Mass Spectrometric Studies on Mouse Hippocampal Synapsins Ia, IIa

Julius Paul Pradeep John, Wei-Qiang Chen, Arnold Pollak, and Gert Lubec*. Department of Pediatrics, Medical University of Vienna, Waehringer Guertel 1...
0 downloads 0 Views 606KB Size
Mass Spectrometric Studies on Mouse Hippocampal Synapsins Ia, IIa, and IIb and Identification of a Novel Phosphorylation Site at Serine-546 Julius Paul Pradeep John, Wei-Qiang Chen, Arnold Pollak, and Gert Lubec* Department of Pediatrics, Medical University of Vienna, Waehringer Guertel 18, 1090 Vienna, Austria Received March 21, 2007

Synapsins are key phosphoproteins in the mammalian brain, and structural research on synapsins is still holding center stage. Proteins were extracted from hippocampal tissue and separated on twodimensional gel electrophoresis (2-DE), and the spots were analyzed by MALDI-TOF-TOF and nanoLC-ESI-MS/MS. Synapsins Ia, IIa, and IIb were unambiguously identified and represented by 15 individual spots on 2-DE. Several serine phosphorylation sites were confirmed, and a novel phosphorylation site was observed at Ser-546 in synapsin IIa in all gels analyzed. Keywords: database conflict • de novo sequencing • mass spectrometry • phosphorylation • synapsin isoforms

Introduction Synapsins are a family of phosphoproteins specially associated with the cytoplasmic surface of the synaptic vesicle membrane.1 Synapsins are expressed in vertebrates, and three genes (synapsins 1, 2, and 3) are observed in mammals.2-4 In vertebrates, synapsins constitute a family of at least five homologous proteins (synapsins Ia, Ib, IIa, IIb, and IIIa) that are derived by alternative splicing from the primary transcript gene.5,6 All the synapsin isoforms exhibit a high degree of sequence homology in their amino terminal regions (the short A- and B-domains and long C-domain) and divergent carboxy terminals that are present in different combinations in each of the synapsins.2 The small carboxy terminal specific to the b-type is present in synapsin isoforms (domain F).7 Synapsins represent one of the abundant families of synaptic proteins, comprising approximately 1% of the total proteins in the brain.8-10 The functional behavior of the synapsins is affected by their phosphorylation states, which are regulated by a variety of physiological and pharmacological messengers that efficiently modify the synaptic function.11 Synapsins are major substrates for multiple protein kinases, including cAMP-dependent protein kinase (PKA), Ca2+/calmodulin protein kinases (CaM kinases) I, II, and IV, and mitogen-associated protein kinase/Erk 1/2 (MAPK/Erk). The presence of phosphorylation sites varies from one synapsin isoform to another.12-14 All the synapsins share consensus sequences for PKA and CaM kinase I, which phosphorylate a serine residue in the amino terminal region (Ser-9). Synapsin I is an excellent substrate for CaM kinase II, which phosphorylates two serine residues in the carboxy terminal (Ser-566 and Ser-603).2 The biological properties of synapsins have been shown by their ability to interact with the synaptic vesicle and the cytoskeleton, actin.15-19 Several reports indicated that synapsins are required to sustain neu* To whom correspondence should be addressed. Phone: +43-1-404003215. Fax: +43-1-40400-3200. E-mail: [email protected]. 10.1021/pr070157r CCC: $37.00

 2007 American Chemical Society

rotransmitter release during high levels of neuronal activity.20-27 This indicates that synapsins participate in neurotransmitter release by regulating a reserve pool of vesicles. Moreover, synapsins are also involved in regulation of the kinetics of release processes.24 Synapsin II is involved in synapse formation during nerve cell growth.28-30 Phosphorylation of synapsin II regulates binding of both the synaptic vesicle and actin filaments.12 In knock-out mouse analysis it was shown that synapsins are involved in maintaining stable vesicles and are required for normal short-term plasticity, suggesting that they constitute important regulatory molecules.22,31,32 Recently, contradictory to the above finding, it was shown that synapsins are not required for normal vesicle exocytosis and recycling.32 Synapsin regulates synaptic plasticity by a Ca2+-dependent pathway in the calyx of the Held synapse.33 Synapsin isoforms exhibit a significant change of heterogenicity in their protein expressional state. Li and co-workers observed the presence of different synapsin splice variants in two-dimensional gel electrophoresis (2-DE) gels, resolved by different pI values and molecular weights.34 The nature of many post-translational modifications (PTMs) associated with synapsin function has been reported. There are several phosphorylation sites confirmed in synapsin I by mass spectrometry (MS) analysis in mammals.35-39 In addition to that, a number of O-linked N-acetylglucosamine (O-GlcNAc) modification sites in synapsin I were identified.40-42 Murrey et al. reported that fucosylation modulates synapsin Ia and Ib expression using synaptic vesicle purification, immunoblots, and gel electrophoresis combined with MS analysis.43 Knowledge of PTMs in synapsins IIa and IIb is very limited, due to the problems associated with protein purification. Zhu et al. reported that synapsin I and synapsin II are substrates for protein Lisoaspartyl methyltransferase in mouse brain using a proteomic approach.44 A recent global phosphoproteomic study on mouse Journal of Proteome Research 2007, 6, 2695-2710

2695

Published on Web 06/19/2007

research articles synaptosomal preparations indicated that synapsin II is phosphorylated at Thr-422 and Ser-426;45 however, the subtype of synapsin II was not defined because in their setting the authors did not identify the isoforms but rather focused on identification of phosphopeptides exclusively. Herein we decided to carry out qualitative studies on phosphorylation of synapsins, with and without phosphorylation enrichment of phosphopeptides by IMAC taken from 2-DE spots, combined with mandatory multiple MS analysis, thus using an appropriate method to determine PTMs on individual synapsin isoforms. To our knowledge, no reports on the heterogenicity of synapsin isoforms in the mouse hippocampus are available, and the issue of identifying several isoforms on a 2-DE gel is a major challenge. The aim of this study was to investigate synapsin isoforms with regard to changes of molecular heterogenicity using 2-DE followed by mass spectrometry. Combination of high-resolution 2-DE with two different MS principles is a reliable method to determine synapsin isoform modifications of sequences and post-translational modifications. Characterization of synapsin isoforms by MALDI-TOFTOF following multienzyme cleavage of synapsins from the mouse hippocampus revealed 15 synapsin expression forms, and nano-LC-ESI-MS/MS following enrichment of phosphopeptides identified phosphorylation sites that may form the basis and tools required for future studies on synapsins at the protein level.

Materials and Methods A. Animals. The subjects were C57BL/6JHim (C57), male, adult mice, approximately 20 weeks old. The animals were housed in standard transparent laboratory cages in a temperature-controlled colony room (22 ( 1 °C). They were maintained on a 12 h artificial light/dark cycle (with lights on at 6 a.m.) and provided with food and water ad libitum. The mice were sacrificed by neck dislocation, and the hippocampi were dissected. Tissue samples were immediately frozen in liquid nitrogen and stored at -80 °C until used for analysis. All experiments on animals were approved by the local animal committee (confirmation number LF1-TVG-17/002-2004). B. Sample Preparation. Hippocampal tissue was powderized and resuspended in 1.0 mL of sample buffer consisting of 7 M urea (Merck, Darmstadt, Germany), 2 M thiourea (Sigma, St. Louis, MO), 4% 3-[(3-cholamidopropyl)dimethylammonio]-1propanesulfonate (CHAPS) (Sigma), 65 mM 1,4-dithioerythritol (Merck, Germany), 1 mM ethylenediaminetraacetic acid (EDTA) (Merck), protease inhibitors complete (Roche, Basel, Switzerland), phosphatase inhibitor cocktail set III (Merck), and 1 mM phenylmethanesulfonyl chloride. The suspension was sonicated for approximately 15 s. After homogenization the samples were left at 22 °C for 1 h and centrifuged at 14000g for 1 h. The supernatant was transferred into Ultrafree-4 centrifugal filter units (Millipore, Bedford, MA) for desalting and concentrating proteins.46 The protein content of the supernatant was determined by the Bradford protein assay system.47 The standard curve was generated using bovine serum albumin, and the absorbance was measured at 595 nm. C. Two-Dimensional Gel Electrophoresis. Protein extracts prepared from each of 10 mice were subjected to 2-DE as described elsewhere.48 A 700 µg portion of hippocampal extracted protein was applied on immobilized pH 3-10 nonlinear gradient strips at their basic and acidic ends. Focusing was started at 200 V, and the voltage was gradually increased to 8000 V over 31 h and then kept constant for a further 3 h 2696

Journal of Proteome Research • Vol. 6, No. 7, 2007

John et al.

(approximately 150000 V h total). After the first dimension, the strips (18 cm) were equilibrated for 15 min in buffer containing 6 M urea, 20% glycerol, 2% SDS, 2% dithiothreitol (DTT) and then for 15 min in the same buffer containing 2.5% iodoacetamide instead of DDT. After equilibration, the strips were loaded onto 9-16% gradient sodium dodecyl sulfate-polyacrylamide gels for two-dimensional separation. The gels (180 mm × 200 mm × 1.5 mm) were run at 40 mA per gel. Immediately after the second dimension run, the gels were fixed for 18 h in 50% methanol, containing 10% acetic acid; the gels were then stained with colloidal Coomassie Blue (Novex, San Diego, CA) for 12 h on a rocking shaker. The molecular masses were determined by running standard protein markers (Biorad Laboratories, Hercules, CA) covering the range 10-250 kDa. pI values of 3-10 were used as given by the supplier of the immobilized pH gradient strips (Amersham Bioscience, Uppsala, Sweden). Excess dye was washed from the gels with distilled water, and the gels were scanned with an ImageScanner (Amersham Bioscience). Electronic images of the gels were recorded using Adobe Photoshop. D. Method for MALDI-TOF-TOF Analysis. 1. Sample Preparation. Gel spots identified as synapsins in parallel experiments by MALDI-TOF-TOF were excised manually and washed with 10 mM ammonium bicarbonate and 50% acetonitrile (ACN) in 10 mM ammonium bicarbonate. After washing, the gel plugs were shrunk by addition of ACN and dried. The dried gel pieces were reswollen with 40 ng/µL trypsin (Promega, Madison, WI) in digestion buffer (consisting of 5 mM octyl β-D-glucopyranoside (OGP) and 5 mM ammonium bicarbonate) and incubated for 4 h at 30 °C: Asp-N digests were performed by addition of 25 mM ammonium bicarbonate containing 25 ng/ µL Asp-N (sequencing grade, Roche Diagnostic, Mannheim, Germany). Lys-C digests were performed by addition of 5 mM ammonium bicarbonate containing 30 ng/µL Lys-C (sequencing grade, Roche Diagnostic) and incubated for 18 h at 31 °C. Extraction was performed with 10 µL of 1% TFA in 5 mM OGP. 2. Protein Identification by MALDI-TOF-TOF. Extracted peptides were directly applied onto a target (AnchorChip, Bruker Daltonics, Bremen, Germany) that was loaded with an R-cyano-4-hydroxycinnamic acid (Bruker Daltonics) matrix thin layer (saturated solution in 100% acetone with 0.1% TFA). The mass spectrometer used in this work was an Ultraflex TOFTOF instrument (Bruker Daltonics) operated in positive ion reflector mode for peptide mass analysis, and the “LIFT” mode was used for tandem mass spectrometry sequencing of peptides using the FlexControl 2.4 software (Bruker Daltonics). An accelerating voltage of 25 kV was used for PMF. Calibration of the instrument was performed externally with [M + H]+ ions of angiotensin I, angiotensin II, substance P, bombesin, and adrenocorticotropic hormones (clips 1-17 and clips 18-39). Each spectrum was produced by accumulating data from 200 consecutive laser shots for PMF. Those samples which were analyzed by PMF from MALDI-TOF were additionally analyzed using LIFT-TOF-TOF-MS/MS from the same target using the LID mode.49 In the LID-MS/MS mode using a long-lifetime N2 laser, all ions were accelerated to 8 kV under conditions promoting metastable fragmentation in the TOF1 stage. After selection of jointly migrating parent and fragment ions in a timed ion gate, the ions were lifted by 19 kV to high potential energy in the LIFT cell. After further acceleration of the fragment ions in the second ion source, their masses could be simultaneously analyzed in the reflector with high sensitivity. Mass spectra were analyzed using the FlexAnalysis 2.4 software.

Mass Spectrometrical Studies on Synapsin Isoforms

PMF and MS/MS datasets were interpreted with MASCOT (Matrix Science Ltd., London, U.K.) software searched against the MSDB 20051115 database (ftp://ftp.ncbi.nih.gov/repository/MSDB/) via BioTools 2.2 software (Bruker Daltonics). A mass tolerance of 25 ppm and MS/MS tolerance of 0.5 Da and one missing cleavage site were allowed. The probability score calculated by the software was used as a criterion for correct identification. Oxidation of methionine and deamidation of Asn and Gln residues were set as variable modifications, and carbamidomethylation of cysteine residues was set as a fixed modification. Mascot results were confirmed manually. Unmatched MS/MS spectra were further analyzed by de novo sequencing analysis. MS/MS spectra were sequenced de novo using BioTool 2.2 software with a RapiDeNovo extension (Bruker Daltonics49), and the top high-scoring candidate sequences for MS/MS spectra were then submitted to an MS BLAST sequence similarity search (http://dove.embl-heidelberg.de/Blast2/ msblast.html), which was based on the most likely de novo sequences. Candidate sequences were edited according to MS BLAST conventions and submitted to MS BLAST searches performed against the nucleic acid database at http://genetics.bwh.harvard.edu/msblast/ under the following settings: scoring table, 99; filter, none; expect, 1000. The statistical significance of hits was evaluated according to the MS BLAST scoring scheme.50 E. Method for Nano-LC-ESI-MS/MS Analysis. 1. Sample Preparation. The gel spots were excised, cut into smaller pieces, and washed with ultrapure water. Coomassie-stained gel bands were destained by several washing steps with 20 mM ammonium bicarbonate and ACN (1:1). The gel pieces were dehydrated by adding ACN. After 5 min of incubation, ACN was removed and the gel fragments were dried in the SpeedVac. Dry gel pieces were incubated for 30 min at 56 °C in 200 µL of 10 mM DTT to reduce disulfide bonds. DTT was washed off, and cysteines were alkylated by incubation with 100 µL of 54 mM iodoacetamide for 20 min at room temperature in the dark. After the washing steps with 50 mM ammonium bicarbonate, the gel was dried and incubated for 4 h at 29 °C with 40 ng/µL trypsin (Promega, Madison, WI) in 50 mM ammonium carbonate buffer, pH 8.5. Asp-N digests were performed by addition of 25 mM ammonium bicarbonate containing 25 ng/ µL Asp-N (sequencing grade, Roche Diagnostic). Lys-C digests were performed by addition of 5 mM ammonium bicarbonate containing 30 ng/µL Lys-C (sequencing grade, Roche Diagnostic) and incubated for 18 h at 31 °C. Reaction was stopped by adding 10% formic acid (FA) to a final concentration of ∼1%. After 10 min of sonication, the supernatant was taken off and the peptides were further extracted by adding approximately 20 µL of 5% FA followed by 10 min of sonication (the procedure was repeated once). 2. Sample Preparation for Subtilisin Digestion. Subtilisin (proteinase from Bacillus subtilis var. biotecus A, Fluka) was dissolved at a concentration of 10 µg/µL in 1 mM HCl and diluted with 6 M urea and 1 M Tris, pH 8.5, at a final concentration of 100 ng/µL. The gel pieces were covered with this concentration and rehydrated for 10 min at 4 °C. The supernatant was removed and replaced by 50 mM ammonium bicarbonate. Digestion was performed for 1 h at 37 °C on a shaker. Reaction was stopped by addition of 10% FA to a final concentration of ∼1%. After 10 min of sonication, the supernatant was taken off and the peptides were further extracted by adding approximately 20 µL of 5% FA followed by 10 min of sonication (the procedure was repeated once).

research articles 3. Protein Identification by Nano-LC-ESI-MS/MS. The peptide mixture obtained from multiproteolytic digests was separated by an UltiMate 3000 dual nano-LC system (Dionex) and analyzed using QSTAR XL (Applied Biosystems, Foster City, CA) equipped with a nano electrospray ionization source. For nano-LC-ESI-MS/MS, the digest was loaded onto a PepMap 100 C18 precolumn (300 µm i.d., 5 mm long cartridge, from Dionex, Amsterdam, The Netherlands) from 0 to 30 min and then separated by a PepMap 100 C18 analytical column (75 µm i.d., 150 mm long cartridge, from Dionex) using a linear gradient of 4% B (solvent A, 0.1% FA; solvent B, 80% ACN/0.08% FA) to 60% B from 0 to 30 min, 90% B constant from 30 to 35 min, and 4% B from 35 to 60 min using the Ultimate micropump at a flow rate of 300 nL/min. As the peptides eluted from the LC system, they were electrosprayed into QSTAR XL. Each cycle consisted of one full scan mass spectrum (m/z 350-1600) followed by MS/MS spectra on the three most abundant peptide ions in the full MS scan. The derived MS datasets were converted to MASCOT generic format flat files by a mascot.dll 1.6b21 (Matrix Science, Boston, MA) script supplied with the AnalystQS 1.1 software (Applied Biosystems) and searched against in-house-licensed MSDB 20051115 and Expasy/TrEMBL databases.51 Multiple forms of PTMs were considered in the search: phosphorylation of serine and threonine, deamidation of Gln and Asn, N-term acetylation. The PHENYX software platform (version 2.0) from GenBio (Geneva, Switzerland) was used to confirm the peptide identification and modification. The raw MS/MS data were converted to a .mgf file format and submitted to PHENYX. Database searches were carried out against the Swiss-Prot/ UniProt database. Top-scoring candidate peptides were manually validated with MASCOT results. F. Protein Identification by LTQ-FT. The peptides were separated on an UltiMate 3000 dual nano-LC system (Dionex). After being desalted for 18 min on a precolumn (PepMAP C18, 0.3 × 5 mm, Dionex) (0.1% TFA, 20 µL/min), the peptides were eluted with a gradient from 0% to 50% B in 90 min and a flow rate of 300 nL/min onto an analytical column (PepMAP C18, 75 µm × 150 mm, Dionex) (solvent A, 5% ACN, 0.1% formic acid; solvent B, 80% ACN, 0.08% formic acid). The nano-HPLC system was coupled online to a hybrid linear ion trap/FT-ICR mass spectrometer (LTQ-FT, Thermo Electron, Bremen, Germany) with a 7 T superconducting magnet via a nano electrospray ionization source (Proxeon Biosystems, Odense, Denmark). The two detectors of the hybrid instrument were operated in parallel mode, during one full scan in the FT-ICR cell (resolution 100000, m/z 300-1800). The five most abundant ions were subjected to MS/MS and neutral loss triggered MS3 analysis in the linear ion trap. Fragmented precursors were excluded from further fragmentation for 60 s (with 5 ppm mass accuracy), and singly charged peptides were generally excluded from MS/MS analysis. Data analysis was performed using the MASCOT (version 2.1.0) software, searching against the NCBI nucleic acid database. G. IMAC Enrichment Using a Pierce Phosphopeptide Isolation Kit. Gallium-chelated IMAC minispin columns (Pierce, Rockford, IL) were used according to the manufacturer’s recommendation with the following modifications. Extracted peptides were evaporated in the Speed-Vac and suspended in 20 µL of 5% acetic acid as the binding buffer; the trypsin- and Asp-N-digested peptides were transferred to columns, left for 30 min at 22 °C, and centrifuged for 1 min at 3000g. The gallium-chelated columns were rinsed once with 50 µL of 0.1% Journal of Proteome Research • Vol. 6, No. 7, 2007 2697

research articles

John et al.

Figure 1. Structure of the synapsins. The individual and shared domains of the five isoforms are composed of domains A-C. In the COOH terminal variable region, the proline- and glycine-rich D domain is shared by synapsins Ia and Ib, the proline-rich G domain is shared by synapsins IIa and II b, and the COOH terminal domain E is present in synapsin Ia, synapsin IIa, and isoform III.

(v/v) acetic acid, twice with 50 µL of 100 mM NaCl in 1% acetic acid, twice with 30% (v/v) ACN in 1% acetic acid, and once with water. The phosphopeptides were then eluted by three additions of 20 µL of 100 mM ammonium bicarbonate. The eluates were pooled and evaporated in the Speed-Vac, the phosphopeptides were suspended in 20 µL of 2% ACN containing 0.1% TFA, and the volume was reduced to 5 µL before nanoLC-ESI-MS/MS analysis. H. Phosphatase Treatment. Trypsin-digested synapsin IIa samples were subjected to phosphatase treatment. The peptide samples were incubated in a solution of 0.5 µL of calf intestine alkaline phosphatase (New England Biolabs, Ipswich, MA) in the presence of 100 mM ammonium bicarbonate for 1 h at 37 °C. The samples were subsequently analyzed by MALDI-MS. I. Protein Quantification. The protein spots were outlined (first automatically and then manually) and quantified using the Proteomweaver software (Definiens, Munich, Germany). The percentage of the volume of the spots representing a certain protein was determined in comparison with the volume of the total proteins present in the 2-DE gel. The values are expressed as means ( standard deviations (SDs) of the percentage of the spot volume in each particular gel after subtraction of the background values.46 J. Bioinformatics Analysis. Protein sequences derived from the NCBI protein database were used for protein sequence analysis. ClustalW (http://www.ebi.ac.uk/clustalw/) was used for multiple sequence alignment searches. ELM (http:// elm.eu.org) was used for functional domain identification.

and post-translational modifications, a mass spectrometry approach was carried out. The protein spots were excised and digested individually with trypsin, Lys-C, and Asp-N and analyzed with MALDI-TOF-TOF, nano-LC-ESI-MS/MS, and LTQ-FT.

Results

The mass spectrometric data of each splicing isoform were compared to identify each synapsin isoform. Synapsin Ia was identified in spots 1 and 2 (Figure 2, Table 1). The PMF map indicated that the tryptic peptide SQSLTNAFNLPEPAPPRPSLSQDEVK (amino acid residues 664-689), which covered the splicing site in synapsin Ia, has an [M + H]+ ion at m/z 2822.38 and was identified as its deamidated form in spot 2 (Figure 2, Table 2). Nano-LC-ESI-MS/MS analysis of Lys-C-digested peptides indicated that peptide [M + 2H]2+ at m/z 952.36, SFASLFSD (amino acid residue 699-776), which covered the splicing site in synapsin Ia, was identified in spot 1 (Table 2). MALDI-TOF-TOF analysis of the tryptic peptide SQSLTNAFSFSESSFFR (amino acid residues 544-560), which covered the splicing site of synapsin IIa, has an [M + H]+ ion at m/z 1941.490 and was identified in spots 3-9 (Figures 2 and 7 (upper panel), Table 2). Moreover, nano-LC-ESI-MS/MS analysis of tryptic peptides indicated that the C-terminal of the synapsin IIa isoform was identified in all the synapsin IIa protein spots. Nano-LC-ESI-MS/MS and LTQ-FT analysis of Asp-N-digested peptides indicated that the peptide [M + 2H]2+ at m/z 772.87, DCNGIAVGPKQVQAS (amino acid numbers 465-479), covering the splicing site of synapsin IIb, was identified in spots 10-15 with a varying degree of deamidated forms in the individual protein spots (Figure 2, Table 2).

A. Analysis of the Synapsin Splicing Isoform. Alternative splicing of synapsin transcripts encodes five known synapsin isoforms: Ia, Ib, IIa, IIb, and IIIa. The differences in the amino acid sequences among these five splicing isoforms are shown in Figure 1. By protein profiling of mouse hippocampal tissue, 15 individual protein spots identified as synapsin I and synapsin II with different molecular weights and pI values were observed (Figure 2). To determine the sequence of synapsin isoforms

Data obtained from MS indicated that an average of 69% sequence coverage was obtained in both synapsin Ia protein spots (spots 1 and 2). In the case of synapsin IIa (spots 3-9) and synapsin IIb (spots 10-15), averages of 78% and 77% were identified with both the MS and MS/MS approaches. In particular, 94% sequence coverage was achieved in spot 12 alone, among which 88% of the sequence was determined by MS/MS analysis (Table 1).

2698

Journal of Proteome Research • Vol. 6, No. 7, 2007

research articles

Mass Spectrometrical Studies on Synapsin Isoforms

Figure 2. Proteins from the mouse hippocampus were subjected to isoelectrofocusing on a 18 cm immobilized pH 3-10 gradient strip and then separated by SDS-PAGE. A 2D gel and partial image of a Coomassie Blue-stained gel image are shown. Synapsin protein spots (marked with arrows) are identified and characterized by mass spectrometry. By protein profiling of mouse hippocampal tissue, 15 individual protein spots identified as synapsin I, synapsin IIa, and synapsin IIb with different molecular weights and pI values were observed in all the 2-DE gels in the same pattern. Table 1. Characterization of Synapsin Isoforms

spot Swiss-Prot no. no.

sequence coverage (%)

protein name

exptl pI,Mr

theor pI, Mr

O88935 O88935 Q64332 Q64332 Q64332

synapsin Ia synapsin Ia synapsin IIa synapsin IIa synapsin IIa

7.86, 82140 7.86, 82140 9, 72100 8.72, 72100 8.16, 72100

9.81, 74097.31 9.81, 74097.31 8.59, 63372.55 8.59, 63372.56 8.59, 63372.57

0.256 ( 0.042 0.233 ( 0.019 0.171 ( 0.048 0.216 ( 0.083 0.306 ( 0.094

62 44 61 70 71

39 13 38 45 44

Q64332 Q64332 Q64332 Q64332 Q64332-2 Q64332-2 Q64332-2

synapsin IIa synapsin IIa synapsin IIa synapsin IIa synapsin IIb synapsin IIb synapsin IIb

7.93, 72100 7.66, 72100 7.3, 72100 7.1, 72100 8.53, 52720 8.16, 52720 7.86, 52720

8.59, 63372.58 8.59, 63372.59 8.59, 63372.60 8.59, 63372.61 7.61, 52451.93 7.61, 52451.94 7.61, 52451.95

0.283 ( 0.084 0.238 ( 0.056 0.233 ( 0.089 0.182 ( 0.081 0.136 ( 0.029 0.281 ( 0.078 0.345 ( 0.050

75 78 66 65 79 85 94

45 41 43 49 42 70 88

7.61, 52451.96

0.541 ( 0.086

81

51

14 Q64332-2 synapsin IIb 6.99, 52720 7.61, 52451.97 15 Q64332-2 synapsin IIb 6.69, 52720 7.61, 52451.98

0.511 ( 0.131 0.389 ( 0.144

79 44

22 7

1 2 3 4 5 6 7 8 9 10 11 12

13 Q64332-2 synapsin IIb 7.4, 52720

av normalizedspot vol (mean ( SD) total MS/MS

PTMsa

Pyro Glu-Q404, Ox-W126(S) P-S9a, P-S62a, Pyro Glu-Q404 Ac-M1a, Ac-M2a Ac-M1a, Ac-M2a, P-S10a, P-S546†, Me-E148 Ac-M1, Ac-M2a, P-S10a, P-S546†, P-T422, P-S426, Me-E249 Pyro Glu-Q94, Ox-W127, Ox-W336 Ac-M1, Ac-M2a, P-S10a, P-S546†, Pyro Glu-Q94, Me-E249 Ac-M1a, Ac-M2a, Ox-W127(S), P-S10a, P-S546† P-S546† P-S546† Ac-M1a, Ac-M2a, Me-E148, Me-E402 Ac-M1, Ac-M2a, P-S10a, Ox-W127(D), Ox-W236(D) Ac-M1, Ac-M2, P-S10a, P-Ser-426, Me-E148, Me-E122, Pyro Glu-Q94, Ox-W127(D,S) Ac-M1a, Ac-M2a, P-S10a, P-Ser-426, Me-E151, Pyro Glu-Q94, Ox-W127(D) Ac-M1a, Ac-M2a, P-S10a, P-Ser-426, T-422, Pyro Glu-Q207 nil

a Key: superscript Roman a, modification only identified with PMF data; Ac, N-acetylation; P, phosphorylation; Me, methylation; Pyro Glu, modification of N-terminal glutamate; Ox, oxidation of tryptophan (D, doubly oxidized; S, singly oxidized); †, novel phosphorylation site Ser-546 identified in all the synapsin IIa protein spots (spots 4-9) except protein spot 3.

B. Expressional Pattern of Synapsin Isoforms. The differences between the individual synapsin isoform proteins in the 2-DE gels were quantitatively analyzed. The average normalized spot volume of each spot is shown in Table 1. The order of the proportions of spot volumes of the 15 2-DE gel spots that contained synapsin isoforms was (spot numbers) 13 > 14 > 15 > 12 > 5 > 6 > 11 > 1 > 7 > 2 > 8 > 4 > 9 > 3 > 10.

C. Structural Characterization of a Polyserine Region in Synapsin IIa. The primary structure of synapsin IIa contains long stretches of 13 serine residues (486-499); neither multiproteolytic nor MS approaches could cover this region. To address this ambiguity, spot 4 and spot 5 were excised and digested with nonspecific enzyme subtilisin and analyzed with nano-LC-ESI-MS/MS analysis, and subsequent search analysis Journal of Proteome Research • Vol. 6, No. 7, 2007 2699

research articles

John et al.

Table 2. Identification of Synapsin Isoforms synapsin isoform

peptide that covers the splicing sitea

synapsin Ia synapsin Ia

579SFASLFSD586

synapsin IIa

449TPPQRPPPQGGPGQPQGMQPPGK472

664SQSLTNAFNLPEPAPPRPSLSQDEVK689, a

observed m/z

Mr(calcd)

Mr(exptl)



charge state

437.25 2823.39

872.48 2822.38

872.39 2822.41

0.09 -0.03

[M + 2H]2+ [M + H]+

1 2

L T

E M

2351.16

2350.16

2350.19

-0.03

[M + H]+

3-9

T

M

deamidation (NQ) synapsin IIa synapsin IIa synapsin IIa synapsin IIa synapsin IIa synapsin IIa synapsin IIa synapsin IIb synapsin IIb synapsin IIb synapsin IIb synapsin IIb synapsin IIb synapsin IIb synapsin IIb synapsin IIb

deamidation (NQ), oxidation (M) 477RLPSGPSLPS486 477RLPSGPSLPSS487 477RLPSGPSLPSSS488 544SQSLTNAFSFSESSFFR560 561SSANEDEAKAETIR574 565DEAKAETIRSLRKSFASLFSD585 571ETIRSLRKSFASLFS585, a 445DSSKTPPQRPPPQGCbLQYIL464 449TPPQRPPPQGCbLQYILDCbNGIAVGPK474 deamidation (NQ)a 449TPPQRPPPQGCbLQYILDCbNGIAVGPK474 2 deamidations (NQ) 449TPPQRPPPQGCbLQYILDCbNGIAVGPK474 4 deamidations (NQ)a 465DCbNGIAVGPKQVQAS479 465DCbNGIAVGPKQVQAS479 deamidation 467NGIAVGPK474 deamidation (NQ) 467NGIAVGPKQVQAS479 deamidation (NQ) 467NGIAVGPKQVQAS479 2 deamidations (NQ)

spot no.

enzymeb

MSc

505.82 549.34 592.86 1941.90 507.59 2371.21 1742.01 1141.85 2877.48

1009.63 1096.66 1183.70 1940.89 1519.76 2370.21 1741.00 2281.69 2876.47

1009.56 1096.59 1183.62 1940.89 1519.71 2370.22 1740.95 2281.15 2876.43

0.07 0.08 0.08 -0.00 0.05 -0.01 0.05 0.54 0.04

[M + [M + 2H]2+ [M + 2H]2+ [M + H]+ [M + 2H]2+ [M + H]+ [M + H]+ [M + 2H]2+ [M + H]+

4, 5 5 5 3-9 3-9 5 5 10, 11, 13 10-15

S S S T T A A A T

E E E M E M E E M

2878.33

2877.32

2877.41

-0.10

[M + H]+

10, 11

T

M

[M +

9

T

M

11 10 13 10, 12 12

A A U A A

E LF E E, LF LF

2880.46

2879.46

2879.38

0.07

773.17 772.87 756.40 635.34 635.84

1544.32 1543.73 756.40 1268.68 1269.66

1542.75 1543.73 756.42 1268.67 1269.66

1.58 0 -0.02 0 0

2H]2+

H]+

[M + 2H]2+ [M + 2H]2+ [M + H]+ [M + 2H]2+ [M + 2H]2+

a Letters in bold indicate the exact site of the amino acid modified with deamidation determined by MS/MS analysis. Key: superscript Roman a, sequence obtained by PMF data; superscript Roman b, peptides modified with carbamidomethyl. b Key: T, trypsin; L, Lys-C; A, Asp-N; U, unspecific cleavage peptide. c Key: M, MALDI-TOF-TOF; E, nano-LC-ESI-MS/MS; LF, LTQ-FT.

with MASCOT identified the peptide at [M + 2H]2+ m/z 505.82, RLPSGPSLPS (amino acid numbers 477-486). Moreover, two peptide sequences corresponding to the sequential removal of the next two serine residues were identified (477RLPSGPSLPSS487, 477 RLPSGPSLPSSS488) in their [M + 2H]2+ state (Supporting Information Table 1). D. Characterization of Synapsin Isoform Migration Differences in a 2-DE Gel. In the 2-DE map of synapsin expression forms (Figure 2), 15 spots had a measurably different pI, but the same molecular weight (Mr); for example, spots 1, and 2, 3-9, and 10-15. Combination of different mass spectrometry and multiproteolytic digest approaches demonstrated that a series of deamidations of glutamine and asparagine residues were identified in the individual protein spots. Due to deamidation, “trains” of spots are proposed to appear in the 2-DE gel.52,53 Individual spots digested with different enzymes and analyzed by MALDI-TOF-TOF, nano-LC-ESI-MS/MS, and LTQ-FT analyses showed that the number of deamidated sites in peptides representing individual protein spots varied from one to another and in some spots the same number of deamidated sites were observed, probably because of relatively low sequence coverage of some individual protein spots (Supporting Information Tables 2-4). MS/MS analysis of the individual spots confirmed the exact deamidation site; for example, the MS/MS spectrum at m/z 3498.716 corresponding to the deamidated glutamine peptide sequence RPPPAQAPAPQPAPQPAPTPSVGSSFFSSLSQAVK (residues 58-92) (residues in bold indicate the amino acids that are modified with deamidation) indicated that the mass difference between b10 (m/z 983.335) and b11 (m/z 1112.166) exactly corresponds to the deamidated glutamine mass (129.04) at amino acid number 69 (Figure 3). However, not in all the protein spots was a deamidated protein site observed. E. N-Acetylation at Met-1 or Met-2. By analyzing trypsinand Asp-N-digested peptides of synapsin IIa and synapsin IIb (spots 3-15) by MALDI-TOF-TOF and nano-LC-ESI-MS/MS analysis, peptides representing N-terminal residues of these protein primary sequences were determined. MALDI-TOF analysis of protein spots digested with Asp-N shows that 2700

Journal of Proteome Research • Vol. 6, No. 7, 2007

mixtures of peptides representing NH4-terminal acetylated methionine (Ac-Met-1) and peptides corresponding to the second methionine (Ac-Met-2) acetylated after cleavage of the initiator methionine were present (Figure 4). The corresponding MS spectrum clearly showed that both N-terminally acetylated peptides (Ac-M1MNFLRRRLS, Ac-M2NFLRRRLS) with a mass increment of 42 Da exist (Figure 4). Parts A and B of Figure 5 show the MALDI-TOF-TOF and de novo sequencing analysis of singly charged ions at m/z 1364.71 and 1233.70 matching the sequences Ac-M1MNFLRRRLS and Ac-M2NFLRRRLS, respectively. The precursor ions [M + H]+ at m/z 1364.71 and 1233.70 were selected for MALDI-TOFTOF analysis, and subsequent de novo sequencing analysis with RapiDeNovo software was carried out. De novo sequencing was performed on the basis of the presence of complementary band y-ion information from the MS/MS spectrum, and de novo sequencing software confirmed the presence of N-terminal acetylation at amino acids Met-1 and Met-2 by generating the peptide sequences Ac-M1MNFLRRRLS and Ac-M2NFLRRRLS, respectively, with high scores (de novo scores 600 and 268). The generated sequence was subsequently submitted to an MSBLAST search against the NCBI nucleic acid database, and the sequence perfectly matched the synapsin IIa (AAH66004) and synapsin IIb (AAC72966) sequences. The MS/MS spectrum [M + H]+ at m/z 1364.71 for the corresponding peptide sequence Ac-M1MNFLRRRLS indicated the presence of a nearly complete b-ion (b1-b9) series and y-ion (y3-y6) information. In particular, observation of the mass of a b1 ion at m/z 174, which corresponds to the mass of acetylated methionine (Met-1), and the presence of an a1 ion at m/z 146 supported the notion that Met-1 was exclusively acetylated (Figure 5A). The MS/MS spectrum [M + H]+ at m/z 1234.71 for the corresponding peptide sequence Ac-M2NFLRRRLS indicated the presence of a b-ion (b1-b8) series and y-ion (y4-y7) information. Moreover, the presence of a b1 ion at 173.94 Da corresponds to the mass of acetylated Met-2, and the presence of an a1 ion at m/z 145.96 in turn indicates that Met-2 is acetylated (Figure 5B). The neutral loss of the characteristic acetyl group (-42 Da) from the respective acetylated precursor ions clearly indicated the

Mass Spectrometrical Studies on Synapsin Isoforms

research articles

Figure 3. MS/MS spectrum of the m/z 3498.716 assignment to sequence 59-93, RPPPAQAPAPQPAPQPAPTPSVGSSFFSSLSQAVK, modified by deamidation of a glutamine residue. The asterisk indicates that the mass of b10 and b11 increased to 129, which corresponds to deamidation of glutamine at amino acid number 69.

Figure 4. MS spectrum of synapsin IIb. The mass ions of the acetylated fragments and of its phosphorylated peptides are indicated.

acetylation of these peptides (Supporting Information Figure 1). This modification was also confirmed in protein spots digested with trypsin analyzed by nano-LC-ESI-MS/MS. The MS/MS spectrum of the precursor ion [M + 2H]2+ at m/z 1443.22 identified the corresponding peptide sequence Ac-

MMNFLR in synapsin IIa (spot 3) and synapsin IIb (spot 9) in its methionine-oxidized state. F. Identification of Known and Novel Phosphorylation Sites. PMF analysis of tryptic peptides obtained from synapsin IIa (spots 3-9) indicated an observed ion at m/z 2021.86 not Journal of Proteome Research • Vol. 6, No. 7, 2007 2701

research articles

John et al.

Figure 5. (A) MS/MS spectrum of the m/z 1365.760 (spot 12) assignment of the identified sequence 1-10, MMNFLRRRLS, which contains N-terminally modified methionine. The asterisk indicates that the mass of the N-terminal methionine residue was increased to m/z 174.13, which corresponds to Met-1 acetylation. (B) MS/MS spectrum of the m/z 1234.710 (spot 12) assignment of the identified sequence 2-10, MNFLRRRLS, which contains N-terminally modified methionine. The spectrum clearly indicates that Met-1 was removed during post-translational processing and subsequent addition of an acetyl group. The asterisk indicates that the mass of the N-terminal methionine residue was increased to m/z 173.949, which corresponds to Met-2 acetylation.

detected in spot 3 but present in spots 4-9; this finding shows that spot 3 is the nonphosphorylated form of the protein spot and spots 4-9 are phosphorylated forms of synapsin IIa. PMF analysis of Asp-N-digested peptides analyzed by MALDI-TOF showed the presence of ions at m/z 1314.68 and 1445.755 that were not present in spots 3 and 10 but again present in protein spots 4, 5, 6, 7, 9 ,11,12, 13, 14, and 15; this shows that spot 3 and spot 10 are spots representing unphosphorylated forms of synapsin IIa and synapsin IIb protein spots in the 2-DE gel, respectively (Figure 2, Table 1). Normally, more acidic phosphorylated protein species would be predominantly expected in the left-hand part of the 2-DE gel (with a lower isoelectric point), and the phosphorylated species would be more prevalent in the right-hand region of the 2-DE gel (higher isolectric point).35 2702

Journal of Proteome Research • Vol. 6, No. 7, 2007

Our experimental data revealed a novel phosphorylation site in synapsin IIa in the domain E region, close to the C-terminal of the protein sequence. Figures 6 and 7 show MALDI-TOFTOF and de novo sequencing analysis of singly charged ions at m/z 2021.86 matching the sequence 544SQpSLTNAFSFSESSFFR560 (residues in bold indicate the amino acids that are modified with phosphorylation) in synapsin IIa. Because phosphorylation of the peptide increases the mass of an ion by 80 Da, PMF data effectively denoted the presence of a phosphopeptide compared to the calculated masses of proteolytic peptides. Protonated molecular ions of tryptic peptide SQSLTNAFSFSESSFFR (residues 544-560) were detected in its monophosphorylated form ([M + H]+ at m/z 2021.86). The observed mass shift of 80 Da corresponds to the addition of a single phosphate group to the corresponding nonphosphoryl-

Mass Spectrometrical Studies on Synapsin Isoforms

research articles

Figure 6. Identification of the phosphorylated peptide by MALDI-MS. MS spectrum of the phosphorylated peptide of synapsin IIb. The mass ion of the unmodified tryptic fragment and of its phosphorylated peptide is indicated. The signal at m/z 1936.420 may correspond to the oxidized form of the metastable fragment ion.

ated peptide ([M + H]+ at m/z 1941.90) (Figure 6). Alkaline phosphatase treatment and MALDI-MS analysis resulted in the disappearance of the monophosphorylated peptide with [M + H]+ at m/z 2021.852 due to dephosphorylation (Supporting Information Figure 2). The mass of m/z 2021.86 was present in all the synapsin IIa protein spots (spots 4-9) except in nonphosphorylated protein spot 3 (Table 1). Observation of the oxidized form of the metastable fragment ion at m/z 1936.420 in the MS spectrum strongly suggested that the peptide SQSLTNAFSFSESSFFR was modified with phosphorylation. However, this peptide contained seven potential phosphorylated sites (Ser-544, Ser-546, Thr-548, Ser-552, Ser-554, Ser-556, and Ser-557), so site-specific localization of the phosphorylation site is of importance. A precursor ion [M + H]+ at m/z 2021.84 was selected for MS/MS analysis, and subsequent de novo sequencing analysis with RapiDeNovo software was carried out. De novo sequencing was performed on the basis of the presence of complementary b- and y-ion information from the MS/MS spectrum, and de novo sequencing software confirmed the presence of phosphorylation at amino acid Ser-456 by generating the peptide sequence 544SQpSLTNAFSFSESSFFR560 with a high score (de novo score 8204) (Figure 7). The generated sequence was subsequently submitted to an MS-BLAST search against the NCBI nucleic acid database, and as a result, the sequence perfectly matched the synapsin IIa sequence. This modification was further verified by the observation of neutral loss of 80 Da (-H3PO3) at m/z 1942.30 as evaluated by MALDITOF-TOF analysis (Figure 8). A nearly complete series of y-ions (y1-y15) and a few b-ion fragments were observed in the MS/ MS spectrum, including several ions corresponding to neutral loss of the phosphate group in the b-ion series (m/z 285.00, 498.82, 683.78, and 831.925) (Figure 7). Close observation of the MS/MS spectrum revealed that the mass difference between y14 and y15 increased to 164, indicating that Ser-546 was specifically phosphorylated as these fragment ions shifted by 80 Da compared with the corresponding fragments generated from the nonphosphorylated peptide (Figure 7). In addition,

as compared to nonphosphorylated peptide fragments, y1-y13 fragmented masses and the b2 ion mass at m/z 216.02 were unchanged, proposing the unique phosphorylation site at Ser546 but not at Ser-544. These results together with the presence of a [b-Pi3] ion at m/z 285.00, i.e., b3-98, demonstrated that Ser-546 is the only residue phosphorylated in this peptide (Figure 7). Phosphorylation of Ser-546 was observed in 10 out of 10 gels examined. All four synapsins are known as excellent substrates for PKA and CaM kinase I as these contain consensus phosphorylation sites in the domain A region. In the synapsin I isoform, it was experimentally proven that Ser-9 (site 1) was phosphorylated by PKA and CaM kinase I, but there are no mass spectrometric confirmations available for synapsin IIa and IIb in the mouse organism. Asp-N-digested peptides of both synapsin IIa and synapsin IIb protein spots (spots 4-9, spots 11-15) and analysis with MALDI-TOF-TOF confirmed that Ser-9 or Ser10 was phosphorylated by PKA and CaM kinase I. Protonated molecular ions of Asp-N-digested peptides Ac-MMNFLRRRLS (residues 1-10) and Ac-MNFLRRRLS (residues 2-10) were detected in their monophosphorylated forms [M + H]+ at m/z 1445.75 and 1314.68, respectively (Figure 4). The MALDI-MS spectrum obtained from Asp-N digestion is shown in Figure 4; the observed mass shift of 80 Da corresponds to the addition of a single phosphate group to the corresponding nonphosphorylated peptide masses at m/z 1365.76 and 1234.70. This mass increment is in agreement with the observed and theoretical masses of the modified peptides. These mass increments are also observed in the oxidized form of these peptides. Precursor ions of phosphorylated peptide ions [M + H]+ at m/z 1445.75 and 1314.68 were subjected to MS/MS analysis. Poor fragmentation patterns in MS/MS spectra were obtained due to the fact that the acetylated N-terminal and phosphorylated C-terminal cannot be protonated anymore. However, ions corresponding to the loss of H3PO3 (-80 Da) were observed at m/z 1365.95 and 1234.75 from respective phosphorylated peptide ions at m/z 1445.75 and 1314.68 (data Journal of Proteome Research • Vol. 6, No. 7, 2007 2703

research articles

John et al.

Figure 7. Identification of a novel phosphorylation site by MALDI-MS/MS and de novo sequence analysis. MALDI-TOF-TOF spectra of the unphosphorylated peptide SQSLTNAFSFSESSFFR (upper panel) and the phosphorylated peptide SQpSLTNAFSFSESSFFR (lower panel). The arrows indicate the 80 Da shifts for the individual fragment ions containing the phosphate moiety. The asterisk indicates that the mass of y14 and y15 increased to 80 Da, representing Ser-546 phosphorylation.

not shown), strongly indicating that Ser-9 or Ser-10 was phosphorylated by PKA and CaM kinase I in the mouse hippocampus. The synapsin Ia protein spot (spot 2) digested with trypsin analyzed with MALDI-TOF confirmed the PKA and CaM kinase I phosphorylation site at Ser-9. The tryptic peptide LSDSNFMANLPNGYMTDLQR (residues 8-27) would have an [M + H]+ ion at m/z 2286.04. If Ser-9 were phosphorylated, it would be at m/z 2366.01. A close examination of all of the PMF data determined an ion at m/z 2366.21 (observed m/z 2367.21) Therefore, spot 2 probably contains phosphorylation at Ser-9 (Table 1). Trypsin digestion of the synapsin Ia protein spot (spot 2) and analysis by MALDI-TOF confirmed the MAPK kinase 2704

Journal of Proteome Research • Vol. 6, No. 7, 2007

phosphorylation site at Ser-62. The tryptic peptide ASTAAPVASPAAPSPGSSGGGGFFSSLSNAVK (residues 54-85) would have an [M + H]+ ion at m/z 2833.39. If Ser-62 were phosphorylated, it would appear at m/z 2913.35. A close examination of PMF data revealed an ion at m/z 2913.48 (observed m/z 2914.48). Therefore, MAPK kinase phosphorylation possibly occurs at Ser-62 (Table 1). G. Identification of Phosphorylation Sites Using IMAC and Nano-LC-ESI-MS/MS. To detect minor sites of synapsin IIa and IIb phosphorylation, peptides generated in the proteolytic digests of trypsin and Asp-N were enriched for phosphopeptides using IMAC (Ga3+)-based preferential enrichment of phosphopeptides and analyzed with nano-LC-ESI-MS/MS. Phosphorylated peptides and sites of phosphorylation from the

research articles

Mass Spectrometrical Studies on Synapsin Isoforms

Figure 8. Identification of the new phosphorylation site by MALDI-TOF-TOF analysis. Demonstration of neutral loss of 80 Da (-H3PO3) from the precursor ion. Table 3. List of Phosphorylation Peptides and Sites of Phosphorylation Identified after IMAC and Subsequent Nano-LC-ESI-MS/MS on Synapsins IIa and IIb spot no.

5 5 5 5 14 14 13 12 12 a

protein name

synapsin IIa synapsin IIa synapsin IIa synapsin IIa synapsin IIb synapsin IIb synapsin IIb synapsin IIb synapsin IIb

phosphopeptidea 544SQpSLTNAFSFSESSFFR560 544SQpSLTNAFSFSESSFFR560 422TPALpSPQRPLTTQQPQSGTLK442 422pTPALpSPQRPLTTQQPQSGTLK442 426pSPQRPLTTQQPQSGTLK442 422pTPALSPQRPLTTQQPQSGTLK442 426pSPQRPLTTQQPQSGTLK442 426pSPQRPLTTQQPQSGTLKEP442 426pSPQRPLTTQQPQSGTLK442

site of phosphorylation

obsd m/z

S-546 S-546 S-426 T-422, S-426 S-426 T-422 S-426 S-426 S-426

1011.40 674.52 776.95 803.70 649.60 776.98 649.60 724.96 649.61

Mr(exptl) Mr(calcd)

2020.79 2020.55 2327.82 2408.08 1945.78 2327.93 1945.79 2171.86 1945.80

2020.86 2020.86 2328.18 2408.15 1945.96 2328.18 1945.96 2172.06 1945.96



expect

-0.07 -0.31 -0.37 -0.07 -0.18 -0.25 -0.17 -0.20 -0.16

1.7E-005 0.0022 0.39 0.47 0.43 2.5 1.2 0.02 0.8

no. of missed charge cleavages enzymeb M Sc

2 3 3 3 3 3 3 3 3

0 0 0 0 0 0 0 0 0

T T T T U T U U U

E E E E E E E E E

pS or pT denotes phosphorylated serine or threonine. b Key: T, trypsin; U, unspecific cleavage peptide from Asp-N digests. c Key: E, nano-LC-ESI-MS/

MS.

trypsin and Asp-N digests from synapsins IIa and IIb are listed in Table 3. The use of IMAC resulted in the confirmation of novel phosphorylation at Ser-546 on synapsin IIa (Supporting Information Figure 3A). The mass spectra of these peptides clearly showed the doubly and triply charged ions of the phosphorylation forms. Trypsin- and Asp-N-digested peptides of both synapsin IIa and synapsin IIb protein spots (spots 5, 12, 13, and 14) were identified as phosphorylated at Thr-422 and Ser-426. The MS/MS spectrum of the triply charged ion at m/z 803.70, 422pTPALSpPQRPLTTQQPQSGTLK442, from the tryptic digests and the unspecific cleavage peptide from the Asp-N digest, [M + 3H]3+ at m/z 724.96, 426pSPQRPLTTQQPQSGTLKEP442, identified them as phosphorylated at Thr-422 and S-426 (Supporting Information Figure 3B). However, Thr-422 and Ser-426 were only identified after IMAC enrichment of phosphopeptides, suggesting that these sites were minor phosphorylation sites. This result supports the recently published phosphorylation sites (Thr-422 and Ser-426) on the synapsin II isoform in the mouse organism.45 H. Confirmation of Pro-44 in Synapsin Ia. On the basis of DNA cloning and sequence analysis of the 5′-terminal synapsin I gene (exon 1), Chin et al.54 published a partial sequence (125) containing leucine instead of proline at position 44 in the mouse organism (GenBank ID AAA79963) (Figure 9). Asp-Ndigested peptides from spot 1 and spot 2 were analyzed by MALDI-TOF-TOF, and the presence of proline at amino acid

44 was confirmed. MS/MS analysis of the Asp-N-digested peptide of spot 1 at m/z 2623.30 and a subsequent MASCOT search showed that it perfectly matched synapsin I, 24-51 (DLQRPQPPPPPPSAASPGATPGSATASA), with a significant score (ion score 30) and confirmed the presence of proline (Figure 10). From spot 2 this peptide was identified in its deamidated form (m/z 2624.300). The MS/MS spectrum indicated that the presence of ions corresponded to the several proline-containing peptides. The mass difference between y7- and y8-ions corresponds to the mass of proline in the MS/MS spectrum, suggesting the presence of proline at amino acid position 44 in the synapsin Ia primary structure in the hippocampal tissue of the mouse (Figure 10). I. Identification of Methylation Sites in Synapsin IIa and Synapsin IIb Protein Spots. Methylation of the acidic residue glutamic acid (E) and aspargine (N) was identified in synapsin IIa and synapsin IIb protein spots. Both trypsin- and Asp-Ndigested peptides analyzed with MALDI-TOF-TOF and nanoLC-ESI-MS/MS analysis identified methylation of glutamic acid at 148 (Glu-148) in synapsin IIa (spot 4) and synapsin IIb (spots 10 and 12), Glu-249 (spots 5 and 6) in synapsin IIa, and Glu-151 and Glu-122 (spot 13) and Glu-402 (spot 10) in synapsin IIb (Table 1). J. Identification of Pyro Glu and Tryptophan Oxidation in Synapsin Isoforms. Other modifications include Pyro Glu at position 404 in synapsin Ia (spots 1 and 2), Pyro Glu-94 in both Journal of Proteome Research • Vol. 6, No. 7, 2007 2705

research articles

John et al.

Figure 9. Comparison of the partial protein sequence of mouse synapsin Ia (NCBI accession no. AAH22954) and deduced amino acid sequences of the 5′-terminal synapsin I gene (exon 1) (GenBank ID AAA79963). The sequences were obtained from the NCBI protein database and GenBank. The alignment was performed using the Clustal W program. MS/MS data provided evidence for the presence of proline rather than lysine in the sequence, thus generating a sequence conflict.

Figure 10. The MALDI-TOF-TOF spectrum of m/z 2622.30 obtained from peptides resulting from Asp-N digestion of synapsin Ia (spot 1) and subsequent MASCOT analysis confirmed the presence of proline at position 44.

synapsin IIa and synapsin IIb (spots 5, 6, 12, and 13), and Pyro Glu-207 in synapsin IIb (spot 14) and were identified by both mass spectrometry approaches. Tryptophan oxidation was identified in singly and doubly oxidized states in synapsin Ia, synapsin IIa, and synapsin IIb protein spots. Wox-126 (spots 1 and 2), Wox-127 (spots 5, 6, 11, 12, and 13), and Wox-336 (spot 5) were observed (Table 1).

Discussion Synapsin isoforms and their post-translational modifications play an important role in the regulation of neurotransmission, but their precise function has been difficult to pinpoint. Detection of synapsin isoforms as protein spots in 2-DE gels from the mouse hippocampus prompted us to investigate the primary structure of synapsin isoforms and their posttranslational modifications. Moreover, protein chemical characterization of synapsin II isoforms has not been studied as extensively as that of synapsin I. A combination of mass spectrometry (MALDI-TOF-TOF, nano-LC-ESI-MS/MS, and LTQ-FT) and enzymatic digestion (trypsin, Asp-N, Lys-C, and 2706

Journal of Proteome Research • Vol. 6, No. 7, 2007

nonspecific enzyme subtilisin) was used, and analysis shows the presence of two synapsin Ia protein spots, seven synapsin IIa protein spots, and six synapsin IIb protein spots in the 2-DE gels from the mouse hippocampus. Combination of multienzyme digestion and mass spectrometry allowed for high sequence coverage of individual synapsin isoforms (Table 1); particularly, 94% sequence coverage was achieved in the case of synapsin IIb protein spots (spot 12). The isolation of individual isoforms of individual synapsins is technically difficult using conventional purification techniques, because of the extent of homology between primary sequences. Moreover, synapsin II is more hydrophobic than synapsin I, and it is therefore difficult to purify synapsin II isoforms from brain tissue. These problems have been overcome by the expression of recombinant synapsin IIa in the eukaryotic expression system.55 Our experimental approach showed that 2-DE-based MS analysis may be a complementary approach for characterization of the primary structure of synapsin II isoforms ex vivo by obtaining high sequence coverage and post-translational modifications. Deamidation of

research articles

Mass Spectrometrical Studies on Synapsin Isoforms

Figure 11. Alignment of amino acid sequences of mouse, rat, bovine, and human synapsin I and II isoforms. The consensus sequence predicted to be phosphorylated by CaM kinase II (Hyd-X-R/K-NB-X-S/T-Hyd, where Hyd ) hydrophobic, NB ) nonbasic, and X may be any residue) is marked in the box. The phosphorylated residue is underlined. Table 4. Identified Phosphorylation Sites and Predicted Consensus Sequence for Kinases Responsible for Phosphorylation on Synapsin Isoforms protein name

phosphopeptide

8LpSDSNFMANLPNGYMTDLQR27 synapsin Ia synapsin IIa and synapsin IIb Ac-M1MNFLRRRLpS10 synapsin IIa and synapsin IIb Ac-M2NFLRRRLpS9 54ASTAAPVApSPAAPSPGSSGGGGFFSSLS synapsin Ia NAVK65 544SQpSLTNAFSFSESSFFR560 synapsin IIa synapsin IIa and synapsin IIb 422pTPALpSPQRPLTTQQPQSGTLK442

a

site of phosphorylation

Ser-9 Ser-10 Ser-9 Ser-62

predicted kinase

CaM kinase I PKA and CaM kinase I PKA and CaM kinase I proline-directed kinase (e.g., MAPK) Ser-546 CaM kinase II Thr-422, Ser-426 proline-directed kinase (e.g., MAPK)

consensus patterna

[RK][RK][ST]... [RK][RK][ST]... [RK][RK][ST]... ...([ST])P... Hyd-X-R/K-NB-X-S/T-Hyd63-65 ...([ST])P...

Hyd ) hyrophobic amino acid. NB ) nonbasic amino acid.

asparagine (N) and glutamine (Q) residues to aspartate (D) and glutamate (E) residues, respectively, is an effect of protein aging and is often observed in 2-DE gels.53,56,57 Deamidation leads to a series of spots with the same Mr, but measurably different pI values.58 The presence of spot trains in the 2-DE gel can be explained by the existence of a heterogeneous mixture of deamidated protein species. Potential sites of deamidation were identified in “thermally stressed” samples of recombinant human growth hormone59 and recombinant tissue plasminogen activator.60 The deamidation pattern (ladder of protein spots) of growth hormone (GH) isoforms in the 2-DE gel was identified and characterized by MS.61 Both PMF and MS/MS analysis of individual protein spots clearly showed that the number of deamidated peptides differs from one spot to the other (Supporting Information Tables 2-4). Herein, the results clearly show the deamidation susceptibility of synapsin isoforms. Deamidation of glutamine to glutamate occurred predominantly in the synapsin IIa and synapsin IIb protein spots (Supporting Information Tables 2-4). This study presents the detailed mapping of the protein sequence of synapsin isoforms by tandem MS and PMF. The major focus of this work was to apply the MS technique to reveal protein characterization and identification of specific synapsin isoforms, as they were not fully shown at the protein level. This was accomplished by analyzing multiple protease digests of synapsin isoforms in 2-DE followed by MALDI-TOFTOF, nano-LC-ESI-MS/MS, or LTQ-FT. All four synapsins are substrates for protein kinases PKA and CaM kinase I, which phosphorylate serine residues in the NH2-terminal region. Synapsin I, but not synapsin II, is an excellent substrate for CaM kinase II, which phosphorylates two serine residues in the COOH-terminal region of synapsin I (sites 1 and 2).2 Synapsin phosphorylations have been well-studied and were found to regulate the function of synapsins.19 Detailed analysis of ex vivo phosphorylation described herein revealed a new phosphorylation site at Ser-546 in synapsin IIa. This new phosphorylated site is located in the C-terminal domain E of synapsin IIa. All synapsin isoforms share the C-terminal

domains of the A-type that play an important role in both the predocking and the postdocking steps of synaptic vesicle exocytosis21,62 and seem to be required for correct targeting of the synapsins to synaptic vesicles.63 However, it was stated in a recent report that domain E increases synaptic efficiency by accelerating both the kinetics of exocytosis and the rate of synaptic vesicle cycling.64 Interestingly, the equivalent site in bovine synapsin I, Ser-666, was identified as phosphorylated in vitro by CaM kinase II38 (Figure 11). The phosphorylation site Ser-666 was part of the consensus sequence known to be phosphorylated by CaM kinase II (Hyd-X-R/K-NB-X-S/T-Hyd, where Hyd ) hydrophobic, NB ) nonbasic, and X may be any residue).65-67 These proposed conserved sequences of CaM kinase II are extremely conserved throughout all synapsin A-type sequences including synapsin IIa (Figure 11). Identification of a new phosphorylated site at Ser-546 in synapsin IIa in the consensus sequence of CaM kinase II prompted us to conclude that both synapsin I and synapsin IIa are most likely substrates for CaM kinase II. The exact function of Ser-546 phosphorylation at the E-domain, however, remains to be evaluated in further functional studies. Table 4 indicates identified phosphorylation and predicted consensus sequences for kinases responsible for phosphorylation on synapsin isoforms using the Web-based ELM (EMBL) program. In addition to a new phosphorylation site, we confirmed two previously characterized PKA and CaM kinase I phosphorylation sites at Ser-9 (site 1) and the MAPK kinase phosphorylation site at Ser-62 in synapsin I through PMF analysis following enzymatic digestion with trypsin. The analysis by PMF and MS/MS experiments with the Asp-N digest of synapsins IIa and IIb for the first time unambiguously identified a PKA and CaM kinase I phosphorylation site at Ser-9 or Ser10 in the mouse organism. A previous study showed that N-terminal phosphorylation of this PKA and CaM kinase I site of all the synapsins modulates their binding to the synaptic vesicle.68 Phosphorylation of synapsin Ia at Ser-568 by CaM kinase II abolishes the bundling activity of actin filaments.69 Journal of Proteome Research • Vol. 6, No. 7, 2007 2707

research articles N-terminal acetylation, together with N-terminal methionine cleavage, is the most common protein modification in eukaryotic cells. Over 40% of all yeast proteins and almost 90% of mammalian proteins are estimated to be N-terminally acetylated.70-73 Protein N-terminal methionine cleavage is an essential cotranslation process that occurs in the cytoplasm of all organisms.74 Cleavage of N-terminal methionine is catalyzed by methionine aminopeptidases (MAPs).75 There are two types of reported MAPs described in eukaryotes, MAP 1 and MAP 2.75,76 However, experimental evidence has been presented that removal of methionine is dependent on whether the penultimate residue has a radius of gyration that is less than 1.29 Å (for example, bulky, acidic, or hydrophobic amino acids). N-terminal acetylation occurs cotranslationally when nascent peptides are between 20 and 50 amino acids long.71,77 The addition of the acetyl group is catalyzed by N-terminal acetyl transferase (NATs).70 In Saccharomyces cervisiae there are known NATs, NatA, NatB, and NatC.74 NATs act on substrates with specific but degenerated N-terminal regions, required for the activity of each NAT.78 In the case of NatB an N-terminal sequence of MD, ME, MN, or MM is required for acetylation.79 NatB acetylates the first methionine of the nascent chain not subjected to methionine cleavage. The synapsin I isoform contains the N-terminal amino acid sequence MN. Synapsins IIa and IIb contain MM in the N-terminal position. N-terminal acetylation was described in bovine synapsin I by LC-MS analysis34 but not in synapsin II isoforms. Our results showed a discrepancy in the N-terminal acetylation process by acetylating first methionine and also acetylating the second methionine after the cleavage of the first methionine in both synapsin II isoforms. On the contrary, our results showed that first methionine could be removed if the penultimate residue has a high radius of gyration value, i.e., Met-2. The loss of methionine could not cause any pI shift and molecular weight change in the 2-DE gels. Finally, we conclude that synapsin IIa and IIb proteins show NH4-terminally acetylated methionine (Ac-Met-1) and a protein corresponding to the second methionine (Ac-Met-2) acetylated after cleavage of the initiator methionine. The biological significance of N-terminal acetylation as well as methionine removal in the amino terminal region close to the PKA and CaM kinase I phosphorylation site certainly deserves further in-depth analysis. In addition to the above-mentioned modifications, we detected methylation of certain glutamic acid and aspargine residues. Methylation of acidic moieties can occur for several different reasons. The most common ones are artifacts from electrophoresis, especially in colloidal Coomassie staining of proteins;80 on the other hand, methylation is an important posttranslational modification with biological consequences. In conclusion, 15 synapsin isoform protein spots were characterized by mass spectrometry as individual synapsin isoforms, synapsin Ia, synapsin IIa, and synapsin IIb. Serine phosphorylation sites were confirmed for Ser-9, Ser-10, Ser62, S-426, and Thr-422, and a novel phosphorylation site was observed at Ser-546 in synapsin IIa. This novel phosphorylation site may control binding of synapsin IIa to synaptic vesicle membranes with the possible consequence of neurotransmitter release on the basis of the knowledge of known phosphorylations as given above and challenges functional neurobiological studies on the role of individual phosphorylation states of synapsins. A series of deamidation sites were observed in the individual protein spots. The results may represent a step forward in the protein chemical characterization of these most 2708

Journal of Proteome Research • Vol. 6, No. 7, 2007

John et al.

important neural elements as the molecular basis of synaptic function is of major interest to neuroscience. Knowledge of the individual isoforms and their modifications is a prerequisite for all neurobiological studies at the protein level. Abbreviations: NAT, N-terminal acetyl transferase; MAPs, methionine aminopeptidases; CaMKs, Ca2+/calmodulin protein kinases; PKA, cAMP-dependent protein kinase; MAPK, mitogenassociated protein kinase; LT-FT, linear ion trap integrated with Fourier transform; OGP, octyl β-D-glucopyranoside; LID, laserinduced dissociation; FA, formic acid; NCBI, National Center for Biotechnology Information.

Acknowledgment. We are highly indebted to Dr. Leila Afjehi-Sadat and Dr. Daniela Pollak for preparing the 2-DE gels, Dr. Ilse Dohnal, University of Vienna, Vienna, Austria, for excellent mass spectrometry support, Robert Mosto¨gl for formatting the manuscript, and the Verein zur Durchfu ¨ hrung der wissenschaftlichen Forschung auf dem Gebiet der Neonatologie und Kinderintensivmedizin (“Unser Kind”). Supporting Information Available: Three figures and four tables. This material is available free of charge via the Internet at http://pubs.acs.org. References (1) Huttner, W. B.; Schiebler, W.; Greengard, P.; De Camilli, P. Synapsin I (protein I), a nerve terminal-specific phosphoprotein. III. Its association with synaptic vesicles studied in a highly purified synaptic vesicle preparation. J. Cell. Biol. 1983, 96, 13741388. (2) De Camilli, P.; Benfenati, F.; Valtorta, F.; Greengard, P. The synapsins. Annu. Rev. Cell. Biol. 1990, 6, 433-460. (3) Hosaka, M.; Sudhof, T. C. Synapsin III, a novel synapsin with an unusual regulation by Ca2+. J. Biol. Chem. 1998, 273, 1337113374. (4) Kao, H. T.; Porton, B.; Czernik, A. J.; Feng, J. A third member of the synapsin gene family. Proc. Natl. Acad. Sci. U.S.A. 1998, 95, 4667-4672. (5) Sudhof, T. C.; Czernik, A. J.; Kao, H. T.; Takei, K. Synapsins: mosaics of shared and individual domains in a family of synaptic vesicle phosphoproteins. Science 1989, 245, 1474-1480. (6) Sudhof, T. C. The structure of the human synapsin I gene and protein. J. Biol. Chem. 1990, 265, 7849-7852. (7) Hilfiker, S.; Pieribone, V. A.; Czernik, A. J.; Kao, H. T. Synapsins as regulators of neurotransmitter release. Philos. Trans. R. Soc. London, B: Biol. Sci. 1999, 354, 269-279. (8) Ueda, T.; Greengard, P. Adenosine 3′:5′-monophosphate-regulated phosphoprotein system of neuronal membranes. I. Solubilization, purification, and some properties of an endogenous phosphoprotein. J. Biol. Chem. 1977, 252, 5155-5163. (9) Goelz, S. E.; Nestler, E. J.; Chehrazi, B.; Greengard, P. Distribution of protein I in mammalian brain as determined by a detergentbased radioimmunoassay. Proc. Natl. Acad. Sci. U.S.A. 1981, 78, 2130-2134. (10) Browning, M. D.; Huang, C. K.; Greengard, P. Similarities between protein IIIa and protein IIIb, two prominent synaptic vesicleassociated phosphoproteins. J. Neurosci. 1987, 7, 847-853. (11) Nestler, E. J.; Greengard, P. Protein phosphorylation in the Nervous System; John Wiley & Sons, Inc.: New York, 1983. (12) Greengard, P.; Valtorta, F.; Czernik, A. J.; Benfenati, F. Synaptic vesicle phosphoproteins and regulation of synaptic function. Science 1993. 259, 780-785. (13) Jovanovic, J. N.; Czernik, A. J.; Fienberg, A. A.; Greengard, P.; Sihra, T. S. Synapsins as mediators of BDNF-enhanced neurotransmitter release. Nat. Neurosci. 2000, 3, 323-329. (14) Sakurada, K.; Kato, H.; Nagumo, H.; Hiraoka, H. Synapsin I is phosphorylated at Ser603 by p21-activated kinases (PAKs) in vitro and in PC12 cells stimulated with bradykinin. J. Biol. Chem. 2002, 277, 45473-45479. (15) Schiebler, W.; Jahn, R.; Doucet, J. P.; Rothlein, J.; Greengard, P. Characterization of synapsin I binding to small synaptic vesicles. J. Biol. Chem. 1986, 261, 8383-8390.

Mass Spectrometrical Studies on Synapsin Isoforms (16) Greengard, P.; Browning, M. D.; McGuinness, T. L.; Llinas, R.; Synapsin, I. a phosphoprotein associated with synaptic vesicles: possible role in regulation of neurotransmitter release. Adv. Exp. Med. Biol. 1987, 221, 135-153. (17) Benfenati, F.; Valtorta, F.; Chieregatti, E.; Greengard, P. Interaction of free and synaptic vesicle-bound synapsin I with F-actin. Neuron 1992, 8, 377-386. (18) Fesce, R.; Benfenati, F.; Greengard, P.; Valtorta, F. Effects of the neuronal phosphoprotein synapsin I on actin polymerization. II. Analytical interpretation of kinetic curves. J. Biol. Chem. 1992, 267, 11289-11299. (19) Valtorta, F.; Greengard, P.; Fesce, R.; Chieregatti, E.; Benfenati, F. Effects of the neuronal phosphoprotein synapsin I on actin polymerization. I. Evidence for a phosphorylation-dependent nucleating effect. J. Biol. Chem. 1992, 267, 11281-11288. (20) Li, L.; Chin, L. S.; Shupliakov, O.; Brodin, L. Impairment of synaptic vesicle clustering and of synaptic transmission, and increased seizure propensity, in synapsin I-deficient mice. Proc. Natl. Acad. Sci. U.S.A. 1995, 92, 9235-9239. (21) Pieribone, V. A.; Shupliakov, O.; Brodin, L.; Hilfiker-Rothenfluh, S. Distinct pools of synaptic vesicles in neurotransmitter release. Nature 1995, 375, 493-497. (22) Rosahl, T. W.; Spillane, D.; Missler, M.; Herz, J. Essential functions of synapsins I and II in synaptic vesicle regulation. Nature 1995, 375, 488-493. (23) Takei, Y.; Harada, A.; Takeda, S.; Kobayashi, K. Synapsin I deficiency results in the structural change in the pre synaptic terminals in the murine nervous system. J. Cell. Biol. 1995, 131, 1789-1800. (24) Hilfiker, S.; Schweizer, F. E.; Kao, H. T.; Czernik, A. J. Two sites of action for synapsin domain E in regulating neurotransmitter release. Nat. Neurosci. 1998, 1, 29-35. (25) Chi, P.; Greengard, P.; Ryan, T. A. Synapsin dispersion and reclustering during synaptic activity. Nat. Neurosci. 2001, 4, 11551157. (26) Humeau, Y.; Doussau, F.; Vitiello, F.; Greengard, P. Synapsin controls both reserve and releasable synaptic vesicle pools during neuronal activity and short-term plasticity in Aplysia. J. Neurosci. 2001, 21, 4195-4206. (27) Gitler, D.; Takagishi, Y.; Feng, J.; Ren, Y. Different presynaptic roles of synapsins at excitatory and inhibitory synapses. J. Neurosci. 2004, 24, 11368-11380. (28) Han, H. Q.; Nichols, R. A.; Rubin, M. R.; Bahler, M. Two sites of action for synapsin domain E in regulating neurotransmitter release. Nature 1991, 349, 697-700. (29) Ferreira, A.; Kosik, K. S.; Greengard, P.; Han, H. Q. Aberrant neurites and synaptic vesicle protein deficiency in synapsin IIdepleted neurons. Science 1994, 264, 977-979. (30) Ferreira, A.; Han, H. Q.; Greengard, P.; Kosik, K. S. Suppression of synapsin II inhibits the formation and maintenance of synapses in hippocampal culture. Proc. Natl. Acad. Sci. U.S.A. 1995, 92, 9225-9229. (31) Rosahl, T. W.; Geppert, M.; Spillane, D.; Herz, J. Short-term synaptic plasticity is altered in mice lacking synapsin I. Cell 1993, 75, 661-670. (32) Ryan, T. A.; Li, L.; Chin, L. S.; Greengard, P.; Smith, S. J. Synaptic vesicle recycling in synapsin I knock-out mice. J. Cell. Biol. 1996, 134, 1219-1227. (33) Sun, J.; Bronk, P.; Liu, X.; Han, W.; Sudhof, T. C. Synapsins regulate use-dependent synaptic plasticity in the calyx of Held by a Ca2+/calmodulin-dependent pathway. Proc. Natl. Acad. Sci. U.S.A. 2006, 103, 2880-2885. (34) Li, K. W.; Hornshaw, M. P.; Van Der Schors, R. C.; Watson, R. Proteomics analysis of rat brain postsynaptic density. Implications of the diverse protein functional groups for the integration of synaptic physiology. J. Biol. Chem. 2004, 279, 987-1002. (35) Zhang, W.; Czernik, A. J.; Yungwirth, T.; Aebersold, R.; Chait, B. T. Matrix-assisted laser desorption mass spectrometric peptide mapping of proteins separated by two-dimensional gel electrophoresis: determination of phosphorylation in synapsin I. Protein Sci. 1994, 3, 677-686. (36) Matsubara, M.; Kusubata, M.; Ishiguro, K.; Uchida, T. Site-specific phosphorylation of synapsin I by mitogen-activated protein kinase and Cdk5 and its effects on physiological functions. J. Biol. Chem. 1996, 271, 21108-21113. (37) Maucuer, A.; Le Caer, J. P.; Manceau, V.; Sobel, A. Specific SerPro phosphorylation by the RNA-recognition motif containing kinase. Eur. J. Biochem. 2000, 267, 4456-4464. (38) McLachlin, D. T.; Chait, B. T. Improved beta-elimination-based affinity purification strategy for enrichment of phosphopeptides. Anal. Chem. 2003, 75, 6826-6836.

research articles (39) DeGiorgis, J. A.; Jaffe, H.; Moreira, J. E.; Carlotti, C. G., Jr. Phosphoproteomic analysis of synaptosomes from human cerebral cortex. J. Proteome Res. 2005, 4, 306-315. (40) Cole, R. N.; Hart, G. W. Mapping sites of O-GlcNAc modification using affinity tags for serine and threonine post-translational modifications. J. Neurochem. 1999, 73, 418-428. (41) Luthi, T.; Haltiwanger, R. S.; Greengard, P.; Bahler, M. Synapsins contain O-linked N-acetylglucosamine. J. Neurochem. 1991, 56, 1493-1498. (42) Wells, L.; Vosseller, K.; Cole, R. N.; Cronshaw, J. M. Mapping sites of O-GlcNAc modification using affinity tags for serine and threonine post-translational modifications. Mol. Cell. Proteomics 2002, 1, 791-804. (43) Murrey, H. E.; Gama, C. I.; Kalovidouris, S. A.; Luo, W. I. Protein fucosylation regulates synapsin Ia/Ib expression and neuronal morphology in primary hippocampal neurons. Proc. Natl. Acad. Sci. U.S.A. 2006, 103, 21-26. (44) Zhu, J. X.; Doyle, H. A.; Mamula, M. J.; Aswad, D. W. Protein repair in the brain, proteomic analysis of endogenous substrates for protein L-isoaspartyl methyltransferase in mouse brain. J. Biol. Chem. 2006, 281, 33802-338013. (45) Munton, R. P.; Tweedie-Cullen, R.; Livingstone-Zatchej, M.; Weinandy, F. Qualitative and Quantitative Analyses of Protein Phosphorylation in Naive and Stimulated Mouse Synaptosomal Preparations. Mol. Cell. Proteomics 2007, 6, 283-293. (46) Weitzdorfer, R.; Hoger, H.; Pollak, A.; Lubec, G. Changes of hippocampal protein levels during postnatal brain development in the rat. J. Proteome Res. 2006, 5, 3205-3212. (47) Bradford, M. M. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal. Biochem. 1976, 72, 248-254. (48) Weitzdoerfer, R.; Fountoulakis, M.; Lubec, G. Reduction of actinrelated protein complex 2/3 in fetal Down syndrome brain. Biochem. Biophys. Res. Commun. 2002, 293, 836-841. (49) Suckau, D.; Resemann, A.; Schuerenberg, M.; Hufnagel, P. A novel MALDI LIFT-TOF/TOF mass spectrometer for Proteomics. Anal. Bioanal. Chem. 2003, 376, 952-965. (50) Shevchenko, A.; Sunyaev, S.; Loboda, A.; Shevchenko, A. Charting the proteomes of organisms with unsequenced genomes by MALDI-quadrupole time-of-flight mass spectrometry and BLAST homology searching. Anal. Chem. 2001, 73, 1917-1926. (51) Chen, W. Q.; Kang, S. U.; Lubec, G. Protein profiling by the combination of two independent mass spectrometry techniques. Nat. Protocols 2006, 1, 1446-1452. (52) Anderson, N. L.; Anderson, N. G. A two-dimensional gel database of human plasma proteins. Electrophoresis 1991, 12, 883-906. (53) Sarioglu, H.; Lottspeich, F.; Walk, T.; Jung, G.; Eckerskorn, C. Deamidation as a widespread phenomenon in two-dimensional polyacrylamide gel electrophoresis of human blood plasma proteins. Electrophoresis 2000, 21, 2209-2218. (54) Chin, L. S.; Li, L.; Greengard, P. Neuron-specific expression of the synapsin II gene is directed by a specific core promoter and upstream regulatory elements. J. Biol. Chem. 1994, 269, 1850718513. (55) Siow, Y. L.; Chilcote, T. J.; Benfenati, F.; Greengard, P.; Thiel, G. Synapsin IIa: expression in insect cells, purification, and characterization. Biochemistry 1992, 31, 4268-4275. (56) Gianazza, E. Iso electric focusing as a tool for the investigation of post-translational processing and chemical modifications of protein. J. Chromatogr., A 1995, 705, 67-87. (57) Tsai, P. K.; Bruner, M. W.; Irwin, J. I.; Ip, C. C.; Oliver, C. N. Origin of the isoelectric heterogeneity of monoclonal immunoglobulin h1B4. Pharm. Res. 1993, 10, 1580-1586. (58) Karty, J. A.; Ireland, M. M.; Brun, Y. V.; Reilly, J. P. Artifacts and unassigned masses encountered in peptide mass mapping. J. Chromatogr., B: Anal. Technol. Biomed. Life Sci. 2002, 782, 363383. (59) Johnson, B. A.; Shirokawa, J. M.; Hancock, W. S.; Spellman, M. W. Formation of isoaspartate at two distinct sites during in vitro aging of human growth hormone. Biol. Chem. 1989, 264, 1426214271. (60) Paranandi, M. V.; Guzzetta, A. W.; Hancock, W. S.; Aswad, D. W. Deamidation and isoaspartate formation during in vitro aging of recombinant tissue plasminogen activator. J. Biol. Chem. 1994, 269, 243-253. (61) Zhan, X.; Giorgianni, F.; Desiderio, D. M. Proteomics analysis of growth hormone isoforms in the human pituitary. Proteomics 2005, 5, 1228-1241. (62) Hilfiker, S.; Benfenati, F.; Doussau, F.; Nairn, A. C. Structural domains involved in the regulation of transmitter release by synapsins. J. Neurosci. 2005, 25, 2658-2669.

Journal of Proteome Research • Vol. 6, No. 7, 2007 2709

research articles (63) Gitler, D.; Xu, Y.; Kao, H. T.; Lin, D. Molecular determinants of synapsin targeting to presynaptic terminals. J. Neurosci. 2004, 24, 3711-3720. (64) Fassio, A.; Merlo, D.; Mapelli, J.; Menegon, A. The synapsin domain E accelerates the exoendocytotic cycle of synaptic vesicles in cerebellar Purkinje cells. Cell Sci. 2006, 119, 4257-4268. (65) Stokoe, D.; Caudwell, B.; Cohen, P. T.; Cohen, P. The substrate specificity and structure of mitogen-activated protein (MAP) kinase-activated protein kinase-2. Biochem. J. 1993, 296, 843849. (66) Songyang, Z.; Lu, K. P.; Kwon, Y. T.; Tsai, L. H.; Filhol, O. A structural basis for substrate specificities of protein Ser/Thr kinases: primary sequence preference of casein kinases I and II, NIMA, phosphorylase kinase, calmodulin-dependent kinase II. Mol. Cell. Biol. 1996, 16, 6486-6493. (67) White, R. R.; Kwon, Y. G.; Taing, M.; Lawrence, D. S.; Edelman, A. M. Definition of optimal substrate recognition motifs of Ca2+calmodulin-dependent protein kinases IV and II reveals shared and distinctive features. J. Biol. Chem. 1998, 273, 3166-3172. (68) Hosaka, M.; Hammer, R. E.; Sudhof, T. C. A phospho-switch controls the dynamic association of synapsins with synaptic vesicles. Neuron 1999, 24, 377-387. (69) Bahler, M.; Greengard, P. Synapsin I bundles F-actin in a phosphorylation-dependent manner. Nature 1987, 326, 704-707. (70) Polevoda, B.; Sherman, F. N-terminal acetyltransferases and sequence requirements for N-terminal acetylation of eukaryotic proteins. J. Mol. Biol. 2003, 325, 595-622. (71) Driessen, H. P.; de Jong, W. W.; Tesser, G. I.; Bloemendal, H. The mechanism of N-terminal acetylation of proteins. CRC Crit. Rev. Biochem. 1985, 18, 281-325.

2710

Journal of Proteome Research • Vol. 6, No. 7, 2007

John et al. (72) Persson, B.; Flinta, C.; von Heijne, G.; Jornvall, H. Structures of N-terminally acetylated protein. Eur. J. Biochem. 1985, 152, 523527. (73) Lee, F. J.; Lin, L. W.; Smith, J. A. Na-acetyltransferase deficiency alters protein synthesis in Saccharomyces cerevisiae. FEBS Lett. 1989, 256, 139-142. (74) Giglione, C.; Boularot, A.; Meinnel, T. Protein N-terminal methionine excision. Cell. Mol. Life Sci. 2004, 61, 1455-1474. (75) Kendall, R. L.; Yamada, R.; Bradshaw, R. A. Co-translational amino-terminal processing. Methods Enzymol. 1990, 185, 398407. (76) Li, X.; Chang, Y. H. Amino-terminal protein processing in Saccharomyces cerevisiae is an essential function that requires two distinct methionine aminopeptidases. Proc. Natl. Acad. Sci. U.S.A. 1995, 92, 12357-12361. (77) Bradshaw, R. A.; Brickey, W. W.; Walker, K. W. N-terminal processing: the methionine aminopeptidase and N alpha-acetyl transferase families. Trends Biochem. Sci. 1998, 23, 263267. (78) Polevoda, B.; Sherman, F. N-alpha -terminal acetylation of eukaryotic proteins. J. Biol. Chem. 2000, 275, 36479-36482. (79) Polevoda, B.; Cardillo, T. S.; Doyle, T. C.; Bedi, G. S.; Sherman, F. Nat3p and Mdm20p are required for function of yeast NatB Nalpha-terminal acetyltransferase and of actin and tropomyosin. J. Biol. Chem. 2003, 278, 30686-30697. (80) Haebel, S.; Albrecht, T.; Sparbier, K.; Walden, P. Electrophoresisrelated protein modification: alkylation of carboxy residues revealed by mass spectrometry. Electrophoresis 1998, 19, 679-686.

PR070157R