prepared and labelled

    with Cy3, for use as an internal standard on all gels. Labeling

    reactions were performed in the dark, at room temperature, for

    30 min and were quenched by incubation with a 50-fold molar

    excess of free lysine over dye for 10 min at room temperature.

    Samples were diluted with an equal volume of solution containing

    7 M urea, 2 M thiourea, 4% CHAPS, 67 mM dithiothreitol (DTT),

    1% Pharmalyte 310 and 0.2% (v/v) Triton X-100.

    2D gel electrophoresis

    The rst dimension of electrophoresis was carried out with narrow

    immobilized pH gradient gels (Immobiline Dry Strip, pH 4.56,

    5.56.7, 69) on a horizontal electrophoresis apparatus (Multiphor

    II; Amersham Pharmacia Biotechnology). Overlapping pH gradients

    were used, to optimize protein resolution (Fig. 1). Immobilized pH

    gradient (IPG) strips (0.5 3 80 mm), containing immobilinesNL 310, were rehydrated in a cassette containing 6 M urea, 2 M

    thiourea, 1% (v/v) CHAPS, 0.5% Pharmalyte 310 and 0.4% DTT.

    Isoelectric focusing was then performed by applying 50 lg labelled

    proteins (for analytical gels) or 500 lg unlabelled proteins (forpreparative gels) to the anodic side. The samples were made to enter

    the IPG strips by applying a low voltage gradient (50 V for 2 h,

    100 V for 2 h, 300 V for 2 h). The gel was then run for 16 h at

    2000 V and 18 h at 3500 V for the pH 4.56 and 5.56.7 gradients,

    and for 1 h at 600 V and then 9 h at 3500 V for the pH 69

    gradient. The strips were then equilibrated by incubating for 10 min

    in 0.1 M Tris-HCl containing 0.5% (v/w) DTT, 36% urea and 30%

    (v/w) glycerol, and then for a further 10 min in a solution of the

    same composition except that DTT was replaced by 4.5%

    iodoacetamide. In the second dimension, strips were subjected to

    13% T, 2.54% C sodium dodecyl sulfatepolyacrylamide gel

    elctrophoresis, using the Iso-Dalt apparatus (Hoeffer, San Francisco,

    CA, USA; T corresponds to the total percentage concentration of

    acrylamide and N,N-methylenebisacrylamide in the gel, and Ccorresponds to the concentration of N,N-methylenebisacrylamideas a percentage (by weight of acrylamide and N,N-methylenebis-acrylamide). Gels were run at 10C, 50 mA for 1.5 h, then at100 mA for 1.5 h and overnight at 185 mA. For each set of

    conditions, we ran three analytical and two preparative gels in

    parallel.

    4.5(a)

    (b)

    200 kDa

    15 kDa

    MW

    pl plpl6 6 96.75.5

    Fig. 1 2D gels images showing the narrow range of overlapping pH

    gradients. (a) Analytical gels. In order to optimize protein resolution,

    the first dimension of electrophoresis was carried out with narrow

    overlapping pH gradient gels (Immobiline Dry Strip, pH 4.56, 5.56.7,

    69). Some 50 lg of each sample was labelled with cyanin dyes and

    loaded on the analytic gels. (b) Preparative gels; 500 lg unlabelled

    proteins were detected by staining with Sypro-Ruby dye after elec-

    trophoresis. The patterns of staining for the cyanin dyes and Sypro-

    Ruby dye were very similar, facilitating accurate matching and picking

    on these preparative gels.

    Proteomic analysis of parkin knockout mice 1261

    2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 12591276

  • Protein visualization

    The Cy2, Cy3 and Cy5 components of each gel were imaged

    individually using mutually exclusive excitation/emission wave-

    lengths on a ProXpress (Perkin Elmer, Norwalk, CT, USA)

    uorescent gel scanner. Gel images were normalized by adjusting

    exposure times according to mean pixel values. Unlabelled proteins

    (preparative gels) were detected by staining with Sypro-Ruby dye

    (Molecular Probes, Eugene, OR, USA) after electrophoresis and

    images were acquired as above.

    Image analysis

    For each condition, pools of six WT and six KO samples were

    labelled with Cy2 and Cy5 respectively, and run on three replicate

    gels together with a Cy3-labelled mixture of all 12 WT and KO

    samples as the in-gel standard. The differential in-gel analysis

    module of the DeCyder software was used to quantify protein spot

    volumes for each in-gel image pair (Cy2Cy3, Cy5Cy3) and

    express the values as a ratio (Cy2/Cy3, Cy5/Cy3). The DeCyder

    biological variation analysis module was subsequently used to

    match the protein spot maps of all replicate gels. This software

    module calculates the average changes in the Cy2/Cy3 and Cy5/Cy3

    ratios across gels and applies statistics (Students t-test) to associatea level of condence with each of those changes. Only the proteins

    presenting variations in the Cy2/Cy3 and Cy5/Cy3 ratios exceeding

    an arbitrary threshold of 1.45, corresponding to a change in

    abundance of 45%, and with a p-value < 0.05, were considered to besignicantly different. Proteins were dened as up-regulated or

    down-regulated if their abundance was higher or lower, respectively,

    in parkin KO mice than in WT mice.

    MS

    As the molecular mass of the labelled protein is 0.5 kDa greaterthan that of the unmodied protein, we labelled the minimum

    number of molecules for each protein and excised the unlabelled

    protein spot rather than the labelled protein spot for mass

    spectrometry. The differences observed in 2D DIGE analyses were

    compared with Sypro-Ruby protein patterns, and spots were selected

    for picking (Ettan spot picker; Amersham Biosciences, Little

    Chalfont, UK) on the basis of this staining pattern. The patterns

    of staining for the cyanin dyes and Sypro-Ruby dye were very

    similar, facilitating accurate matching and picking (Fig. 1).

    Up-regulated proteins were excised from a preparative gel contain-

    ing a KO sample and down-regulated proteins were excised from a

    preparative gel containing a WT sample.

    The proteins were reduced with DTT (Sigma, Poole, UK),

    alkylated with iodoacetamide and digested with trypsin (modied

    trypsin, sequencing grade; Roche, Indianapolis, IN, USA) overnight

    at 37C, using the automatic DIGESTPRO digester from ABIMED(Longenfeld, Germany). Tryptic digests were dried under vacuum in

    a Speed-Vac. Samples were resuspended in 4 lL 0.1% formic acid.A 0.5-lL aliquot of each sample was used to measure automaticallythe mass ngerprint on a Bruker Reex III MALDITOF mass

    spectrometer (Bruker-Daltonix GmbH, Bremen, Germany) in

    positive ion reector mode using delayed extraction. The measured

    tryptic peptide masses were transformed automatically, through the

    MS BioTools program, into input used by Mascot software (Matrix

    Science, London, UK) to search the National Center for Biotech-

    nology Information (NCBI) database.

    To conrm some of the ngerprints, tryptic digests were

    separated by HPLC, using the LC-Packings system (San Francisco,

    CA, USA), including an injector (Famos), a concentrator (Switchos)

    and a pump (Ultimate). The ow rate was adjusted to 200 nL/min. A

    gradient was used, starting at 2% Acetonitrile (AcCN) in 0.1%

    formic acid for 1 min, increased to 50% AcCN over 40 min, and

    nally to 90% AcCN over 10 min. A 1-lL aliquot was injected fromthe autosampler (in user dened program mode) into a

    15 cm 75 lm fused silica column packed with PLRP-S 5 lm(Polymer Laboratories).

    The LC system was connected to an ion trap mass spectrometer

    (LCQ Deca; Finnigan Corp, San Jose, CA, USA), run by Xcalibur

    software. The spray voltage was set at 2.1 kV, the temperature of the

    ion transfer tube was set at 180C and the normalized collisionenergies were set at 35% for MS/MS. We used dynamic exclusion.

    The sequences of the uninterpreted CID spectra were identied by

    correlation with the peptide sequences present in the NCBI non-

    redundant protein database, using the SpectrumMill program

    (Millenium Pharmaceuticals, Cambridge, MA, USA).

    Protein annotation and data handling

    The MALDI-TOF MS and MSMS analyses that followed our 2D

    DIGE approach were successful for about 74% (159 spots) of all

    spots showing altered abundance in parkin KO mice. Owing to post-translational modications and the use of overlapping pH gradients,

    the 159 protein spots identied in both the cortex and the striatum

    from 2- and 12-month-old mice corresponded to 87 different

    proteins. To ensure a high quality of annotation, the proteins

    identied were compared with those in the Swissprot sequence

    database, using the Blast2 algorithm. A relational database

    containing protein annotations and experimental results was created

    (Access; Microsoft Corporation, Redmond, WA, USA) to facilitate

    analysis.

    Quantitative western blot analyses

    CDCRel-1 and calretinin protein levels were analysed in samples from

    the cortex of 2- and 12-month-old mice respectively (n 5 forWTorparkinKOmice).We ran about 40 lg total protein extract in each laneof a 15-well, precast 412% gradient sodium dodecyl sulfate

    polyacrylamide mini gel (Invitrogen, Carlsbad, CA, USA). After

    electrophoresis, the proteins were transferred to nitrocellulose lters

    (Protran, Schleicher & Schuell Bioscience, Dassel, Germany). The

    lter was blocked by incubation in 0.2% Tween and 5% non-fat milk

    powder in phosphate-buffered saline, followed by anti-calretinin

    (1 : 10000; Swant, Bellinzona, Switzerland), anti-CDCRel1

    (1 : 5000) or anti-actin (1 : 2000; gift from Sigma, St Louis, MO,

    USA) antibodies. Secondary antibodies radiolabelled with 125I

    (1 : 200; IM 131 and IM 134, Amersham Biosciences) were visual-

    ized by phosphoimaging and quantied by Aida analysis software

    (Raytest Isotopenmessgeraete GmbH, Straubenhardt, Germany).

    Oxyblot analyses

    Protein carbonyls were assayed by western blot analysis in brains of

    2- and 12-month-old WT and KO mice, according to the

    manufacturers instructions (Oxyblot; Chemicon, Temecula, CA,

    USA). In brief, 15 lg protein from individual cortex and striatumextracts obtained in 50 mM Hepes containing 150 mM NaCl, 10%

    glycerol, 1% Triton X-100, 100 mM NaF, 0.2 mM Na3VO4 and

    1262 M. Periquet et al.

    2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 12591276

  • complete protease inhibitors (Roche) was reacted with 2,4-di-

    nitrophenylhydrazine and western blotted using a primary antibody

    specic to dinitrophenylhydrazone-derivatized residues (Oxyblot;

    Chemicon) and a 125I-labelled secondary antibody (Amersham

    Biosciences) or a non-radioactive secondary antibody (Oxyblot;

    Chemicon). Protein carbonyls were visualized by phosphoimaging

    or revealed using enhanced chemioluminescence and quantied by

    densitometry. Blots were subsequently reprobed for actin immuno-

    reactvity (1 : 2000; Sigma) and revealed using enhanced chemio-

    luminescence (Pierce, Rockford, IL, USA).

    Results

    Analysis of cortical and striatal mouse tissues at 2 and12 months of age using 2D DIGE technology led to theidentication of 159 differentially regulated protein spotsbetween WT and parkin KO mice. In 2D analysis, proteinsare frequently detected in more than one spot, indicating thepresence of either different isoforms and/or post-translationalmodications. In order to optimize protein resolution and tobetter detect these different isoforms, the rst dimension ofelectrophoresis was carried out with narrow overlapping pHgradient gels (Immobiline Dry Strip, pH 4.56, 5.56.7, 69;Fig. 1). The differentially regulated spots were rst analysedby MALDITOF MS, using peptide mass ngerprints anddatabase searches. Proteins not identied by this methodwere subjected to MS/MS, followed by a search of sequencedatabases. With these techniques, we identied 87 uniqueproteins that differed in abundance by at least 45% in WTand parkin KO mice (Tables 13). In 2-month-old mice, thenumber of proteins found to be differentially regulated in thecortex and the striatum and the proportions of up- and down-regulated proteins were similar (Table 1). In contrast, in12-month-old mice, the majority of the proteins with alteredabundance were found in the striatum and most of them wereup-regulated (p < 0.05) (Table 1). Approximately 20% (18of 87) of the identied proteins were differentially regulatedin both 2- and 12-month-old parkin KO mice and a similarpercentage (14/87) were dysregulated in both the cortex andthe striatum, at one or both of the ages examined (Tables 4and 5). Overall, 46 proteins that increased in abundance wereidentied, 31 that decreased in abundance, three that changedtheir pattern of regulation between 2 and 12 months, and

    seven that had a change in their electrophoretic mobility. Themobility variants were represented by at least two independ-ent spots with different isoelectric points of a sameprotein subjected to opposite regulation in one and the sameexperiment (status +/ in Tables 25). They usually corres-pond to different isoforms of a protein that has undergonepost-translational modication, i.e. a shift in phosphorylationstatus, as illustrated in Fig. 2(c). Other examples of changesin staining intensity are illustrated in Fig. 2.Classication of the differentially regulated proteins

    according to the specic keywords attributed to them in theSwissprot database (Table 6) showed that a certain propor-tion of the modulated proteins were linked to energymetabolism (glycolysis, ATP synthesis, avoprotein, hydro-gen ion transport, FAD, NAD/NADP, mitochondrion) andprotein processing pathways (heat shock, proteasome, pro-tein biosynthesis, ligase, chaperone, transit peptide, isom-erase). The frequent occurrence of the keywords kinase andGTP binding suggested that cell signalling pathways werelikely to be disrupted, together with vesicle trafcking andcytoskeletal dynamics processes in which proteins withkinase and GTPase activities play major roles.Extensive literature searches in Pubmed, to characterize

    each protein, led to the identication of 12 distinctfunctional categories, each including at least two proteinsup- or down-regulated in the absence of the parkin gene(Fig. 3, Tables 2 and 3). Sixty-seven of the 87 identiedproteins fell into these categories; nine, represented byseveral spots with either increased or decreased abundance,a situation compatible with post-translational modicationsor a change in the pattern of regulation between 2- and12-month-old mice, were not included in Fig. 3. Elevenproteins could not be assigned to established functionalcategories based on their putative functions. Among theseproteins, the calcium-binding protein calretinin was found.Calretinin, a brain-specic, potentially neuroprotective pro-tein (Mura et al. 2000; Tsuboi et al. 2000), was down-regulated in the cortex of 12-month-old KO mice (Table 3).The down-regulation of this protein was conrmed byquantitative western blot following the one-dimensional gelelectrophoresis of protein samples from mouse cortex(Fig. 4).

    Table 1 Number of proteins that differed

    significantly in abundance in WT and parkin

    KO mice Age (months)

    Cortex Striatum

    Increased Decreased Altered EM Increased Decreased Altered EM

    2 15 12 5 19 18 3

    12 4 4 0 23 5 0

    The total number of matched protein spots that differed in abundance by at least 45% in WT and

    KO samples is shown for 2- and 12-month-old mice. Note that the sum of these proteins differs

    from the total number of 87 identified proteins, because some of them are regulated in two

    structures or at two ages. Fold differences were calculated from the mean standardized abundance

    of triplicate spots. Only means with p values < 0.05 were included. EM, electrophoretic mobility.

    Proteomic analysis of parkin knockout mice 1263

    2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 12591276

  • Table 2 Proteins differentially regulated in the striatum of WT and parkin KO mice

    SwissProt or gi accession Protein name

    No. and status of spots

    Striatum

    2 months

    Striatum

    12 months

    Energy metabolism (n 17)ALFC_MOUSE Fructose-bisphosphate aldolase C [Fragment] 1

    ATPA_MOUSE ATP synthase a chain, mitochondrial 3+ 2+

    CISY_HUMAN Citrate synthase 1 1

    DLDH_MOUSE Dihydrolipoamide dehydrogenase 1+

    DHSA_HUMAN Succinate dehydrogenase flavoprotein subunit 1+

    G3P_MOUSE Glyceraldehyde-3-phosphate dehydrogenase 2+

    KAD3_MOUSE GTP:AMP phosphotransferase mitochondrial 1+ 1+

    KPY2_MOUSE Pyruvate kinase, M2 isozyme 1/2 4

    LDHA_MOUSE L-Lactate dehydrogenase A chain 1

    MDHC_MOUSE Malate dehydrogenase, cytoplasmic 1

    NUBM_HUMAN NADH-ubiquinone oxidoreductase 51-kDa subunit 1+

    ODPB_RAT Pyruvate dehydrogenase E1 component b subunit 1

    PGK1_MOUSE Phosphoglycerate kinase 1 *1

    SCB2_MOUSE Succinyl-CoA ligase [GDP-forming] b-chain, mitochondrial 1+

    THIL_RAT Acetyl-CoA acetyltransferase, mitochondrial 1+

    UCR1_RAT Ubiquinol-cytochrome c reductase complex core protein I 1+

    539926/13435978 Acetyl-CoA C-acetyltransferase 1+

    Signal transduction (n 8)ARK1_RAT b-Adrenergic receptor kinase 1 *1

    CRK_MOUSE Proto-oncogene C-crk 1

    DPY2_MOUSE Dihydropyrimidinase-related protein-2 (CRMP-2) 5/2+ 5+

    P2BA_MOUSE Ser/Thr protein phosphatase 2B catalytic subunit, a isoform 1+

    PP1B_HUMAN Ser/Thr protein phosphatase PP1-b catalytic subunit 1+

    PTNB_MOUSE Protein tyrosine phosphatase, non receptor type 1-1 1

    143Z_MOUSE 14-3-3 prot f/d 1

    MPP3_HUMAN MAGUK p55 subfamily member 3 1+

    Vesicular trafficking (n 8)GDIR_HUMAN rho GDP-dissociation inhibitor 1 2+

    NSF_MOUSE N-ethylmaleimide-sensitive fusion protein 1- 1+

    ST1B_MOUSE Syntaxin 1B 1

    STB1_MOUSE Syntaxin-binding protein 1 1 1+

    Septin family

    SEP5_MOUSE Septin 5 1+ 1+

    SEP7_MOUSE Septin 7 1+

    Y202_HUMAN Septin-like protein KIAA0202 1+

    8922712 Hypothetical protein FLJ10849 (Septin 2 or 6 homologue) 1+ 1+

    Protein folding (n 2)GR75_MOUSE Stress-70 protein 1+

    HS7C_MOUSE Heat-shock cognate 71-kDa protein 1

    Stress/detoxification (n 4)DHCA_MOUSE Carbonyl reductase [NADPH] 1 1+

    GTP2_MOUSE Glutathione S-transferase P 2 *2

    LGUL_MOUSE Glyoxalase I *1+

    TRXB_MOUSE Thioredoxin reductase 1+

    Cytoskeleton (n 4)AR20_HUMAN ARP2/3 complex 20-kDa subunit 1

    CAP1_MOUSE Adenyl cyclase-associated protein 1 *1/1+

    DYN1_MOUSE Dynamin-1 1 1+

    TBA1_MOUSE Tubulin a1 chain 3+

    1264 M. Periquet et al.

    2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 12591276

  • This approach conrmed that a large proportion of thedifferentially regulated proteins were involved in energymetabolism (nup 9, ndown 10, nup/down 4), includingthe glycolytic pathway, the Krebs cycle and the mitochond-rial respiratory chain. The differentially regulated proteinsalso played roles in signal transduction pathways (nup 6, ndown 4, nup/down 1), vesicle trafcking (nup 6,ndown 1, nup/down 2), cytoskeletal dynamics (nup 2, ndown 2, nup/down 2), protein folding (nup 3,ndown 3) and degradation (nup 2, ndown 1), and theoxidative stress response and/or detoxication processes(nup 4, ndown 1). These latter changes occurred in theabsence of modications in protein oxidation, as revealed bythe western blot analysis of protein carbonyls in brain lysatesof 2-month-old (not shown) and 12-month-old WT and KO

    mice (Fig. 5). Other categories included proteins involved inlipid metabolism (nup 3, ndown 2), amino acid andprotein biosynthesis pathways (nup 2, ndown 2), RNAprocessing (nup 1, ndown 1) and neurotransmitter hand-ling (nup 2).The proteins that increased in abundance in parkin KO

    mice are potential substrates of Parkins E3 ubiquitinproteinligase activity. Three members of the membrane-associatedguanylate kinase (MAGUK) family of neuronal scaffoldingproteins (p55 subfamily members 2, 3 and 6) were foundamong the proteins found to be exclusively up-regulated inparkin KO mice. In addition, members of the septin family(CDCRel-1/septin5, septin 7, septin-like protein KIAA0202,hypothetical septin 2 or 6 homologue FLJ10849) (Tables 2and 3) were up-regulated both in the cortex and in the

    Table 2 (Continued)

    SwissProt or gi accession Protein name

    No. and status of spots

    Striatum

    2 months

    Striatum

    12 months

    Protein degradation (n 3)PSB5_MOUSE Proteasome subunit beta type 5 1+

    UBL1_MOUSE Ubiquitin carboxyterminal hydrolase L1 1+

    18490720 Deubiquitinating enzyme OTUB1 1

    Lipid metabolism (n 3)ACDV_MOUSE Acyl-CoA dehydrogenase, very-long-chain specific 1+

    PCCA_RAT Propionyl CoA carboxylase a chain 1+

    6678760 Lysophospholipase 1 1

    Protein biosynthesis (n 2)SYY_HUMAN Tyrosyl tRNA synthetase 1+

    SYTC_HUMAN Threonyl tRNA synthetase 1

    Amino acid synthesis (n 2)GLNA_MOUSE Glutamine synthetase 1

    SRR_MOUSE Serine racemase 1+

    RNA processing (n 3)HE47_RAT Probable ATP-dependent RNA helicase p47 1

    PCB2_MOUSE Poly(rC)binding protein 1+

    Neurotransmitter metabolism (n 2)GABT_RAT 4-Aminobutyrate aminotransferase, mitochondrial 1+ 2+

    TY3H_MOUSE Tyrosine 3-hydroxylase 1+

    Others (n 7)ALBU_MOUSE Serum albumin 1

    ARHY_MOUSE ADP-ribosylarginine hydrolase 1+

    CAH2_MOUSE Carbonic anhydrase II 3

    FCE2_MOUSE Low-affinity immunoglobulin epsilon FC receptor 1+

    KPR1_HUMAN Ribose-phosphate pyrophosphokinase I 1

    POR1_MOUSE Voltage-dependent anion-selective channel protein 1 2+

    SPEE_MOUSE Spermidine synthase 1+

    2D analysis allows the resolution of different isoforms and/or post-translational modifications of a same protein. Thus, several dysregulated spots

    can correspond to a unique protein. These isoforms were in some cases all up-regulated (status +) or down-regulated (status ) in parkin KO mice,

    or subjected to opposite regulation, owing to altered electrophoretic mobility (status +/). Owing to the use of overlapping pH gradients, the same

    protein could also be detected twice in two consecutive pH gradients gels. In all the tables of the article, the number of spots corresponding to the

    same protein is noted next to the status associated. *Proteins differing in abundance by a factor > 2.

    Proteomic analysis of parkin knockout mice 1265

    2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 12591276

  • Table 3 Proteins differentially regulated in the cortex of WT and parkin KO mice

    SwissProt or gi accession Protein name

    No. and status of spots

    Cortex 2 months Cortex 12 months

    Energy metabolism (n 13)ATPA_MOUSE ATP synthase a chain, mitochondrial 1/1+ 2+

    ATPB_MOUSE ATP synthase b chain, mitochondrial 1

    DHSA_HUMAN Succinate dehydrogenase flavoprotein subunit 1+

    ENOG_MOUSE c Enolase 1/1+

    G3P_MOUSE Glyceraldehyde-3-phosphate dehydrogenase 3/1+

    LDHA_MOUSE L-Lactate dehydrogenase A chain 1

    MDHC_MOUSE Malate dehydrogenase, cytoplasmic 2

    NUCM_HUMAN NADH-ubiquinone oxidoreductase 49-kDa subunit 1-

    ODPA_MOUSE Pyruvate dehydrogenase E1 component a subunit 1+

    PGK1_MOUSE Phosphoglycerate kinase 1 *1

    UCR1_RAT Ubiquinol-cytochrome c reductase complex core protein I 1+

    UCR2_MOUSE Ubiquinol-cytochrome c reductase complex core protein 2 1

    128325765 NADH-ubiquinone oxidoreductase PDSW subunit HS homolog 1

    Signal transduction (n 4)DPY2_MOUSE Dihydropyrimidinase-related protein-2 (CRMP-2) 2/3+

    9910474 MAGUK p55 subfamily member 6 1+

    7709986 Sumo-1 activating enzyme subunit 2 1+

    MPP2_HUMAN MAGUK p55 subfamily member 2 1+

    Vesicular trafficking (n 3)NSF_MOUSE N-ethylmaleimide sensitive fusion protein 1+

    SEP7_MOUSE Septin 7 1+

    SNX5_MOUSE Sorting nexin 5 *1+

    Protein folding (n 6)GR75_MOUSE Stress-70 protein 2+

    GR78_MOUSE 78-kDa glucose-regulated protein 1

    HS72_MOUSE Heat-shock-related 70-kDa protein 2 1

    HS7C_MOUSE Heat-shock cognate 71-kDa protein 2

    OS94_MOUSE Heat-shock 70-related protein APG-1 1+

    TCP2_MOUSE T-complex protein 1, a subunit B 1+

    Stress/detoxification (n 2)ACON_HUMAN Aconitate hydratase, mitochondrial 2+

    GTP2_MOUSE Glutathione S-transferase P 2 *1

    Cytoskeleton (n 2)ACTG_HUMAN c-Actin 1

    SPCN_MOUSE Spectrin a chain, brain 2+

    Protein degradation (n 1)UBA1_MOUSE Ubiquitin-activating enzyme E1 type 1 1+ 1+

    Lipid metabolism (n 2)CAO1_MOUSE Acyl-coenzyme A oxidase 1, peroxisomal 1+

    MTE1_MOUSE Acyl CoA thioester hydrolase 1

    Others (n 5)A1A3_RAT Sodium/potassium-transporting ATPase a-3 chain 1

    CLB2_MOUSE Calretinin 1

    DD19_MOUSE ATP-dependent RNA helicase DDX19, dead box protein 1+

    MO25_MOUSE No known function 1

    POR1_MOUSE Voltage-dependent anion-selective channel protein 1 1/1+

    2D analysis allows the resolution of different isoforms and/or post-translational modifications of a same protein. Thus, several dysregulated spots

    can correspond to a unique protein. These isoforms were in some cases all up-regulated (status +) or down-regulated (status ) in parkin KO mice,

    or subjected to opposite regulation, owing to altered electrophoretic mobility (status +/). Owing to the use of overlapping pH gradients, the same

    protein could also be detected twice in two consecutive pH gradients gels. In all the tables of the article, the number of spots corresponding to the

    same protein is noted next to the status associated. *Proteins differing in abundance by a factor > 2.

    1266 M. Periquet et al.

    2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 12591276

  • striatum, at both ages (Tables 4 and 5). In particular, vespots, migrating with the same molecular weight but withdifferent isoelectric points, were identied as the Parkinsubstrate CDCRel-1/septin5 (M. Duchesne, personal com-munication). The abundance of one of these spots increasedby at least 45% in the striatum of 2- and 12-month-old parkinKO mice compared with WT mice (Tables 2 and 4). Inaddition, the levels of a second spot were increased by 33%in the striatum of 12-month-old mice (p < 0.021). As wedecided arbitrarily to take into consideration only the mostsignicant protein abundance changes (> 45%), this proteinis not listed in the tables presented. A reproducible increasein total CDCRel-1 levels was conrmed in individual cortexsamples (Fig. 4) and pooled striata (data not shown) of

    parkin KO mice, resolved by conventional one-dimensionalgel electrophoresis and analysed by quantitative westernblotting. However, this increase did not reach statisticalsignicance. This is probably due to the fact that the vedifferent CDCRel-1 isoforms with identical molecular weightmigrate as a single band on one-dimensional gels.Additional Parkin substrates showed altered abundance in

    our parkin KO model. Huynh et al. (2003) reported thatsynaptotagmin XI and synaptotagmin I are ubiquitylated byParkin. In our study, synaptotagmin I was found to beup-regulated by 40% (p < 0.001) in the cortex of 2-month-old mice (data not shown). In addition, several spotscorresponding to the tubulin a-1 chain (Ren et al. 2003)were increased in abundance in the striatum of 12-month-oldparkin KO mice (Table 2). Finally, we also analysed thelevels of our previously identied Parkin substrate p38 (Cortiet al. 2003) by one-dimensional western blotting, although itwas not identied as being altered in abundance in our 2D

    Table 5 Proteins differentially regulated in cortex and striatum at 2

    and/or 12 months

    SwissProt or

    gi accession

    Age

    (months)

    No. and status

    of spots Fold

    Energy metabolism

    ATPA_MOUSE 2/12 1/8+ > 1.45

    DHSA_HUMAN 2 2+ > 1.45

    G3P_MOUSE 2 3/3+ >1.45

    LDHA_MOUSE 2/12 2 > 1.45

    MDHC_MOUSE 2/12 3 > 1.45

    PGK1_MOUSE 2/12 2 > 1.45

    UCR1_RAT 2/12 2+ > 1.45

    Signal transduction

    DPY2_MOUSE 2/12 7/10+ > 1.45

    Vesicular trafficking

    NSF_MOUSE 2/12 12+ > 1.45

    SEP7_MOUSE 2/12 2+ > 1.45

    Protein folding

    GR75_MOUSE 12 3+ > 1.45

    HS7C_MOUSE 2 3 > 1.45

    Stress/detoxification

    GTP2_MOUSE 2 3 > 2.00

    Others

    POR1_MOUSE 2/12 1/3+ > 1.45

    2D analysis allows the resolution of different isoforms and/or post-

    translational modifications of a same protein. Thus, several dysregu-

    lated spots can correspond to a unique protein. These isoforms were

    in some cases all up- (status +) or down-regulated (status -) in parkin

    KO mice, or subjected to opposite regulation, owing to altered elec-

    trophoretic mobility (status +/-). Owing to the use of overlapping pH

    gradients, the same protein could also be detected twice in two con-

    secutive pH gradient gels. In all the tables of the article, the number of

    spots corresponding to the same protein is noted next to the status

    associated.

    Table 4 Proteins differentially regulated at both 2 and 12 months

    SwissProt/gi accession

    No. and status

    of spots Fold

    Energy metabolism

    ATPA_MOUSE 1/8+ > 1.45

    CISY_HUMAN 2 > 1.45

    KAD3_MOUSE 2+ > 1.45

    KPY2_MOUSE 5/2+ > 1.45

    LDHA_MOUSE 2 > 1.45

    MDHC_MOUSE 3 > 1.45

    PGK1_MOUSE 2 > 2.00

    UCR1_RAT 2+ > 1.45

    Signal transduction

    DPY2_MOUSE 7-/10+ > 1.45

    Vesicular trafficking

    NSF_MOUSE 1/2+ >1.45

    STB1_MOUSE 1/1+ > 1.45

    Septin family

    SEP5_MOUSE 2+ > 1.45

    SEP7_MOUSE 2+ > 1.45

    8922712 2+ > 1.45

    Cytoskeleton

    DYN1_MOUSE 1/1+ > 1.45

    Protein degradation

    UBA1_MOUSE 2+ > 1.45

    Neurotransmitter metabolism

    GABT_RAT 3+ > 1.45

    Others

    POR1_MOUSE 1/3+ > 1.45

    2D analysis allows the resolution of different isoforms and/or post-

    translational modifications of a same protein. Thus, several dysregu-

    lated spots can correspond to a unique protein . These isoforms were

    in some cases all up-regulated (status +) or down-regulated (status )

    in parkin KO mice, or subjected to opposite regulation, owing to altered

    electrophoretic mobility (status +/). Owing to the use of overlapping

    pH gradients, the same protein could also be detected twice in two

    consecutive pH gradients gels. In all the tables of the article, the

    number of spots corresponding to the same protein is noted next to the

    fold difference associated.

    Proteomic analysis of parkin knockout mice 1267

    2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 12591276

  • DIGE analysis. The quantitative analysis of protein samplesfrom individual striata from 12-month-old KO and WT micedid not reveal any signicant change in abundance of p38levels (Fig. 4).

    Discussion

    Nowadays, 2D DIGE is the most powerful 2D polyacryla-mide gel electrophoresis-based approach for widespreadprotein proling. We used this recent technology to compareprotein expression proles in parkin KO and WT mice, usinga pool of samples as an internal standard and the dedicatedDeCyder analysis software developed by Amersham Bio-sciences (Tonge et al. 2001; Gharbi et al. 2002; Yan et al.2002). Owing to the large amount of material required for 2DDIGE, our analyses were performed on pools of brainextracts obtained from six animals for each condition. Thisexperimental paradigm gives an appropriate indication of the

    average biological differences between groups of samples,although it does not provide information on inter-animalvariation. The validity of this approach was demonstrated ina previous study showing that similar results are obtainedwhen individual animals or pooled samples are analysed by2D DIGE (Tonge et al. 2001). We found that 87 proteinsdiffered in abundance by at least 45% in parkin KO and WTmice.Classication of these proteins led to the identication of

    12 major functional categories affected by inactivation of theparkin gene. The most frequently represented categoryincluded proteins related to energy metabolism, particularlyto the glycolytic pathway, the Krebs cycle and the mitoch-ondrial respiratory chain. This functional class has also beenshown to be affected at the mRNA and/or protein levels inother models of cell degeneration and in neurodegenerativediseases (Loring et al. 2001; Gozal et al. 2002; Napolitanoet al. 2002; Seong et al. 2002; Tilleman et al. 2002a, 2002b;Xie et al. 2002; Kuhn et al. 2003). These proteins wereeither up- or down-regulated, or subject to post-translationalmodication, such that the overall consequences of theirdifferential regulation are difcult to predict. In the absenceof a neurodegenerative phenotype, these changes probably

    Fig. 2 2D gel images showing selected differentially regulated pro-

    teins. (a) An up-regulated spot (boxed protein) identified as aconitase

    hydratase, which was 1.97 times more abundant in the cortex of

    2-month-old KO than in WT mice. (b). A down-regulated spot (boxed

    protein) identified as phosphoglycerate kinase 1, which was 2.09 times

    less abundant in the cortex of 2-month-old KO than in WT mice. (c)

    Two isoforms of a chain ATP synthase protein, which were regulated

    differently in the cortex at the age of 2 months. The first isoform was

    down-regulated by at least 60% ( 1.66), whereas the second morebasic variant was up-regulated to the same extent ( 1.63), suggestinga shift due to phosphorylation or other post-translational modifications.

    Squares and circles indicate downward and upward changes in parkin

    KO mice respectively.

    Table 6 Functions of differentially regulated proteins assessed by

    keyword analysis

    Occurrences SwissProt keyword Confidence index

    16 Glycolysis 20.66

    6 Heat_shock 13.34

    5 ATP_synthesis 13.34

    7 Flavoprotein 7.37

    5 Hydrogen_ion_transport 6.46

    5 Proteasome 6.25

    8 Ligase 6.16

    5 FAD 5.56

    8 Lyase 5.52

    7 NADP 5.29

    5 Protein_biosynthesis 5.13

    8 Chaperone 4.85

    8 NAD 4.71

    18 Transit_peptide 4.68

    8 Kinase 4.64

    11 Acetylation 4.04

    25 Oxidoreductase 3.86

    7 Magnesium 3.79

    6 Isomerase 3.58

    9 GTP binding 3.37

    22 Mitochondrion 3.21

    Only keywords occurring more than four times were taken into account

    and their relative significance (confidence index 3) was determinedby normalizing the observed frequency of each keyword to the relative

    frequency in the Swissprot database as a whole. Only entries for

    mouse proteins were considered.

    1268 M. Periquet et al.

    2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 12591276

  • reect an adaptive regulation of cellular energy in parkin KOmice, as suggested by the altered abundance of enzymesinvolved in glycolysis (glyceraldehyde-3-dehydrogenase,c-enolases, pyruvate kinase) and energy regeneration(subunits of the mitochondrial ATP synthase).Several lines of evidence suggest that dysfunction of the

    ubiquitin-dependent proteasomal degradation pathway plays

    a major role in the pathophysiology of both familial andsporadic PD. Changes in proteins related to this pathwayhave previously been reported at the transcriptional level incell and mouse models of PD (Cadet et al. 2001; Ryu et al.2002; Kuhn et al. 2003). In parkin KO mice, the abundanceof several stress-induced chaperones was altered, includingheat-shock protein 70-related proteins, the osmotic stressprotein Osp94 and the T complex protein 1, which plays arole in the folding of actin and tubulin, and probably also ofother cytoskeletal proteins (Dunn et al. 2001). Of note,several cytoskeletal proteins were altered in abundance inparkin KO mice, including the Parkin substrate tubulin a-1chain (Ren et al. 2003). In addition, the PD-associatedprotein UCH-L1, and the proteasome subunit b type 5 weremore abundant in parkin KO mice, whereas the deubiquityenzyme OTU-domain Uba1-binding protein (OTUB1) wasless abundant. These modications were paralleled bychanges in the levels of several enzymes linked to cellularstress and detoxication processes. In particular, the level ofthe antioxidant protein Glutathione S-transferase P2 (GTP2)was decreased in both striatum and cortex from 2-month-oldparkin KO mice, whereas other proteins (carbonyl reductase,glyoxalase I, thioredoxin reductase) known to be protectiveagainst oxidative stress-induced neurodegeneration (Chenet al. 2004) increased in abundance in these mice. Thesechanges might reect an adaptive response to high concen-trations of free radicals, as suggested by our previousobservation that reduced glutathione concentrations are highin both the striatum and in fetal mesencephalic neuronalcultures from parkin KO mice (Itier et al. 2003). In youngand aged parkin KO mice, however, the levels of protein

    Fig. 3 Functional distribution of proteins

    differentially regulated in parkin KO and WT

    mice. The major functional categories of all

    the proteins identified in cortex and striatum

    are plotted in a lateral bar graph as a per-

    centage of the total that increased in

    abundance (right) and the total that

    decreased in abundance (left). Only func-

    tional categories with two or more members

    are shown for proteins differing in abun-

    dance by at least 45% in the two types of

    mice. Proteins with several isoforms, some

    of which were up-regulated and some of

    which were down-regulated, were exclu-

    ded.

    Fig. 4 Quantitative western blot analyses of CDCRel-1, calretinin and

    p38 in the cortex of WT and parkin KO mice. A slight but reproducible

    increase in CDCRel-1 protein levels was observed in KO mice,

    whereas calretinin protein levels were significantly reduced. In con-

    trast, p38 levels were similar in WT and KO mice. Data were obtained

    by normalizing the relative intensities of CDCRel-1, calretinin and p38

    signals to the intensity of the respective actin signal in each sample.

    The mean CDCRel-1/actin, calretinin/actin and p38/actin ratios were

    set arbitrarily at 1. Values are expressed as mean SEM (n 5animals per group). *p < 0.05 versus WT (Students t-test). Results

    representative of at least three independent experiments are shown.

    Proteomic analysis of parkin knockout mice 1269

    2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 12591276

  • carbonyls were comparable to those of WT mice, indicatingthat antioxidant defences and/or detoxication processes areefcient in these animals.Several proteins involved in vesicle trafcking or function

    were also affected in parkin KO mice, including componentsof the SNARE (soluble N-ethylmaleimide sensitive fusionprotein (NSF) attachment protein receptors) complex: NSF,syntaxin 1B and syntaxin-binding protein 1. In particular,four members of the septin protein family (which is involved

    in vesicle transport and exocytosis, but also in cytokinesis,protein scaffolding and several other cellular processes),including the Parkin substrate septin5/CDCRel-1, were moreabundant in parkin KO mice than in WT mice. Members ofthis protein family accumulate in neurobrillary tangles andglial brils in Alzheimers disease, and in lewy bodies in PD(Kitada et al. 1998; Ihara et al. 2003). CDCRel-1 and therelated protein CDCRel-2 also accumulate in the brains ofPD sufferers with parkin gene mutations (Zhang et al. 2000;Choi et al. 2003). Regulation of the degradation of synapticvesicle-associated proteins by Parkin, which is present onsynaptic vesicles (Kubo et al. 2001), may modulate neuro-transmitter release. Loss of this function might be partiallyresponsible for the observed abnormalities in dopaminergicand glutamatergic neurotransmission in parkin KO mice(Itier et al. 2003). The up-regulation of three members of theMAGUK p55 subfamily of synaptic scaffolding proteins mayalso be involved in these changes in synaptic function.Interestingly, a previous study demonstrated a direct inter-action between Parkin and the MAGUK family membercalcium/calmodulin-dependent serine protein kinase (CASK)and suggested that CASK might target Parkin to specializedfunctional membrane domains where it may modulate theactivity of NMDA receptors (Fallon et al. 2002).While this work was in progress, Palacino et al. (2004)

    reported a differential proteomic analysis of another parkinKO mouse model, using conventional 2D gel technologyand silver-stained gels. Surprisingly, they found consistentdecreases in the abundance of only 13 proteins and alteredelectrophoretic mobility of an additional one. Decreasedabundance of proteins involved in mitochondrial function,i.e subunits of complexes I and IV, was associated with areduction in the respiratory capacity of striatal mitochon-dria from parkin KO mice. This mouse model alsoexhibited decreased levels of proteins involved in protec-tion against oxidative stress, decreased serum antioxidantcapacity and, in contrast to our model, increased proteinand lipid peroxidation. Only two (pyruvate dehydrogenaseE1a1 and glyoxalase I) of the 13 proteins identied inPalacinos study were also found to be changed inabundance in our parkin KO mice. These proteins showeda decrease in abundance in Palacinos study, whereas theywere increased in abundance in our parkin KO mice,raising the possibility that these proteins represent falsepositives in either or both studies. However, this surprisingdiscordance might also result from the identication ofdifferent protein isoforms or post-translational variants,which would be dysregulated differently in the two studies,as was the case in a previous report (Choi et al. 2004b).Indeed, our proteomic analysis was performed using abroader range of pH gradients, which might have led to theidentication of protein isoforms that were not resolved onthe 310 pH gradient gel used by Palacino. The consid-erably greater number of proteins identied in our study

    (a)

    (b)

    Fig. 5 Levels of protein carbonyls are similar in the cortex and stria-

    tum of WT and parkin KO mice. Analysis of individual cortex (a) and

    striatum (b) samples from 12-month-old animals revealed comparable

    levels of oxidatively damaged proteins in WT and parkin KO mice.

    Equivalent protein loading was confirmed by western blotting using an

    anti-actin antibody. C, protein samples in which 2,4-din-

    itrophenylhydrazine was omitted; M, molecular weight protein stand-

    ard; BSA, oxidatively modified bovine seum albumin.

    1270 M. Periquet et al.

    2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 12591276

  • Table

    7P

    rote

    ins

    diffe

    rentially

    regula

    ted

    betw

    eenparkin

    KO

    and

    WT

    mic

    eand

    identified

    inoth

    er

    pro

    teom

    icstu

    die

    sanaly

    zin

    gneuro

    degenera

    tive

    models

    or

    dis

    ord

    ers

    Sw

    issP

    rot

    or

    giaccessio

    nP

    rote

    innam

    eS

    tatu

    sD

    isease

    Modeland/o

    rtissue

    Refe

    rences

    Energ

    ym

    eta

    bolis

    m

    ALF

    C_M

    OU

    SE

    Fru

    cto

    se-b

    isphosphate

    ald

    ola

    se

    C

    PD

    parkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    [Fra

    gm

    ent]

    S

    CZ

    DH

    um

    an

    bra

    inP

    rabakara

    netal.

    2004

    AT

    PA

    _M

    OU

    SE

    AT

    Psynth

    asea

    chain

    ,m

    itochondrial

    +P

    Dparkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    na

    AD

    Tau

    transgenic

    mic

    eT

    illem

    anetal.

    2002a

    AT

    PB

    _M

    OU

    SE

    AT

    Psynth

    aseb

    chain

    ,m

    itochondrial

    P

    Dparkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    +A

    DH

    um

    an

    bra

    inT

    sujietal.

    2002

    D

    SH

    um

    an

    bra

    inK

    imetal.

    2000

    DLD

    H_M

    OU

    SE

    Dih

    ydro

    lipoam

    ide

    dehydro

    genase

    P

    Dparkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    na

    AD

    Tau

    transgenic

    mic

    eT

    illem

    anetal.

    2002a

    DH

    SA

    _H

    UM

    AN

    Succin

    ate

    dehydro

    genase

    flavopro

    tein

    +A

    Dparkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    subunit

    +A

    DG

    SK

    3b

    transgenic

    mic

    eT

    illem

    anetal.

    2002b

    EN

    OG

    _M

    OU

    SE

    cE

    nola

    se

    /+

    PD

    parkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    +A

    DH

    um

    an

    bra

    inS

    chonberg

    eretal.

    2001

    S

    CZ

    DH

    um

    an

    bra

    inP

    rabakara

    netal.

    2004

    na

    AD

    Tau

    transgenic

    mic

    eT

    illem

    anetal.

    2002a

    G3P

    _M

    OU

    SE

    Gly

    cera

    ldehyde

    3-p

    hosphate

    dehydro

    genase

    /+

    PD

    parkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    +A

    DH

    um

    an

    bra

    inS

    chonberg

    eretal.

    2001

    P

    DM

    PT

    Pm

    ice

    m

    itochondria

    SN

    Jin

    etal.

    2005

    S

    CZ

    DH

    um

    an

    bra

    inP

    rabakara

    netal.

    2004

    na

    AD

    Tau

    transgenic

    mic

    eT

    illem

    anetal.

    2002a

    NU

    CM

    _H

    UM

    AN

    NA

    DH

    -ubiq

    uin

    one

    oxid

    ore

    ducta

    se

    A

    Dparkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    49-k

    Da

    subunit

    A

    DG

    SK

    3b

    transgenic

    mic

    eT

    illem

    anetal.

    2002b

    OD

    PA

    _M

    OU

    SE

    Pyru

    vate

    dehydro

    genase

    E1

    com

    ponent

    +P

    Dparkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    asubunit

    S

    CZ

    DH

    um

    an

    bra

    inP

    rabakara

    netal.

    2004

    P

    DM

    PT

    Pm

    ice

    m

    itochondria

    SN

    Jin

    etal.

    2005

    P

    Dparkin

    KO

    mic

    e,

    ventr

    alm

    idbra

    inP

    ala

    cin

    oetal.

    2004

    UC

    R1_R

    AT

    Ubiq

    uin

    ol-cyto

    chro

    mec

    reducta

    se

    com

    ple

    x+

    AD

    parkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    core

    pro

    tein

    I

    AD

    Hum

    an

    bra

    inK

    imetal.

    2000

    S

    CZ

    DH

    um

    an

    bra

    inP

    rabakara

    netal.

    2004

    Sig

    naltr

    ansduction

    DP

    Y2_M

    OU

    SE

    Dih

    ydro

    pyrim

    idin

    ase-r

    ela

    ted

    pro

    tein

    -2+

    /P

    Dparkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    (CR

    MP

    -2)

    A

    DH

    um

    an

    bra

    inT

    sujietal.

    2002

    A

    DH

    um

    an

    bra

    inS

    chonberg

    eretal.

    2001

    (G

    lyfo

    rm)

    AD

    Hum

    an

    bra

    inK

    annin

    enetal.

    2004

    S

    CZ

    DH

    um

    an

    bra

    inP

    rabakara

    netal.

    2004

    +(O

    xfo

    rm)

    AD

    P

    DH

    um

    an

    bra

    inC

    hoietal.

    2004a

    +(O

    xfo

    rm)

    AD

    Hum

    an

    bra

    inC

    aste

    gnaetal.

    2002b

    +A

    DG

    SK

    3b

    transgenic

    mic

    eT

    illem

    anetal.

    2002b

    Proteomic analysis of parkin knockout mice 1271

    2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 12591276

  • Table

    7(C

    ontinued)

    Sw

    issP

    rot

    or

    giaccessio

    nP

    rote

    innam

    eS

    tatu

    sD

    isease

    Modeland/o

    rtissue

    Refe

    rences

    P2B

    A_M

    OU

    SE

    Ser/

    Thr

    pro

    tein

    phosphata

    se

    2B

    cata

    lytic

    +P

    Dparkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    subunit,a

    isofo

    rm+

    AD

    GS

    K3b

    transgenic

    mic

    eT

    illem

    anetal.

    2002b

    143Z

    _M

    OU

    SE

    14-3

    -3pro

    tf/

    d

    PD

    parkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    P

    DM

    PT

    Pm

    ice

    m

    itochondria

    SN

    Jin

    etal.

    2005

    na

    AD

    Tau

    transgenic

    mic

    eT

    illem

    anetal.

    2002a

    Vesic

    ula

    rtr

    affi

    ckin

    g

    NS

    F_M

    OU

    SE

    N-e

    thylm

    ale

    imid

    esensitiv

    efu

    sio

    n

    pro

    tein

    +/

    PD

    parkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    A

    DH

    um

    an

    bra

    inS

    chonberg

    eretal.

    2001

    +S

    CZ

    DH

    um

    an

    bra

    inP

    rabakara

    netal.

    2004

    +A

    DG

    SK

    3b

    transgenic

    mic

    eT

    illem

    anetal.

    2002b

    ST

    B1_M

    OU

    SE

    Synta

    xin

    -bin

    din

    gpro

    tein

    1+

    /P

    Dparkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    +P

    DM

    PT

    Pm

    ice

    m

    itochondria

    SN

    Jin

    etal.

    2005

    SE

    P7_M

    OU

    SE

    Septin

    7+

    PD

    parkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    S

    CZ

    DH

    um

    an

    bra

    inP

    rabakara

    netal.

    2004

    Pro

    tein

    fold

    ing

    HS

    72_M

    OU

    SE

    Heat-

    shock-r

    ela

    ted

    70-k

    Da

    pro

    tein

    2

    PD

    parkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    S

    CZ

    DH

    um

    an

    bra

    inP

    rabakara

    netal.

    2004

    na

    AD

    Tau

    transgenic

    mic

    eT

    illem

    anetal.

    2002a

    Str

    ess/d

    eto

    xifi

    cation

    AC

    ON

    _H

    UM

    AN

    Aconitate

    hydra

    tase,

    mitochondrial

    +P

    Dparkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    S

    CZ

    DH

    um

    an

    bra

    inP

    rabakara

    netal.

    2004

    DH

    CA

    _M

    OU

    SE

    Carb

    onylre

    ducta

    se

    [NA

    DP

    H]

    1+

    PD

    parkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    S

    CZ

    DH

    um

    an

    bra

    inP

    rabakara

    netal.

    2004

    +A

    DD

    SH

    um

    an

    bra

    inB

    alc

    zetal.

    2001

    LG

    UL_M

    OU

    SE

    Gly

    oxala

    se

    I+

    PD

    parkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    +P

    DM

    PT

    Pm

    ice

    m

    itochondria

    SN

    Jin

    etal.

    2005

    -

    PD

    parkin

    KO

    mic

    e,

    ventr

    alm

    idbra

    inP

    ala

    cin

    oetal.

    2004

    Cyto

    skele

    ton

    DY

    N1_M

    OU

    SE

    Dynam

    in-1

    +/

    PD

    parkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    +/

    SC

    ZD

    Hum

    an

    bra

    inP

    rabakara

    netal.

    2004

    na

    AD

    Tau

    transgenic

    mic

    eT

    illem

    anetal.

    2002a

    SP

    CN

    _M

    OU

    SE

    Spectr

    ina

    chain

    ,bra

    in+

    PD

    parkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    S

    CZ

    DH

    um

    an

    bra

    inP

    rabakara

    netal.

    2004

    TB

    A1_M

    OU

    SE

    Tubulin

    achain

    +P

    Dparkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    A

    DG

    SK

    3b

    transgenic

    mic

    eT

    illem

    anetal.

    2002b

    S

    CZ

    DH

    um

    an

    bra

    inP

    rabakara

    netal.

    2004

    1272 M. Periquet et al.

    2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 12591276

  • might be explained by technological differences. First,cyanin dye staining is more sensitive than silver stainingand has a greater quantication range, extending linearlyover four orders of magnitude (Patton 2000). Second, thepooled internal standard used with the 2D DIGE technol-ogy allows detection of changes in protein abundance thatcannot be seen in pairwise comparisons of individualbiological samples (Friedman et al. 2004). Third, the useof overlapping pH gradients in our study increased theresolution of 2D gels. Finally, differences in the number,nature and status of the differentially regulated proteinsmight also result from differences in the tissues and agesanalysed. Palacinos study was conducted on ventralmidbrain of 8-month-old mice, whereas our proteomicanalysis was performed on cortex and striatum from 2- and12-month-old mice.In conclusion, we have identied changes in the abun-

    dance of a large number of proteins belonging to variousfunctional categories in our parkin KO model. Several ofthese functional categories have already been linked to cell/animal models of PD as well as to other neurodegenerativeconditions in previous differential gene expression orproteomic analyses, raising the question of their specicity.The categories most reproducibly affected at the mRNAand/or protein levels in these studies include proteinsinvolved in energy metabolism (Loring et al. 2001; Gozalet al. 2002; Napolitano et al. 2002; Seong et al. 2002;Tilleman et al. 2002a, 2002b; Xie et al. 2002; Kuhn et al.2003), protein folding and degradation (Cadet et al. 2001;Ryu et al. 2002; Kuhn et al. 2003) and detoxicationprocesses (Balcz et al. 2001; Prabakaran et al. 2004). Otherfunctional classes, such as proteins implicated in vesicletrafcking, cytoskeletal dynamics and protein folding anddegradation, were also frequently identied in severalproteomic analyses on brains from patients affected byschizophrenia, Downs syndrome or Alzheimers disease(Schonberger et al. 2001; Castegna et al. 2002a, 2002b;Choi et al. 2004a; Tilleman et al. 2002a, 2002b; Kadotaet al. 2004; Prabakaran et al. 2004; Jin et al. 2005).Detailed analysis of the protein abundance changes reportedin the various proteomic studies available so far in the eldof neurodegeneration and psychiatric conditions (Table 7)revealed that 26 of the 87 proteins identied in our parkinKO model (30%) were also affected in other studies. Theseproteins cover the major functional classes shown in Fig. 3.Therefore, in general, common pathways appear to bealtered in these models, although the nature of the affectedproteins is often different. However, our study also hints atthe possible specic involvement of members of the septinand MAGUK protein families in parkin-related PD. Indeed,only one previous study reported the down-regulation of aseptin (septin7) in human brain lysates from patients withschizophrenia (Prabakaran et al. 2004) (Table 7) and, to ourknowledge, alterations in MAGUK protein abundance wereTa

    ble

    7(C

    ontinued)

    Sw

    issP

    rot

    or

    giaccessio

    nP

    rote

    innam

    eS

    tatu

    sD

    isease

    Modeland/o

    rtissue

    Refe

    rences

    Pro

    tein

    degra

    dation

    UB

    L1_M

    OU

    SE

    Ubiq

    uitin

    carb

    oxyte

    rmin

    al

    hydro

    lase

    L1

    +P

    Dparkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    S

    CZ

    DH

    um

    an

    bra

    inS

    chonberg

    eretal.

    2001

    P

    DA

    DH

    um

    an

    bra

    inC

    hoietal.

    2004b

    D

    SM

    ouse

    ES

    cells

    Kadota

    etal.

    2004

    +(O

    xfo

    rm)

    AD

    Hum

    an

    bra

    inC

    aste

    gnaetal.

    2002a

    Pro

    tein

    bio

    synth

    esis

    SY

    Y_H

    UM

    AN

    Tyro

    syltR

    NA

    synth

    eta

    se

    +P

    Dparkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    S

    CZ

    DH

    um

    an

    bra

    inP

    rabakara

    netal.

    2004

    Am

    ino

    acid

    synth

    esis

    GLN

    A_M

    OU

    SE

    Glu

    tam

    ine

    synth

    eta

    se

    P

    Dparkin

    KO

    mic

    e,

    cort

    exstr

    iatu

    mP

    eriquetetal.

    2005

    +A

    DG

    SK

    3b

    transgenic

    mic

    eT

    illem

    anetal.

    2002b

    +(O

    xfo

    rm)

    AD

    Hum

    an

    bra

    inC

    aste

    gnaetal.

    2002a

    Ox,

    oxid

    ized;

    Gly

    ,gly

    cosyla

    ted;

    PD

    ,P

    ark

    insons

    dis

    ease;

    AD

    ,A

    lzheim

    ers

    dis

    ease;

    DS

    ,D

    ow

    ns

    syndro

    me;

    SC

    ZD

    ,schiz

    ophre

    nia

    ;S

    N,

    substa

    ntia

    nig

    ra;

    +,

    incre

    ased

    abundance,

    ,

    decre

    ased

    abundance;

    +/-

    ,altere

    dele

    ctr

    ophore

    tic

    mobili

    ty;

    na,

    data

    not

    availa

    ble

    .

    Proteomic analysis of parkin knockout mice 1273

    2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 12591276

  • not observed in any of the previously reported differentialproteomic analyses.Overall, although further functional studies are required to

    validate the proteins identied in our study and link themmechanistically to the molecular events underlying parkin-related Parkinsons disease, these data already constitute avaluable reference bank for future investigations into thepathological mechanisms involved in the early stages of thisdisease.

    Acknowledgements

    We thank Lydia Guennec for technical assistance, Frederic Darios

    and Francisco Araujo for helpful discussions, and Merle Ruberg for

    critical reading of the manuscript. This work was supported by the

    Fondation pour la Recherche Medicale, the VERUM foundation,

    Fondation de France and APOPIS (Abnormal proteins in the

    pathogenesis of neurodegenerative disorders an integrated project

    funded by the EU under the Sixth Framework Programme; Priority:

    Life Science for Health, contract no. LSHM-CT-2003-503330).

    References

    Balcz B., Kirchner L., Cairns N., Fountoulakis M. and Lubec G. (2001)Increased brain protein levels of carbonyl reductase and alcoholdehydrogenase in Down syndrome and Alzheimers disease.J Neural Transm Suppl. 2001; (61), 193201.

    Cadet J. L., Jayanthi S., McCoy M. T., Vawter M. and Ladenheim B.(2001) Temporal proling of methamphetamine-induced changesin gene expression in the mouse brain: evidence from cDNA array.Synapse 41, 4048.

    Castegna A., Aksenov M., Aksenova M., Thongboonkerd V., Klein J. B.,Pierce W. M., Booze R., Markesbery W. R. and Buttereld D. A.(2002a) Proteomic identication of oxidatively modied proteinsin Alzheimers disease brain. Part I: creatine kinase BB, glutaminesynthase, and ubiquitin carboxy-terminal hydrolase L-1. FreeRadic. Biol. Med. 33, 562571.

    Castegna A., Aksenov M., Thongboonkerd V., Klein J. B., Pierce W. M.,Booze R., Markesbery W. R. and Buttereld D. A. (2002b) Proteo-mic identication of oxidatively modied proteins in Alzheimersdisease brain. Part II: dihydropyrimidinase-related protein 2, alpha-enolase and heat shock cognate 71. J. Neurochem. 82, 15241532.

    Chen F., Wollmer M. A., Hoerndli F., Munch G., Kuhla B., Rogaev E. I.,Tsolaki M., Papassotiropoulos A. and Gotz J. (2004) Role forglyoxalase I in Alzheimers disease. Proc. Natl Acad. Sci. USA101, 76877692.

    Choi J., Forster M. J., McDonald S. R., Weintraub S. T., Carroll C. A.and Gracy R. W. (2004a) Proteomic identication of specicoxidized proteins in ApoE-knockout mice: relevance to Alzhei-mers disease. Free Radic. Biol. Med. 36, 11551162.

    Choi J., Levey A. I., Weintraub S. T., Rees H. D., Gearing M., Chin L. S.and Li L. (2004b) Oxidative modications and down-regulation ofubiquitin carboxyl-terminal hydrolase L1 associated with idio-pathic Parkinsons and Alzheimers diseases. J. Biol. Chem. 279,13 25613 264.

    Choi P., Snyder H., Petrucelli L. et al. (2003) SEPT5_v2 is a parkin-binding protein. Brain Res. Mol. Brain Res. 117, 179189.

    Corti O., Hampe C., Koutnikova H. et al. (2003) The p38 subunit of theaminoacyltRNA synthetase complex is a Parkin substrate: linking

    protein biosynthesis and neurodegeneration. Hum. Mol. Genet. 12,14271437.

    DekkerM. C., Bonifati V. and vanDuijn C.M. (2003) Parkinsons disease:piecing together a genetic jigsaw. Brain 126, 17221733.

    Dev K. K., van der Putten H., Sommer B. and Rovelli G. (2003) Part I:parkin-associated proteins and Parkinsons disease. Neurophar-macology 45, 113.

    Dunn A. Y., Melville M. W. and Frydman J. (2001) Review: cellularsubstrates of the eukaryotic chaperonin TRiC/CCT. J. Struct. Biol.135, 176184.

    Fallon L., Moreau F., Croft B. G., Labib N., Gu W. J. and Fon E. A.(2002) Parkin and CASK/LIN-2 associate via a PDZ-mediatedinteraction and are co-localized in lipid rafts and postsynapticdensities in brain. J. Biol. Chem. 277, 486491.

    Farrer M., Chan P., Chen R. et al. (2001) Lewy bodies and parkin-sonism in families with parkin mutations. Ann. Neurol. 50,293300.

    Forno L. S. (1987) The Lewy body in Parkinsons disease. Adv. Neurol.45, 3543.

    Friedman D. B., Hill S., Keller J. W., Merchant N. B., Levy S. E., CoffeyR. J. and Caprioli R. M. (2004) Proteome analysis of human coloncancer by two-dimensional difference gel electrophoresis and massspectrometry. Proteomics 4, 793811.

    Gharbi S., Gaffney P., Yang A., Zvelebil M. J., Cramer R., WatereldM. D. and Timms J. F. (2002) Evaluation of two-dimensionaldifferential gel electrophoresis for proteomic expression analysisof a model breast cancer cell system. Mol. Cell. Proteomics 1,9198.

    Goldberg M. S., Fleming S. M., Palacino J. J. et al. (2003) Parkin-decient mice exhibit nigrostriatal decits but not loss of dopam-inergic neurons. J. Biol. Chem. 278, 43 62843 635.

    Gouider-Khouja N., Larnaout A., Amouri R., Sfar S., Belal S.,Ben Hamida C., Ben Hamida M., Hattori N., Mizuno Y. andHentati F. (2003) Autosomal recessive parkinsonism linked toparkin gene in a Tunisian family. Clinical, genetic and pathologicalstudy. Parkinsonism Relat. Disord. 9, 247251.

    Gozal E., Gozal D., Pierce W. M., Thongboonkerd V., Scherzer J. A.,Sachleben L. R. Jr, Brittian K. R., Guo S. Z., Cai J. and Klein J. B.(2002) Proteomic analysis of CA1 and CA3 regions of rat hippo-campus and differential susceptibility to intermittent hypoxia.J. Neurochem. 83, 331345.

    Hattori N. and Mizuno Y. (2004) Pathogenetic mechanisms of parkin inParkinsons disease. Lancet 364, 722724.

    Hayashi S., Wakabayashi K., Ishikawa A., Nagai H., Saito M.,Maruyama M., Takahashi T., Ozawa T., Tsuji S. and Takahashi H.(2000) An autopsy case of autosomal-recessive juvenile parkin-sonism with a homozygous exon 4 deletion in the parkin gene.Mov. Disord. 15, 884888.

    Huynh D. P., Scoles D. R., Nguyen D. and Pulst S. M. (2003) Theautosomal recessive juvenile Parkinson disease gene product,parkin, interacts with and ubiquitinates synaptotagmin XI. Hum.Mol. Genet. 12, 25872597.

    Ihara M., Tomimoto H., Kitayama H., Morioka Y., Akiguchi I.,Shibasaki H., Noda M. and Kinoshita M. (2003) Association of thecytoskeletal GTP-binding protein Sept4/H5 with cytoplasmicinclusions found in Parkinsons disease and other synucleinopa-thies. J. Biol. Chem. 278, 24 09524 102.

    Imai Y., Soda M. and Takahashi R. (2000) Parkin suppresses unfoldedprotein stress-induced cell death through its E3 ubiquitinproteinligase activity. J. Biol. Chem. 275, 35 66135 664.

    Itier J. M., Ibanez P., Mena M. A. et al. (2003) Parkin gene inactivationalters behaviour and dopamine neurotransmission in the mouse.Hum. Mol. Genet. 12, 22772291.

    1274 M. Periquet et al.

    2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 12591276

  • Jiang H., Jiang Q. and Feng J. (2004) Parkin increases dopamine uptakeby enhancing the cell surface expression of dopamine transporter.J. Biol. Chem. 279, 54 38054 386.

    Jin J., Meredith G. E., Chen L., Zhou Y., Xu J., Shie F. S., Lockhart P.and Zhang J. (2005) Quantitative proteomic analysis of mitoch-ondrial proteins: relevance to Lewy body formation and Parkin-sons disease. Brain Res. Mol. Brain Res. 134, 119138.

    Kadota M., Nishigaki R., Wang C. C., Toda T., Shirayoshi Y., Inoue T.,Gojobori T., Ikeo K., Rogers M. S. and Oshimura M. (2004)Proteomic signatures and aberrations of mouse embryonic stemcells containing a single human chromosome 21 in neuronal dif-ferentiation: an in vitro model of Down syndrome. Neuroscience129, 325335.

    Kanninen K., Goldsteins G., Aurrola S., Alafuzoff I. and Koistinaho J.(2004) Glycosylation changes in Alzheimers disease as revealedby a promeotic approach. Neurosci. Lett. 367, 235240.

    Kim S. H., Vikolinsky R., Coiins N. and Cubec G. (2000) Decreasedlevels of complex III core protein I and complex V beta chain inbrains from patients with Alzheimers disease and Down Syn-drome. Cell. Mol. Life. Sci. 57, 18101816.

    Kitada T., Asakawa S., Hattori N., Matsumine H., Yamamura Y.,Minoshima S., Yokochi M., Mizuno Y. and Shimizu N. (1998)Mutations in the parkin gene cause autosomal recessive juvenileparkinsonism. Nature 392, 605608.

    Klein C., Pramstaller P. P., Kis B. et al. (2000) Parkin deletions in afamily with adult-onset, tremor-dominant parkinsonism: expandingthe phenotype. Ann. Neurol. 48, 6571.

    Kubo S. I., Kitami T., Noda S., Shimura H., Uchiyama Y., Asakawa S.,Minoshima S., Shimizu N., Mizuno Y. and Hattori N. (2001) Parkinis associated with cellular vesicles. J. Neurochem. 78, 4254.

    Kuhn K., Wellen J., Link N., Maskri L., Lubbert H. and Stichel C. C.(2003) The mouse MPTP model: gene expression changes indopaminergic neurons. Eur. J. Neurosci. 17, 112.

    Lohmann E., Periquet M., Bonifati V. et al. (2003) How much pheno-typic variation can be attributed to parkin genotype? Ann. Neurol.54, 176185.

    Loring J. F., Wen X., Lee J. M., Seilhamer J. and Somogyi R. (2001) Agene expression prole of Alzheimers disease. DNA Cell Biol. 20,683695.

    Lucking C. B., Durr A., Bonifati V. et al. (2000) Association betweenearly-onset Parkinsons disease and mutations in the parkin gene.French Parkinsons Disease Genetics Study Group. N. Engl.J. Med. 342, 15601567.

    Mori H., Kondo T., Yokochi M., Matsumine H., Nakagawa-Hattori Y.,Miyake T., Suda K. and Mizuno Y. (1998) Pathologic and bio-chemical studies of juvenile parkinsonism linked to chromosome6q. Neurology 51, 890892.

    Mura A., Feldon J. and Mintz M. (2000) The expression of the calciumbinding protein calretinin in the rat striatum: effects of dopaminedepletion and L-DOPA treatment. Exp. Neurol. 164, 322332.

    Napolitano M., Centonze D., Calce A., Picconi B., Spiezia S., Gulino A.,Bernardi G. and Calabresi P. (2002) Experimental parkinsonismmodulates multiple genes involved in the transduction of dopam-inergic signals in the striatum. Neurobiol. Dis. 10, 387395.

    Paisan-Ruiz C., Jain S., Evans E. W. et al. (2004) Cloning of the genecontaining mutations that cause PARK8-linked Parkinsons dis-ease. Neuron 44, 595600.

    Palacino J. J., Sagi D., Goldberg M. S., Krauss S., MotZ. C., Wacker M.,Klose J. and Shen J. (2004) Mitochondrial dysfunction and oxi-dative damage in parkin-decient mice. J. Biol. Chem. 279, 1861418 622.

    Patton W. F. (2000) A thousand points of light: the application ofuorescence detection technologies to two-dimensional gel elec-trophoresis and proteomics. Electrophoresis 21, 11231144.

    Perez F. A. and Palmiter R. D. (2005) Parkin-decient mice are not arobust model of parkinsonism. Proc. Natl Acad. Sci. USA 102,21742179.

    Periquet M., Latouche M., Lohmann E. et al. (2003) Parkin mutationsare frequent in patients with isolated early-onset parkinsonism.Brain 126, 12711278.

    Prabakaran S., Swatton J. E., Ryan M. M. et al. (2004) Mitochondrialdysfunction in schizophrenia: evidence for compromised brainmetabolism and oxidative stress. Mol. Psychiatry 9, 643.

    Ren Y., Zhao J. and Feng J. (2003) Parkin binds to alpha/beta tubulin andincreases their ubiquitination and degradation. J. Neurosci. 23,33163324.

    Ryu E. J., Harding H. P., Angelastro J. M., Vitolo O. V., Ron D. andGreene L. A. (2002) Endoplasmic reticulum stress and the unfol-ded protein response in cellular models of Parkinsons disease.J. Neurosci. 22, 10 69010 698.

    Schonberger S. J., Edgar P. F., Kydd R., Faull R. L. and Cooper G. J.(2001) Proteomic analysis of the brain in Alzheimers disease:molecular phenotype of a complex disease process. Proteomics 1,15191528.

    Seong J. K., Kim do K., Choi K. H., Oh S. H., Kim K. S., Lee S. S. andUm H. D. (2002) Proteomic analysis of the cellular proteinsinduced by adaptive concentrations of hydrogen peroxide in humanU937 cells. Exp. Mol. Med. 34, 374378.

    Shimura H., Hattori N., Kubo S. et al. (2000) Familial Parkinson diseasegene product, parkin, is a ubiquitinprotein ligase. Nat. Genet. 25,302305.

    Singleton A. B., Farrer M., Johnson J. et al. (2003) Alpha-synucleinlocus triplication causes Parkinsons disease. Science 302, 841.

    Swatton J. E., Prabakaran S., Karp N. A., Lilley K. S. and Bahn S.(2004) Protein proling of human postmortem brain using2-dimensional uorescence difference gel electrophoresis (2-DDIGE). Mol. Psychiatry 9, 128143.

    Tilleman K., Stevens I., Spittaels K., Haute C. V., Clerens S., Van DenBergh G., Geerts H., Van Leuven F., Vandesande F. and Moens L.(2002a) Differential expression of brain proteins in glycogensynthase kinase-3 transgenic mice: a proteomics point of view.Proteomics 2, 94104.

    Tilleman K., Van den Haute C., Geerts H., van Leuven F., Esmans E.L. and Moens L. (2002b) Proteomics analysis of the neuro-degeneration in the brain of tau transgenic mice. Proteomics 2,656665.

    Tonge R., Shaw J., Middleton B., Rowlinson R., Rayner S., Young J.,Pognan F., Hawkins E., Currie I. and Davison M. (2001) Validationand development of uorescence two-dimensional differential gelelectrophoresis proteomics technology. Proteomics 1, 377396.

    Tsuboi K., Kimber T. A. and Shults C. W. (2000) Calretinin-containingaxons and neurons are resistant to an intrastriatal 6-hydroxydop-amine lesion. Brain Res. 866, 5564.

    Tsuji T., Shiozaki A., Kohno R., Yoshizato K. and Shimohama S. (2002)Proteonic proling and neurodegeneration in Alzheimers disease.Neurochem. Res. 27, 12451253.

    Valente E. M., Abou-Sleiman P. M., Caputo V. et al. (2004) Hereditaryearly-onset Parkinsons disease caused by mutations in PINK1.Science 304, 11581160.

    Von Coelln R., Thomas B., Savitt J. M., Lim K. L., Sasaki M., Hess E. J.,Dawson V. L. and Dawson T. M. (2004) Loss of locus coeruleusneurons and reduced startle in parkin null mice. Proc. Natl Acad.Sci. USA 101, 10 74410 749.

    van de Warrenburg B. P., Lammens M., Lucking C. B., Denee P.,Wesseling P., Booij J., Praamstra P., Quinn N., Brice A. andHorstink M. W. (2001) Clinical and pathologic abnormalities in afamily with parkinsonism and parkin gene mutations. Neurology56, 555557.

    Proteomic analysis of parkin knockout mice 1275

    2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 12591276

  • Xie T., Tong L., Barrett T., Yuan J., Hatzidimitriou G., McCann U. D.,Becker K. G., Donovan D. M. and Ricaurte G. A. (2002) Changesin gene expression linked to methamphetamine-induced dopam-inergic neurotoxicity. J. Neurosci. 22, 274283.

    Yan J. X., Devenish A. T., Wait R., Stone T., Lewis S. and Fowler S.(2002) Fluorescence two-dimensional difference gel electrophor-esis and mass spectrometry based proteomic analysis of Escheri-chia coli. Proteomics 2, 16821698.

    Zabel C., Chamrad D. C., Priller J., Woodman B., Meyer H. E., Bates G.P. and Klose J. (2002) Alterations in the mouse and human prot-

    eome caused by Huntingtons disease. Mol. Cell. Proteomics 1,366375.

    Zhang Y., Gao J., Chung K. K., Huang H., Dawson V. L. and Dawson T.M. (2000) Parkin functions as an E2-dependent ubiquitinproteinligase and promotes the degradation of the synaptic vesicle-asso-ciated protein, CDCrel-1. Proc. Natl Acad. Sci. USA 97, 13 35413 359.

    Zimprich A., Biskup S., Leitner P. et al. (2004) Mutations in LRRK2cause autosomal-dominant parkinsonism with pleomorphicpathology. Neuron 44, 601607.

    1276 M. Periquet et al.

    2005 International Society for Neurochemistry, J. Neurochem. (2005) 95, 12591276