Abstract
Protein phosphorylation is a globally adopted and tightly controlled post-translational modification, and represents one of the most important molecular switching mechanisms that govern the entire spectrum of biological processes. In the central nervous system, it has been demonstrated that phosphorylation of key proteins mediating chromatin remodeling and gene transcription plays an important role controlling brain development, synaptogenesis, learning and memory. Many studies have focused on large scale identification of phosphopeptides in brain tissue. These studies have identified phosphorylation site specific motifs useful for predicting protein kinases substrates. In this study, we applied a previously developed quantitative approach, stable isotope labeling of amino acids in mammals (SILAM), to quantify changes in the phosphorylation of nuclear proteins between a postnatal day one (p1) and a p45 rat brain cortex. Using a 15N labeled rat brain as an internal standard, we quantified 705 phosphopeptides in the p1 cortex and 1477 phosphopeptides in the p45 cortex, which translates to 380 and 585 phosphoproteins in p1 and p45 cortex, respectively. Bioinformatic analysis of the differentially modified phosphoproteins revealed that phosphorylation is upregulated on multiple components of chromatin remodeling complexes in the p1 cortex. Taken together, we demonstrated for the first time the usefulness of employing stable isotope labeled rat tissue for global quantitative phosphorylation analysis.
Keywords: phosphorylation, quantification, mass spectrometry, brain development, nucleus
Introduction
The covalent addition of a phosphate group onto a serine, threonine, or tyrosine residue is predicted to occur on almost a third of proteins in higher organisms 1. In fact, protein phosphorylation by kinases and dephosphorylation by phosphatases is by far the most effective and versatile molecular switching mechanism, which governs the entire spectrum of biological processes, from the flow of genetic information to the control of cell cycle, cell growth, differentiation and death. In the nucleus, it has been previously demonstrated that protein phosphorylation is an imperative regulatory mechanism controlling gene expression 2, 3. In the brain, these phosphorylation dependent changes in gene expression have been demonstrated to regulate neuronal differentiation 4, synaptogenesis 5, as well as learning and memory 6. Mitogen-and stress-activated kinase 1 (MSK1) is a nuclear kinase with such a role. MSK1 knock-out mice exhibit a deficiency in histone phosphorylation in the brain. Phenotypically, these mice show deficits in multiple learning and memory tasks 7, which is consistent with the general hypothesis that chromatin remodeling is necessary for memory formation 8. The importance of protein phosphorylation is further emphasized by reports of its dysregulation in human diseases. For example, the tau protein promotes the assembly and maintenance of the microtubule structure, and these functions are regulated by phosphorylation 9. Hyperphosphorylation of tau causes it to aggregate into neurofibrillary tangles, a histopathological hallmark of Alzheimer’s disease and a hypothesized contributor to human dementia 10. Furthermore, it has been suggested that the distinct combinations or arrays of phosphorylated and dephosphorylated proteins generate neuronal activity and modulate synaptic strength, but these phosphoprotein modules are poorly understood 11.
The study of phosphorylation on individual proteins has provided tremendous insight into individual protein function, however, these studies provide little information on phosphorylation modules. It is therefore difficult to appreciate the phosphorylation-mediated signal transduction cascade as a whole. In order to identify and understand these phosphorylation modules, large-scale mass spectrometry (MS) analysis has been employed 12. Mass spectrometry-based analyses of protein phosphorylation are hindered by two factors. First, phosphorylation is often present at a low stoichiometry making most phosphorylation difficult to detect in complex samples. To overcome this problem, enrichment techniques have been developed including immobilized affinity chromatography (IMAC) 13-15, metal oxide affinity chromatography (MOAC) 16, 17, strong cation exchange chromatography 18, and immunopurification 19. Further hindering phosphoprotein analysis is the less informative fragmentation pattern generated by phosphopeptides compared to unmodified peptides. In the mass spectrometer, the phosphorylation modification is more labile than the peptide backbone during collision induced dissociation (CID), which can result in the neutral loss of phosphoric acid 20. This hampers the identification of the phosphopeptide, since this neutral loss fragmentation pattern yields fewer sequence ions. Precursor ion mapping and MS3 strategies, however, have taken advantage of this phenomenon to further improve the identification of phosphopeptides 18, 21. In the MS3 strategy, the neutral loss ion is selected for further fragmentation, and a more informative fragmentation pattern is produced allowing higher quality identification of the phosphopeptides 18. Overall, hundreds to thousands of phosphoproteins have been identified from different biological samples using these methods 15, 18, 22.
To provide insight into the dynamics of these phosphorylation modules, laboratories have begun to quantify changes in phosphorylation events induced by different biological conditions. Quantification can be achieved by comparing a peptide with an identical peptide that is labeled with heavy isotopes (e.g. 13C or 15N) 23, 24. Given that a mass spectrometer can recognize the mass difference between light and heavy peptides, an abundance ratio between the labeled and unlabeled peptides can then be calculated from the respective ion chromatograms 25, 26. To label a protein sample with stable isotopes, either metabolic labeling or in vitro labeling can be employed 27, 28. Alterations in protein expression induced by a stimulus can be determined by analyzing two samples utilizing the same labeled internal standard 25. Metabolic labeling has advantages over in vitro labeling techniques since it exploits the cell’s translational machinery to label all the proteins, while some in vitro labeling techniques use chemical reactions to label proteins with only certain functional groups 29. In addition, some in vitro techniques label peptides after digestion, and then the light and heavy samples are mixed, while metabolic labeling allows for the mixture of light and heavy samples before any sample preparation, such as the isolation of a specific organelle. Thus, metabolic labeling reduces the systematic errors that may accumulate during sample preparation between the heavy and light samples 25. Metabolic labeling is routinely used in simple systems, such as yeast and cultured mammalian cells, and has even been applied to simple organisms, including C. elegans and D. melanogaster, to quantify hundreds to thousands of unmodified and phosphorylated peptides 14, 30. In comparison, few studies have performed large-scale quantitative phosphorylation analysis on mammalian tissue, and those that have employed in vitro labeling techniques 31, 32.
In order to study animal models of disease, we developed the strategy SILAM (Stable Isotope LAbeling of Mammals) to metabolically label an entire mammal for quantitative MS analysis 33, 34. This strategy combines the necessity of studying mammalian tissues with the quantitative advantage of metabolic labeling. We previously demonstrated that labeling a rat with 15N for two generations had no adverse health effects and generated an entire animal highly enriched with 15N that was phenotypically normal 34. We validated the SILAM strategy by quantifying alterations in unmodified peptides in liver tissue induced by a systemic injection of cyclohexamide and in brain tissue during postnatal development 33, 35. Here, we demonstrate the use of SILAM for quantitative MS analysis of phosphorylation in mammalian brain during development.
Experimental Section
Nuclear enriched sample preparation
Sprague-Dawley rats were labeled with 15N as previously described 33, 34. Briefly, a female rat was fed a 15N labeled protein diet starting after weaning, remaining on the 15N protein diet throughout its pregnancy, and while feeding its pups. On postnatal day 45 (p45), the pups were subjected to halothane by inhalation until unresponsive, and the brains were quickly removed, frozen with liquid nitrogen, and stored at −80°C. The 15N enrichment was determined to be 94% using a previously described protocol 36. Brains from unlabeled Sprague-Dawley rats at p1 and p45 were obtained and stored in an identical manner as the 15N labeled brains except they were quickly dissected before freezing. All methods involving animals were approved by the Institutional Animal Research Committee and accredited by the American Association for Accreditation of Laboratory Animal Care.
Three snap-frozen p1 and p45 rat brain cortices, as well as 15N enriched rat brain were homogenized in the buffer (1 g of tissue/10 ml of buffer) containing 4mM HEPES, 0.32 M sucrose, protease and phosphatase inhibitors(Roche, Indianapolis, IN) in a Teflon hand held dounce grinder. After determining the protein concentration with a BCA protein assay (Pierce, Rockford, IL), homogenates from either p1 or p45 cortices were mixed at a 1:1(wt/wt) ratio with the 15N brain homogenate. The 14N/15N mixture was added to 10ml of buffer and then, was centrifuged at 800 × g for 15 minutes. The resulting pellets were resuspended in 1ml of buffer containing 0.5% NP-40, and then, incubated on ice for 2 hours. The lysate was added to 10ml of buffer and centrifuged at 800 x g for 15 minutes. The resulting 0.5% NP-40 insoluble pellets were nuclear fraction 37 and were homogenized in 500ul of buffer and protein concentration was determined with a BCA protein assay.
Western Analysis
Nuclear preparations were solubilized in 2% SDS, separated by 8-16 % Tris-Glycine gel(Invitrogen, Carlsbad, CA), and electrophoretically transferred to polyvinylidene difluoride membranes (Millipore, Billerica, MA). Membranes were probed with the following antibodies: rabbit glutamate receptor 2/3 antibody and H3 histone antibody (Upstate Biotechnology, Temecula, CA). The membranes were then incubated with perioxidase-conjugated AffiPure goat anti-mouse or anti-rabbit secondary antibodies (Jackson Immunoresearch, West Grove, PA), and the immunoreactive bands were visualized using SuperSignal West Pico chemiluminscent substrate (Pierce, Rockford, IL).
Trypsin digestion and enrichment of phosphopeptides using Immobilized Metal Affinity Chromatography (IMAC)
One milligram of the p1 and the p45 14N/15N mixtures were precipitated with trichloroacetic acid at a final concentration of 20% for 30 minutes, and washed twice with cold acetone. The pellets were then solublized by sonication with 4 M urea/5x Invitrosol (Invitrogen, Calsbad, CA), reduced with 10 mM dithiothreitol and alkylated with 10 mM iodoacetomide, both for 30 minutes. The solutions were diluted with 4x volumes of 100 mM Tris-HCl, and then digested with trypsin (1:100 enzyme/substrate) overnight at 37 °C. After digestion, the enzymatic reaction was terminated using 5% acetic acid.
The enrichment of phosphopeptides was performed using a gallium-based IMAC column (Pierce, Rockford, IL), according to manufacturer’s protocols with minor modification. Briefly, about 100 μg of protein digest in 5% of acetic acid was loaded onto each IMAC column. After two washes with 0.1% acetic acid and two washes with 0.1% acetic acid plus 10% acetonitrile, the bound peptides were eluted four times with 20 μl of 100 mM ammonium bicarbonate, pH 9. The resulting eluate was acidified with 5% formic acid before mass spectrometry analysis.
Analysis of phosphopeptides by Multi-Dimensional Protein Identification Technology (MudPIT) and Linear Iontrap-Orbitrap
The eluted peptides from each IMAC column were analyzed by one MudPIT experiment, and each sample was analyzed in triplicate. As a quantitative control, digestions without phosphopeptide enrichment from either p1 or p45 14N/15N mixture were also analyzed in triplicate. The MudPIT experiment was based on a previous method 38 with modifications tailored to phosphopeptide analysis. Peptides were pressure-loaded onto a 100-μm i.d. fused silica capillary column packed with a 5 cm long, 5 μm Partisphere strong cation exchanger (SCX, Whatman, Clifton, NJ) and a 5 cm, 5 μm Gemini C18 material (Phenomenex, Ventura, CA), with the SCX end fritted with immobilized Kasil 1624 (PQ Corperation, Valley forge, PA). After desalting, a 100-μm i.d. capillary with a 5-μm pulled tip packed with 15 cm 5-μm Jupiter C18 material was attached to the SCX end with a ZDV union, and the entire column was placed inline with an Eksigent pump (Eksigent Technologies, Dublin, CA) and analyzed using a 4-step separation. Three buffer solutions used were: 5% acetonitrile/0.1% formic acid (buffer A); 80% acetonitrile/0.1% formic acid (buffer B), and 500 mM ammonium acetate/5% acetonitrile/0.1% formic acid (buffer C). The first step consisted of a 100 min gradient from 0-100% buffer B. Steps 2-3 had the following profile: 3 min of 100% buffer A, 5 min of X% buffer C, a 10 min gradient from 0-15% buffer B, and a 130 min gradient from 15-45% buffer B, followed by a 20 min gradient increase to 100% buffer B, and a reverse of gradient to 100% buffer A. The 5 min buffer C percentages (X) were 30, 70% and 100% respectively. As peptides were eluted from the microcapillary column they were electrosprayed directly into a hybrid LTQ linear ion trap and Orbitrap (Thermo, San Jose, CA) with the application of a distal 2.4 kV spray voltage. A cycle of one full-scan with 60,000 resolution at 400 m/z by Orbitrap (400-1400 m/z) followed by five data-dependent MS2 scan plus neutral loss-dependent MS3 scan by LTQ was repeated continuously throughout each step of the multidimensional separation. Normalized collision energy of 35% was used while acquiring the MS2 and MS3 spectra.
Identification, quantification of phosphopeptides and phosphoproteins; bioinformatic analysis
MS2 and MS3 spectra were analyzed using the following software analysis protocol. Both spectra were searched with the ProLucid algorithm 39 against the EBI rat IPI database (ftp://ftp.ebi.ac.uk/pub/databases/IPI/, version 3.17, releasing date May 18, 2006), that was concatenated to a decoy database in which the sequence for each entry in the original database was reversed. The search parameters include a static cysteine modification of 57.02146 amu and differential modification on serine, threonine and tyrosine residues of 79.9663 amu. Trypsin specificity was required for all peptides. The database search results were assembled and filtered using the DTASelect program with a spectra level false positive rate of less than 0.5%, mass accuracy of 5 ppm. Under such filtering conditions, the estimated false positive rate was below 1% at the peptide level and below 3% at the protein level.
The assembled search result file was used to obtain quantitative ratios between 14N (sample) and 15N (reference) using the software Census 40. Census allows users to filter peptide ratio measurements based on a correlation threshold because the correlation coefficient (values between zero and one) represents the quality of the correlation between the unlabeled and labeled chromatograms, and can be used to filter out poor quality measurements. In this study, only peptide ratios with correlation values greater than 0.5 were used for further analysis. Because both p1 and p45 cortices were mixed at 1:1 ratio with the common internal standard 15N brain, the ratio difference between p1 and p45 ratios for the same phosphopeptide represents the difference in the phosphorylation level at that specific site in the protein. For singleton analysis, we required the 14N/15N ratio to be greater than 5.0 and the threshold score to be greater the 0.5. The threshold score ranges from zero to one and represents the quality of the singleton analysis with one being the most stringent. For any peptides that were assigned a 14N/15N ratio and determined to be singleton, we relied on the 14N/15N ratio and it was not considered a singleton. In addition, the resulting list of phosphopeptides was also validated using an automated phosphorylation validation program Debunker 41. For Debunker filtering, we required a Debunker output score of at least 0.95 for a peptide for it to be considered as a validated phosphopeptide. In the end, the quantified and validated phosphopeptides and proteins were assembled as the final output.
For Gene Ontology (GO) analysis, annotations were obtained from www.geneontology.org. Almost all nuclear proteins were annotated with multiple molecular functions. For the construction of the pie graph (Figure 3C), the first molecular function was chosen.
Figure 3.
Quantification phosphopeptides. (A) Histograms of log transformed phosphopeptide ratios between p1 or p45 cortices and a common internal standard (15N labeled rat whole brain). (B) Number of peptides quantified and Debunker validated phosphopeptides identified in MS2. (C) Number of peptides quantified and Debunker validated phosphopeptides identified in MS3. (D) GO annotation of quantified phosphoproteins in the p1 and p45 cortex using the category of molecuclar function.
The Pearson’s correlation analysis between total and phosphorylated proteins was performed using Prism (San Diego, CA). The pathway analysis and protein-protein interaction network analysis was performed using Ingenuity Pathway Analysis software (Mountain View, CA).
Results
High confidence identification of phosphopeptides from brain nuclear extraction
The experimental procedure is outlined in Figure 1. To assess the general applicability of using 15N enriched tissue as a common internal standard for quantitative phosphoproteomic measurements, as well as to survey the nuclear phosphorylation events associated with postnatal brain development, postnatal day one (p1) and postnatal day forty five (p45) rat brain cortical homogenates were mixed with a 15N labeled p45 rat brain homogenate at a 1:1(wt/wt) ratio. Following a nuclear enrichment, the 14N/15N mixtures were digested with trypsin. The phosphopeptides were enriched using immobilized metal ion affinity chromatography (IMAC), and then analyzed by multi-dimensional protein identification technology (MudPIT) with neutral loss dependent MS3. The resulting spectra were searched with ProLucid 39, and filtered with DTASelect 42 with a peptide false positive rate less than 1%. Finally, the peptides were quantified with Census, which extracts the 14N and 15N chromatograms for each peptide and determines the 14N/15N ratio using linear regression analysis 43. Each preparation was analyzed in triplicate, and the results were pooled to generate the final output.
Figure 1.

An outline of the experiment. Postnatal day one (p1) and postnatal day forty five (p45) rat cortical homogenates were mixed with a 15N p45 whole rat brain homogenate at a 1:1 ratio. Following a nuclear enriched preparation, the 14N/15N mixtures were digested with trypsin. Half of the digests was subject to phosphopeptide enrichment using IMAC, and then analyzed by MudPIT with neutral loss dependent MS3. The other half of the digests was directly analyzed by MudPIT. The resulting spectra were searched with ProLucid and filtered with DTASelect, and then, the identified peptides were quantified with Census. Each preparation was analyzed in triplicate and the results were pooled to generate the final output.
The dramatic enrichment of nuclear proteins was demonstrated by western blot analysis using an antibody against histone H3 comparing a total tissue homogenate and a nuclear enriched preparation (Figure 2A). In contrast, the immunoreactivity of a synaptic marker (ionotropic glutamate receptor 2/3) showed a large decrease in the nuclear sample. From the MudPIT analysis, we identified 785 and 739 proteins resulting from 2614 and 2815 peptide identifications from the p1 and p45 samples, respectively. This represents identifications of both phospho- and non-phosphopeptides (Figure 2B). The p1 cortex yielded 551 proteins from 1523 unique phosphopeptides, and the p45 cortex yielded 560 proteins from 2121 unique phosphopeptides. Thus, phosphopeptides represented 58% and 75% of our identifications from p1 and p45 cortices, respectively. The relatively low percentage of phosphopeptides versus total (modified plus unmodified) peptides might be due to the highly complex nature of brain nuclear extract. Although this number of identified phosphorylated peptides is comparable with other reports in the brain 32, 44, each analysis of the same sample resulted in an increased number of phosphopeptides suggesting more analyses are required for a more complete phosphoproteome coverage of our complex sample (Figure S1). In addition, for more complete phosphoproteome coverage, it has been suggested that the combination of different phosphopeptide enrichment strategies is required 45. Several large-scale analyses of different phosphoproteomes have revealed a consistent trend of the relative abundance of Ser, Thr or Tyr phosphorylation sites15, 22, 31. In our dataset, the distribution showed a similar relative abundance of 84.5%, 13.4%, and 2.1% for Ser, Thr and Tyr, respectively, in both p1 and p45 samples (Figure 2C). The relative ratios of singly, doubly and triply phosphorylated peptides is also in proximity with previous studies 22, 31, which were 74%, 23.6% and 2.4%, respectively (Figure 2D). Overall, our phosphopeptide identifications appear to be consistent with previous phosphoproteomic analyses.
Figure 2.
Phosphopeptide identification. (A) Western blot analysis using antibodies against a nuclear marker (histone H3) and a synaptic marker (GluR2/3). Twenty-five micrograms of total cortical lysate and the nuclear enriched preparation from a p45 rat were compared. (B) A comparison of total protein/peptide identifications and phosphoprotein and phosphopeptide identifications from the p1 and p45 cortex. (C) Relative abundance of serine (pS), threonine (pT) and tyrosine (pY) phosphorylated peptides in p1 and p45 cortex. (D) Distribution of singly, doubly and triply phopshorylated peptides in the p1 and p45 cortex. (E) table shows respectively the number of phosphopeptides identifide from MS2, MS3 spectra, the number of overlapped phosphopeptides identified from both MS2 and MS3 spectra, percent of phosphopeptides identified solely from MS3 spectra, and the number and the percent of identified phosphopeptides that were validated using Debunker.
During the collision activation dissociation (CID) process in an ion-trap mass spectrometer, phosphoserine and phosphothreonine frequently lose phosphoric acid while phosphotyrosine occasionally loses metaphosphoric acid. Thus, the appearance of fragment ions with an m/z 32, 49 or 98 less than the peptide precursor ions in MS2 spectra strongly indicates that the peptide contains at least one phosphoserine, phosphothreonine residue. This neutral loss event is commonly used to trigger a data dependent MS3 in phosphopeptide analysis to increase both the number and confidence of identifications 18, 46. In our study, a prominent precursor ion neutral loss of 98, 49 or 32 in the MS2 scan is selected for further fragmentation, and the fragmentation pattern appears in the MS3 spectra. As illustrated in Figure 2E, 71% and 80% of the phosphopeptides identified from MS3 spectra were also identified from MS2 spectra, providing greater confidence for these phosphopeptides. To further increase our confidence in our phosphopeptide identifications, we applied a machine-learning program, Debunker 41, to validate phosphopeptide identifications derived from the MS2 spectra. Debunker uses a support vector machine binary classification to derive a probability score for phosphopeptide identifications from the CID spectra. Prominent spectral features such as neutral loss of precursor ions, neutral loss of fragment ions, and intensity of b or y ion series are incorporated to calculate a probability score as an assessment of the validity of the phosphopeptide identification. A predictive value from 0 to 1 is assigned to the possible phosphorylation event. A value less than 0.5 indicates that the phosphorylation prediction is negative, while a value greater than 0.5 indicates that the prediction is positive for a phosphorylation event. Values close to 1 are strongly predictive of a phosphorylation event. Requiring a predictive value greater than 0.95 in both samples, 86.5% of the phosphopeptides were determined as highly confident (Figure 2E).
The two sets of validated phosphopeptides were site-localized using a binomial probability approach similar to what has been described previously 22, 47. They were then separately analyzed by Motif-X algorithm 48 to extract phosphorylation sequence motifs. The default significance value was set to 10-6 for the sequence analysis and at least 20 occurences were required to form the motif with the resulting motifs listed in Figure S2 and S3. Largely consistent with a previous analysis on HeLa cell nuclear proteome 18, the major motifs generated from either p1 or p45 cortices were proline directed and acidiphilic sites. Motifs between p1 and p45 cortices were similar except for a single motif, xPxxxKS(p)PxxxKx, which was found exclusively in p45 cortices. Among the proteins that contain this motif are neurofilament proteins (NFs). It has been shown that mitogen-activated protein kinases (Erk1, Erk2) preferentially recognize this motif 49. Phosphorylation of NFs by Erk1 and Erk2 at the KSP motif is thought to result in conformational change of sidearms of NF proteins and affect their association with each other and with microtubules. These phosphorylation events lead to the dynamic reorganization of neuronal cytoskeleton and thus the remodeling of synapse 49, a process that is predominantly seen in adult brain (p45).
Quantification of brain nuclear proteome in different developmental stages
Phosphopeptide ratios from either p1 or p45 cortices against the common 15N internal standard are plotted as histograms in Figure 3A. For p1 cortex, the range of magnitude in phosphopeptide ratios spans from 50 fold less to 10 fold more. In comparison, the range of magnitude for that of p45 cortices spans from 10 fold less to 10 fold more. Overall, we quantified 968 and 1674 phosphopeptides for the p1 and p45 cortices respectively, representing a quantification efficiency of 63.6% and 78.9% (Figure 3B and Table S1 for the complete list). The high confidence of our phosphopeptide identifications also extended to our quantified phosphopeptides. A high percentage (86.5%) of quantified MS2 peptides was validated by Debunker with a probability score of 0.95 or greater. In addition, we quantified the peptides identified from MS3 spectra (Figure 3C and Table S2 for the complete list). Since Census uses peptide chromatograms generated from the MS spectra for quantification 43, the ratios of the quantified MS3 peptides were identical to the ratios of the MS2 peptides from which they were derived. Furthermore, there were phosphopeptides quantified only with the MS3 spectra because the MS2 fragmentation didn’t generate enough sequence information to identify these peptides. Overall, 180 and 137 peptides (29% and 20% of all MS3 peptides) were quantified exclusively from MS3 identifications in p1 and p45 cortex, respectively.
Using Gene Ontology, we determined that 43% (p1 cortex) and 38% (p45 cortex) of the annotated quantified phosphoproteins had nuclear localization. In contrast, 17% of the proteins in the rat GO database are described as nuclear proteins indicating an enrichment of nuclear proteins supporting our western blot analysis. Corresponding to our western blot analysis, there were non-nuclear proteins in our sample. Also, it should be noted that Gene Ontology often annotates a protein with mulitple localization, which reflects the literature. Thus, the percentage of nuclear proteins using the GO analysis may include proteins with that possess both cytoplasmic and nuclear localizations. The distribution of the molecular functions of these nuclear proteins was similar for the p1 and p45 samples and was combined in Figure 3C. Forty-eight percent of the quantified phosphoproteins were annotated to bind or regulate DNA. These were mostly proteins that regulated transcription, including beta-catenin, glucocorticoid receptor DNA-binding factor 1, and methyl-CpG-binding protein 2(MeCP2). Another large percentage of proteins were annotated to bind RNA or regulate RNA, including proteins that regulate RNA splicing. Numerous proteins involved in phosphorylation were quantified, and the majority of the proteins in this group were kinases, including protein kinase C (PKC), glycogen synthase kinase (GSK), 5’-AMP-activated protein kinase(AMPK), MAP/microtubule affinity-regulating kinase 2 (MARK2), and dual specificity tyrosine-phosphorylation-regulated kinase (Dyrk). In addition, phosphatases were also quantified within this functional protein group. Other functional categories quantified were structural proteins such as cytoskeletal proteins and nuclear pore proteins. Thus, we quantified many different functional classes of nuclear phosphoproteins.
It was impossible to derive a 14N/15N ratio for some phosphopeptides because a chromatographic peak for the corresponding 15N internal standard peptide was non-existent or of extremely low signal. A large change in the abundance of phosphopeptide, however, has the greatest probability of indicating biological significance. This scenario has been designated as “singleton” indicating that only the 14N peak was observed 43. Census can re-examine peptides that did not pass the original criteria for a high quality 14N/15N ratio for singleton peptides 43. We observed 191 14N singleton events in the p1 analysis and 102 14N singleton events in the p45 analysis. Since it is possible that the absence of a corresponding 15N peak could indicate the 14N peak is a misidentified peptide, we required singleton peptides to be annotated as nuclear proteins to be considered as true singleton peptides. With this additional requirement, we observed 56 14N singleton nuclear peptides in the p1 analysis and 8 14N singleton nuclear peptides from the p45 analysis (Table S3). We next examined the frequency of these singleton peptides within the three MudPIT analyses of each development time point. In the p1 cortex, one nuclear singleton peptide was observed in all three MudPIT analyses, six nuclear singleton peptides were observed in two of the MudPIT analyses, and the rest were only observed in one MudPIT analysis. In the p45 cortex, one of the nuclear singleton peptides was observed in two MudPIT analyses and the rest were observed in only one of the three MudPIT analyses. Since our 15N internal standard was a p45 whole rat brain, the singleton peptides in the p1 analysis are potentially important phosphorylation events in neuronal development. Furthermore, it was expected that the p45 analysis would yield less singleton peptides since the sample and the internal standard were obtained from rats of the same age. The singleton peptides that were identified in the p45 analysis are most likely due to differences between whole brain and the cortical region, but we cannot rule out an isotopic effect of the 15N labeling.
Clustering and functional classification of nuclear phosphoproteins
To further examine phosphoproteome changes during mouse brain development, we analyzed samples from p1 and p45 cortices using the identical protocol that has been described except we omitted the IMAC enrichment step. We will reference proteins from this analysis as “total” proteins (Table S5). These quantified proteins were employed to determine if the alterations observed with the phosphoproteins were due to changes in phosphorylation events or changes in protein expression. There were 194 proteins quantified in both p1 and p45 samples and both in total and phosphorylation enriched preparations (Table S4). A global comparison of the ratios (p1/p45) between phosphorylated and total proteins by Pearson’s correlation showed that there was statistically significant correlation between the two ratios (Figure 4A, R=0.427, p<0.0001).
Figure 4.
Clustering of phosphoproteins. (A) Correlation of p1/p45 ratios between total protein and phosphorylated protein. Proteins are clustered based on eight categories depending on the changes of either total or phosphorylated protein ratios, with different colors indicating different clusters (See Table S4 for color code). R-value was calculated using Pearson’s correlation (p<0.0001). (B) Number of proteins in each of the eight categories. With respect to p1/p45 ratio, 1 indicates a ratio change of more than or equal to two fold, -1 indicates a ratio change of less than or equal to 0.5, while 0 indicates the ratio was between 0.5 and 2. (C) Ingenuity analysis of significantly changed functional categories using Fisher’s exact test. (D) Protein-protein interaction network that represent the function of cell cycle and DNA replication, recombination and repair. Red indicates an increased level of protein phosphorylation and green indicates a decreased level of phosphorylation.
These 194 proteins were then filtered based on the following criteria: a p1/p45 ratio of greater than 2 was considered as upregulated, while a ratio of less than 0.5 was considered as downregulated. The combinations of either up- or downregulation in either phosphoprotein or total protein categorize the proteins into 8 groups (Figure 4A and 4B). The majority of the phosphoprotein and total protein analyses were in agreement based on these criteria. In both phosphorylated and total protein analyses, 94 proteins showed no change, 35 proteins were observed to be downregulated in the p1 cortex, and 17 proteins were upregulated in the p1 cortex.
The remaining 48 proteins with differential changes between total and phosphoproteins suggest important protein phosphorylation regulation during cortical development. These proteins were further analyzed using the Ingenuity pathway analysis software (http://www.ingenuity.com/). Ingenuity uses Fisher’s exact test to identify significantly over represented functional categories in the input data against the Ingenuity Pathways Knowledge Base. With a significance cutoff value of 0.05 (-log(p)=1.301), the analysis identified proteins in cell cycle, cellular assembly and organization, DNA replication, recombination and repair, as the top three significantly over represented categories (Figure 4C). Several other categories including gene expression, cell morphology, developmental disorder, nervous system development and function were also significantly represented. Consistent with the Gene Ontology annotation results which showed an enrichment of nuclear proteins, all of these functional groups are intimately connected to the nucleus. We observed a high amount of overlap between these functional groups. For example, phosphoproteins involved in the regulation of the cell cycle were the largest common functional group with 14 proteins in this category, and all 10 proteins that were assigned to the category of DNA replication, recombination, and repair were also assigned to the cell cycle category. In addition, ten out of the 13 proteins assigned to the cell assembly and organization group were also assigned to the cell cycle category. This overlap likely stems from multiple biological functions assigned to these phosphoproteins.
Ingenuity also identified several protein-protein interaction modules, the top scored module was involved in cell cycle control, cell assembly and organization, as well as DNA replication, recombination and repair (Figure 4D). In this module, many proteins known to form the SWI/SNF chromatin remodeling complex, including SMARCA4 (Brg1), SMARCC2, polybromo 1 (PBRM1), chromodomain helicase DNA-binding protein 4 (CHD4), and remodeling and splicing factor (RSF1) 50, 51, were found to have upregulated phosphorylation events in p1 compared to p45 with Brg1 showing the greatest increase in phosphoryation at S1382. With the exception of transformer-2 alpha (TRA2A) and importin alpha 4 (KPNA3), the other 10 proteins identified in this network showed increase in phosphorylation in the p1 cortex. Therefore, it appears that many proteins controlling cell growth and chromatin remodeling are regulated by phosphorylation at specific sites in rat cortices during the early stage of brain development.
Regulated phosphorylation at different sites of the same protein
It is widely observed that different phosphorylation sites on the same protein can be differentially regulated. For example, it has been demonstrated that acute administration of the psychostimulant, amphetamine, causes an increase in the phosphorylation of the N-methyl-D-aspartate receptor NR1 subunit at serine 890 and 896, but not serine 897 in the brain52. As depicted in Figure 5A and 5B, two different proteins, heterogeneous nuclear ribonucleoprotein C (hnRNP C) and methyl-CpG-binding protein 2 (MeCP2), exhibited distinct abundance distribution patterns of phosphorylation at different residues. hnRNP C is a member of the heterogeneous nuclear ribonucleoproteins family of proteins that have been reported to be involved in DNA repair, telomere biogenesis, and gene expression 53. MeCP2 is a member of the family of methyl-CpG-binding domain proteins that function as long-range transcriptional repressors that mediate developmental silencing by binding to methylated DNA 54, 55. Phosphopeptides derived from hnRNP C showed both up- and downregulation in the p1 cortex, while phosphopeptides from MeCP2 showed either downregulation or no change in the p1 cortex. In contrast, peptides derived from total protein analysis of those two proteins showed much unanimous distribution pattern, either no change for hnRNP C, or downregulation of MeCP2 in the p1 cortex (Figure 5, C and D). We next plotted the distribution of standard deviation of the relative phosphoproteins ratios for all the proteins quantified in either the phosphorylated or total protein samples (Figure 5E). The distribution curves for standard deviation of quantified phosphoproteins appears to overlap with that of quantified total proteins with a few exceptions. This suggests that differentially regulated phosphorylation at different sites of the same protein is not a frequent occurrence in our quantitative analysis. However, it is possible that such differentially phosphorylated proteins may be below the limits of detection of the mass spectrometer or may be transient in nature preventing their detection.
Figure 5.
Phosphorylation levels are regulated at different sites for a subset of proteins. Shown are distribution of quantification ratios of multiple phosphopeptides with sites indicated (A) and multiple unmodified peptides (B), for a protein hnRNP C. (C and D) Similar to (A and B) except that the ratio distributions shown are for MeCP2. (E) Distribution of standard deviation of protein ratios calculated by Census. Here the standard deviation is the precision of peptide ratio measurements that are used to calculate protein ratio. The curve shows the percentage of proteins from either total proteins or phosphoproteins in each interval of standard deviation values.
Two specific examples of regulation of phosphorylation restricted to a discrete portion of a protein are shown in Figure 6. Figure 6A and 6B show a previously undocumented phosphopeptide, MPFQAS(p)PGGK (S229), from the developmentally regulated transcription factor MeCP2. There was a slight increase of this peptide in p1 relative to p45 cortex. Figure 6C and 6D showed phosphorylation of two serine residues of MeCP2, AETS(p)ESSGSAPAVPEASAS(p)PK, with the second serine being designated as S80 by a previous study 31. We observed dramatically less phosphorylation of this peptide in p1 cortex. Interestingly, there is almost no change between p1 and p45 cortex of another phosphopeptide, AGS(p)LESDGCPKEPAK (S421), a site previously reported to be phosphorylated in an neuronal activity dependent manner 56. As a reference, the peptide derived from total protein analysis revealed no changes between p1 and p45 cortex (Figure 6E and 6F). We also observed site-specific regulation of phosphorylation for the transcriptional regulator Brg1. Brg1 has been shown to play a critical role in mammalian neuronal differentiation 57. We found that for a documented phosphopeptide , EVDYSDS(p)LTEK (S1382) 18, there was an over 15 fold increase in S1382 phosphorylation in p1 compared with p45 cortex (Figure 6G and 6H), but a much less (3 fold) increase in a peptide derived from total protein (Figure 6I and 6J), indicating a dramatic increase in phosphorylation at this site in early brain development, but a decrease when the brain approaches maturity. The MS2 spectra of above mentioned phosphopeptides were shown in Figure S4. Thus, regulated phosphorylation at specific sites of these proteins may play important roles driving postnatal brain development.
Figure 6.

Reconstructed chromatograms for peptides from two selected proteins. (A through D) shows peptide precursor ion chromatograms (blue) for two different phophopeptides of MeCP2 from either p1 or p45 cortex, against that of the common internal standard (red). The calculated ratio (sample/reference) by Census is shown in each chromatogram. The chromatograms for an unmodified peptide from either p1 or p45 are shown in E and F. (G and H) Chromatograms of a phosphopeptide from Brg1 shows ratios of either p1 or p45 against the 15N peptide, while the p1 or p45 ratios of an unmodified peptide from Brg1 of either against the 15N peptide are shown in I and J. The phosphorylation site was indicated as (p).
Discussion
In this study, we used 15N labeled rat tissue as an internal standard combined with IMAC-based phosphopeptide enrichment to quantify protein phosphorylation in different stages of postnatal brain development. Our IMAC strategy consistently resulted in more than 50% of identifications being phosphopeptides facilitating the quantification of phosphopeptides. In total, we quantified over 1500 phosphopeptides in each sample giving rise to over 500 quantified phosphoproteins. Additionally, by clustering the quantified nuclear phosphoproteins based on their abundance of phosphorylation, we found that there was a significant correlation between the phosphorylation level changes and the total protein expression changes from different developmental stages of rat cortex. Finally, we discovered regulated, site-specific phosphorylation changes in two important transcriptional regulators, MeCP2 and Brg1.
Even with phosphopeptide enrichment strategies, the low stoichiometry of phosphorylation events often limits the identification of a phosphoprotein to one peptide. We have applied several strategies, however, to achieve high confidence in our identification and quantification of phosphopeptides. First, a decoy database search strategy 58 was used to control the false positive rate at peptide level to be below 1%. Second, after the identification of peptides, a support vector machine algorithm, Debunker, was applied to derive a binary classifier for each peptide filtering out peptides from low-quality spectra and the spectra lacking phosphopeptide fragmentation patterns commonly observed in CID 41. In this study, only a small fraction of identified phosphopeptides (<15%) were filtered out based on stringent criteria. Third, using an Orbitrap mass analyzer to acquire MS spectra enabled us to distinguish phosphorylation from other modifications of similar mass. For example, protein sulfonation at each serine or threonine residue results in a molecular mass increment of 79.9568 59, a mass shift that is 0.0095 amu different from that of phosphorylation which can be distinguished for peptides that are under 1900 Da by Orbitrap with a 5 ppm accuracy. In addition, the increased accuracy of peptide precursor ion measurement greatly improves the quantification using extracted chromatograms 60. Finally, neutral loss dependent acquisition of MS3 spectra was applied to further increase the confidence of the identifications. A low percentage (~20%) of MS2 identifications were confirmed by MS3, which is consistent with previous observations 18, 61. There are several potential reasons as to why a phosphopeptide may be identified only from the MS2 scan. It is most likely that the number of fragment ions in the MS3 scan is not large enough to identify a phosphopeptide due to the insufficient trapping of the neutral loss peptide ions. Alternatively, an MS3 scan event may not be triggered when a phosphopeptide analyzed by an MS2 scan does not undergo complete neutral loss of phosphate under CID 32, or proline-directed fragmentation in MS2 generates ions that are more abundant than the neutral loss peptide ions. However, over 70% of the identifications generated from MS3 spectra matched that of MS2 spectra. The remaining, high-quality identifications from MS3 spectra thus provided additional identifications, and these can increase the number of quantified phosphopeptides as a whole. The mechanism underlying phosphopeptides identified solely from MS3 scans is that CID in MS2 may generate a dominant neutral loss peptide ion and less or undetectable fragment ions, which leads to a complete lack of peptide identifications or the identification of an incorrect phosphopeptide. Thus, employing MS3 analysis, Debunker, decoy database searching, and a high resolution and high mass accuracy mass spectrometer generate a particularly confident quantified phosphoprotein analysis.
A previous report performed global mass spectrometry analysis of nuclear phosphoproteins from a HeLa cell line 18. Although different nuclear extract methods were performed, this report also identified greater than 50% contaminant or unknown proteins. A direct comparison of individual phosphoproteins between these studies is difficult, because our starting material consisted of multiple rat cell types embedded within extracellular matrices, while the sample of this previous report consisted of a single human cell type cultured in vitro. In addition, different phosphopeptide enrichment strategies were employed between the two studies. However, both reports demonstrated the identification of a large percentage of DNA binding proteins, kinases, and proteins involved in protein splicing. Furthermore, both reports identified proline-directed and acidiphilic kinase motifs as the major motif identified suggesting this dominants nuclear phosphorylation. In addition to phosphopeptide identification, our study focused on quantification of phosphopeptides. We have previously employed SILAM analysis to quantify proteins from both liver and brain tissues 33, 35. In the brain analysis, we identified changes in protein expression during postnatal development, and over 50% of the annotated proteins quantified have been previously described in the literature using other quantification methods 35. Nevertheless, quantification of phosphoproteins provides unique challenges to mass spectrometry. The largest challenge is that phosphorylation sites can be differentially regulated on the same protein, as demonstrated by our analysis of MeCP2 and Brg1. Although many laboratories have successfully performed large-scale phosphoprotein quantification, there are very few laboratories that have combined the complexity of mammalian tissue with quantitative phosphorylation analysis. One report quantified a small number of phosphoproteins from brain 31, while more recently in another report, the iTRAQ strategy was employed to quantify 1339 phosphorylation sites from the post-synaptic density (PSD) from brain 32. ITRAQ labels and mixes peptides after the isolation of the PSD and trypsin digestion, while using metabolically labeling, the 14N and 15N samples are mixed before fractionation, which reduces the accumulation of the systematic errors during sample preparation. Because of differences in phosphopeptide enrichment strategies and mass spectrometers employed, it is difficult to compare quantitative phosphoproteome analysis using SILAM to the analysis used by Trinidad et al., but future studies directly comparing these two strategies would be valuable.
Our results also highlight the importance of using an unmodified protein ratio as a control to interpret the phosphoprotein ratio changes. We observed the majority of the phosphopeptides have a similar expression pattern as the unmodified peptides from the same protein. Among the proteins quantified with both phosphorylation ratios and total ratios, over 70% of the proteins were either unchanged or the change in expression occurred concurrently. This is in contrast with EGF signaling, where site-specific phosphorylation levels frequently changed without total protein changes 22. This might be the result of the different systems analyzed in these two studies. First, the study of EGF signaling was performed on a less complex system, mammalian cell culture, and under EGF stimulation to achieve signal transduction, while we were comparing the differences in protein phosphorylation in brain tissues between two developmental stages. Furthermore, in the EGF signaling cascade, the entire signal transduction occurs rapidly in the time scale of minutes. While in our study of postnatal brain development, the signaling event happens in the time scale of weeks 62 allowing the steady growth of neurons and glia to increase the brain mass. It is therefore conceivable that many important signaling proteins are up- or down regulated as a whole together with the phosphorylation of specific sites. In addition, Trinidad et al 32 observed similar trends between protein expression and phosphorylation in their analysis of brain tissue. Thus, this indicates that the biological significance of many of the phosphorylation events we quantified is equally crucial to the p1 cortex as to the p45 cortex.
Alterations in the expression of phosphopeptides between p1 and p45 samples, however, did not always correspond to the alterations in the expression of the unmodified peptides. For example, the average p1/p45 ratio of the phosphopeptides from the Brg1 protein was 7.5 while the average p1/p45 ratio of the unmodified peptides was 1.7 suggesting a large increase in phosphorylation of Brg1 at p1 compared to p45. Alternatively, the unmodified Rap1-interacting factor 1 (RIF1) expression was low (-5.9) at p1 compared to p45 while the phosphopeptides of RIF1 were relatively unchanged (1.85) between the p1 and p45 samples indicating more phosphorylation at p1 compared to p45. These observations suggest that these phosphorylation events are important for cellular processes early in postnatal brain development. Previous reports examining individual phosphorylation events have also demonstrated changes in protein phosphorylation during brain development 63-65. For example, it has been reported that in juvenile mouse cortices, visual stimulation induces the phosphorylation of mitogen- and stress-activated kinase (MSK), cAMP responsive element-binding protein (CREB), and histone H3, while this induced phosphorylation of CREB and H3, but not MSK, was greatly reduced in adult mouse cortices 65. Therefore, we have identified novel regulated phosphorylation on known sites that could provide further understanding into the signaling of the developing cortex.
Cerebral cortical development is a dynamic process that requires complex but precise coordination of cell proliferation, death, migration, differentiation, dendritic arborization, and synaptogenesis. Many of the phosphorylation signaling pathways are involved in this process, including mulitple mitogen-activated protein kinase pathways and the Wnt pathway 66, 67. These pathways are activated by a variety of extracellular growth factors and result in the phoshorylation of nuclear proteins to alter transcription. It is still unknown, however, how these mutliple phosphorylation signaling pathways work together to promote the precise coordination of the numerous biological processes during development. Adding to the complexity is that the cortex consists of multiple different types of neurons in addition to different types of glial cells, and the localization of specific growth factor receptors can vary dramatically between different cortical regions68. Thus, our analysis most likely represents global or the most abundant nuclear changes in phosphorylation found throughout the cortex. By using Ingenuity pathway analysis and the literature, we observed that many of the proteins forming the chromatin remodeling complex, and proteins that interact with the complex had an increase in phosphorylation in the p1 cortex. Neurogenesis peaks at birth for most neurons in rat brain, and synaptogenesis peaks between p14 and p21 62. The phosphorylation of Brg1 and other related proteins might play an important role in these processes. Brg1 is a component of the SWI/SNF chromatin remodeling complex with an evolutionarily conserved DNA-dependent ATPase domain 69. Consistent with our findings, previous studies have shown the requirement of Brg1 phosphorylation in mitotic cells for the SWI/SNF complex to be excluded from condensed chromatin, and this might be part of the mechanism leading to transcriptional arrest during mitosis 70. It was later shown that Brg1 is required for neural differentiation in mammals through interaction with Ngnr1 and NeuroD transcription factors 57. Chromatin remodeling, DNA modification and histone modification is known to play a crucial role in activity-dependent dendritic development and neuronal plasticity 71, 72. A recent study demonstrated that the neuronal chromatin-remodeling Brg/Brm-associated factor (nBAF) complex promotes dendritic morphogenesis, and that the process involves a combinatorial selection of an actin-interacting protein BAF53b and requires Brg/Brm ATPase activation 73. Therefore, our finding of increased phosphorylation at specific sites of Brg1 and related proteins will likely provide further insights for the understanding of how neuronal activity lead to the control of dendritic growth, neurogenesis and therefore brain development.
Neurons and glia are two main classes of cells making up the nervous system, and both encompass many specialized sub-types. As these cells exit the cell cycle, they migrate and differentiate establishing the form of the nervous system. In humans, these processes occur between the third trimester and the first years of life. In the rat, these same processes are condensed into the first month of postnatal life, and thus, generate an experimental model for human brain development. The molecular mechanisms underlying human brain development are not fully elucidated, but further research can provide potential therapies for poorly understood neurodevelopmental disorders, such as Rett syndrome, autism, and schizophrenia 74-76. In Rett syndrome for example, mutations in MeCP2 have been demonstrated to cause the majority of the cases, but how these mutations cause the deleterious phenotype is unknown75. One report suggests that the phosphorylation of MeCP2 plays a role in progression of the disease 56. Since phosphorylation plays an essential role in neural activity, global quantitative phosphorylation analysis becomes indispensable for neuroscience research. For the first time, we demonstrate the value of employing a metabolically labeled rat tissue for global quantitative phosphorylation analysis of brain tissue and highlight the importance of phosphorylation events in neuronal development.
Supplementary Material
There are four supplemental figures and five supplemental tables in this manuscript. Figure S1 shows the total number of phosphopeptides in each of the accumulating MudPIT runs. Figure S2 and S3 list the sequence motif of phosphopeptides from p1 and p45 cortical nuclear proteome. Figure S4 shows the annotated MS2 spectra of the phosphopeptides quantified and illustrated in Figure 6. Table S1 lists Ratios of phosphopeptides identified by MS2 from p1 or p45 cortex over a common internal standard (15N labeled rat brain). Table S2 lists ratios of phosphopeptides identified by MS3 from p1 or p45 cortex over a common internal standard (15N labeled rat brain). Table S3 lists phosphopeptides with large differences in phosphorylation ratios (singleton peptides). Table S4 lists phosphoprotein and total protein ratios between p1 and p45 cortex. Table S5 lists the ratios of total proteins from p1 or p45 cortex over a common internal standard (15N labeled rat brain).
Supporting Information Softwares ProLucid, Census and Debunker are available for download via the following website: http://fields.scripps.edu/download.php.
The MS2 and MS3 spectra of the reported phosphopeptides are available at the author’s website: http://fields.scripps.edu/yateslab/public/08_jpr_liao/
Supporting figures and tables are available free of charge via the Internet at http://pubs.acs.org
Ratios of phosphopeptides identified by MS2 from p1 or p45 cortex over a common internal standard (15N labeled rat brain). Column A lists peptide sequences with phosphorylation site indicated as (p). Column B and C list the protein index and name, respectively, from which the peptide sequence was derived. Column D lists the ratio of each phosphopeptide derived from p1 cortex over the same peptide derived from 15N brain. Column E, same as Column D except that the ratio was p45 cortex over 15N brain. Column F and G list the probability value (generated by Debunker) of each peptide listed in Column A, identified in p1 and p45, respectively. Column H lists the charge state of each peptide listed in Column A.
Ratios of phosphopeptides identified by MS3 from p1 or p45 cortex over a common internal standard (15N labeled rat brain). Same as Table S1 except that the phosphopeptides were identified by MS3.
List of phosphopeptides with large differences in phosphorylation ratios (singleton peptides). Column A lists peptide sequences with phosphorylation site indicated as (p). Column B and C list the protein index and name, respectively, from which the peptide sequence was derived. Column D lists the ratio of each phosphopeptide derived from p1 cortex over the same peptide derived from 15N brain. Column E, same as Column D except that the ratio was p45 cortex over 15N brain. Column F and G list the ratio of each protein from p1 or p45 cortex respectively over the protein from 15N brain. Column H and I, probability value (generated by Debunker) of each peptide listed in Column A, identified in p1 and p45, respectively. Column J, charge state of the phosphopeptide in Column A.
Phosphoprotein and total protein ratios between p1 and p45 cortex. Column A and B, protein index and name respectively. Column C, annotation of cellular localization for each protein.Column D and E, relative ratio between p1 and p45 cortex of phosphoproteins (D) and total proteins (E). Column F and G, labels of relative expression ratios of phosphoproteins (F) and total proteins (G) between p1 and p45, whereby, -1 indicates a ratio of <0.5, 0 indicates a ratio of between 0.5 and 2.0, and 1 indicates a ratio of >2.0. Column H list the cluster number of the proteins based on their relative expression ratio changes in phosphoprotein as well as total protein. Column I gives the color code used to plot the correlation graph in Figure 4A.
Ratios of total proteins from p1 or p45 cortex over a common internal standard (15N labeled rat brain). Column A and B, protein index and name respectively. Column C, the ratio of each protein derived from p1 cortex over the same protein derived from 15N brain. Column D, standard deviation (S.D.) of multiple peptide ratios used to derive the protein ratio. In cases where one peptide was used to identify the protein, S.D. was not available (n/a). Column E and F, same layout as Column C and D except that the protein ratios were from p45 cortex. Note that some proteins were quantified in either p1 or p45 cortex but not in both, in such cases the sample with the proteins not quantified labeled as “n/a”.
Total number of phosphopeptides increase as the function of the increasing number of MudPIT runs.
Sequence motif logo of phosphopeptides from p1 and p45 cortical nuclear proteome.
Sequence motif logo of phosphopeptides from p1 and p45 cortical nuclear proteome.
Annotated MS2 spectra of the phosphopeptides quantified and illustrated in Figure 6.
Acknowledgments
The authors thank Yates’ lab members for critical reading of the manuscript, and acknowledge financial support from National Institutes of Health grants BIMR P30 NS057096, 5R01 MH067880-02 to JRY.
References
- 1.Johnson SA, Hunter T. Kinomics: methods for deciphering the kinome. Nat Methods. 2005;2(1):17–25. doi: 10.1038/nmeth731. [DOI] [PubMed] [Google Scholar]
- 2.Meyer T, Vinkemeier U. STAT nuclear translocation: potential for pharmacological intervention. Expert Opin Ther Targets. 2007;11(10):1355–65. doi: 10.1517/14728222.11.10.1355. [DOI] [PubMed] [Google Scholar]
- 3.Huang H, Tindall DJ. Dynamic FoxO transcription factors. J Cell Sci. 2007;120(15):2479–87. doi: 10.1242/jcs.001222. [DOI] [PubMed] [Google Scholar]
- 4.Nagaraj K, Hortsch M. Phosphorylation of L1-type cell-adhesion molecules--ankyrins away! Trends Biochem Sci. 2006;31(10):544–6. doi: 10.1016/j.tibs.2006.07.010. [DOI] [PubMed] [Google Scholar]
- 5.Raymond LA, Blackstone CD, Huganir RL. Phosphorylation of amino acid neurotransmitter receptors in synaptic plasticity. Trends Neurosci. 1993;16(4):147–53. doi: 10.1016/0166-2236(93)90123-4. [DOI] [PubMed] [Google Scholar]
- 6.Limback-Stokin K, Korzus E, Nagaoka-Yasuda R, Mayford M. Nuclear calcium/calmodulin regulates memory consolidation. J Neurosci. 2004;24(48):10858–67. doi: 10.1523/JNEUROSCI.1022-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Chwang WB, Arthur JS, Schumacher A, Sweatt JD. The nuclear kinase mitogen- and stress-activated protein kinase 1 regulates hippocampal chromatin remodeling in memory formation. J Neurosci. 2007;27(46):12732–42. doi: 10.1523/JNEUROSCI.2522-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Wood MA, Hawk JD, Abel T. Combinatorial chromatin modifications and memory storage: a code for memory? Learn Mem. 2006;13(3):241–4. doi: 10.1101/lm.278206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Alonso AC, Zaidi T, Grundke-Iqbal I, Iqbal K. Role of abnormally phosphorylated tau in the breakdown of microtubules in Alzheimer disease. Proc Natl Acad Sci U S A. 1994;91(12):5562–6. doi: 10.1073/pnas.91.12.5562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Iqbal K, Grundke-Iqbal I. Developing pharmacological therapies for Alzheimer disease. Cell Mol Life Sci. 2007;64(17):2234–44. doi: 10.1007/s00018-007-7221-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Mansuy IM, Shenolikar S. Protein serine/threonine phosphatases in neuronal plasticity and disorders of learning and memory. Trends Neurosci. 2006;29(12):679–86. doi: 10.1016/j.tins.2006.10.004. [DOI] [PubMed] [Google Scholar]
- 12.Matsuoka S, Ballif BA, Smogorzewska A, McDonald ER, 3rd, Hurov KE, Luo J, Bakalarski CE, Zhao Z, Solimini N, Lerenthal Y, Shiloh Y, Gygi SP, Elledge SJ. ATM and ATR substrate analysis reveals extensive protein networks responsive to DNA damage. Science. 2007;316(5828):1160–6. doi: 10.1126/science.1140321. [DOI] [PubMed] [Google Scholar]
- 13.Andersson L, Porath J. Isolation of phosphoproteins by immobilized metal (Fe3+) affinity chromatography. Anal Biochem. 1986;154(1):250–4. doi: 10.1016/0003-2697(86)90523-3. [DOI] [PubMed] [Google Scholar]
- 14.Cantin GT, Venable JD, Cociorva D, Yates JR., 3rd Quantitative phosphoproteomic analysis of the tumor necrosis factor pathway. J Proteome Res. 2006;5(1):127–34. doi: 10.1021/pr050270m. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Villen J, Beausoleil SA, Gerber SA, Gygi SP. Large-scale phosphorylation analysis of mouse liver. Proc Natl Acad Sci U S A. 2007;104(5):1488–93. doi: 10.1073/pnas.0609836104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Pinkse MW, Uitto PM, Hilhorst MJ, Ooms B, Heck AJ. Selective isolation at the femtomole level of phosphopeptides from proteolytic digests using 2D-NanoLC-ESI-MS/MS and titanium oxide precolumns. Anal Chem. 2004;76(14):3935–43. doi: 10.1021/ac0498617. [DOI] [PubMed] [Google Scholar]
- 17.Pinkse MW, Mohammed S, Gouw JW, van Breukelen B, Vos HR, Heck AJ. Highly robust, automated, and sensitive online TiO2-based phosphoproteomics applied to study endogenous phosphorylation in Drosophila melanogaster. J Proteome Res. 2008;7(2):687–97. doi: 10.1021/pr700605z. [DOI] [PubMed] [Google Scholar]
- 18.Beausoleil SA, Jedrychowski M, Schwartz D, Elias JE, Villen J, Li J, Cohn MA, Cantley LC, Gygi SP. Large-scale characterization of HeLa cell nuclear phosphoproteins. Proc Natl Acad Sci U S A. 2004;101(33):12130–5. doi: 10.1073/pnas.0404720101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Rush J, Moritz A, Lee KA, Guo A, Goss VL, Spek EJ, Zhang H, Zha XM, Polakiewicz RD, Comb MJ. Immunoaffinity profiling of tyrosine phosphorylation in cancer cells. Nat Biotechnol. 2005;23(1):94–101. doi: 10.1038/nbt1046. [DOI] [PubMed] [Google Scholar]
- 20.DeGnore JP, Qin J. Fragmentation of phosphopeptides in an ion trap mass spectrometer. J Am Soc Mass Spectrom. 1998;9(11):1175–88. doi: 10.1016/S1044-0305(98)00088-9. [DOI] [PubMed] [Google Scholar]
- 21.Steen H, Kuster B, Fernandez M, Pandey A, Mann M. Detection of tyrosine phosphorylated peptides by precursor ion scanning quadrupole TOF mass spectrometry in positive ion mode. Anal Chem. 2001;73(7):1440–8. doi: 10.1021/ac001318c. [DOI] [PubMed] [Google Scholar]
- 22.Olsen JV, Blagoev B, Gnad F, Macek B, Kumar C, Mortensen P, Mann M. Global, in vivo, and site-specific phosphorylation dynamics in signaling networks. Cell. 2006;127(3):635–48. doi: 10.1016/j.cell.2006.09.026. [DOI] [PubMed] [Google Scholar]
- 23.Oda Y, Huang K, Cross FR, Cowburn D, Chait BT. Accurate quantitation of protein expression and site-specific phosphorylation. Proc Natl Acad Sci U S A. 1999;96(12):6591–6. doi: 10.1073/pnas.96.12.6591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Conrads TP, Alving K, Veenstra TD, Belov ME, Anderson GA, Anderson DJ, Lipton MS, Pasa-Tolic L, Udseth HR, Chrisler WB, Thrall BD, Smith RD. Quantitative analysis of bacterial and mammalian proteomes using a combination of cysteine affinity tags and 15N-metabolic labeling. Anal Chem. 2001;73(9):2132–9. doi: 10.1021/ac001487x. [DOI] [PubMed] [Google Scholar]
- 25.MacCoss MJ, Wu CC, Liu H, Sadygov R, Yates JR., 3rd A correlation algorithm for the automated quantitative analysis of shotgun proteomics data. Anal Chem. 2003;75(24):6912–21. doi: 10.1021/ac034790h. [DOI] [PubMed] [Google Scholar]
- 26.Gruhler A, Olsen JV, Mohammed S, Mortensen P, Faergeman NJ, Mann M, Jensen ON. Quantitative phosphoproteomics applied to the yeast pheromone signaling pathway. Mol Cell Proteomics. 2005;4(3):310–27. doi: 10.1074/mcp.M400219-MCP200. [DOI] [PubMed] [Google Scholar]
- 27.Goodlett DR, Keller A, Watts JD, Newitt R, Yi EC, Purvine S, Eng JK, von Haller P, Aebersold R, Kolker E. Differential stable isotope labeling of peptides for quantitation and de novo sequence derivation. Rapid Commun Mass Spectrom. 2001;15(14):1214–21. doi: 10.1002/rcm.362. [DOI] [PubMed] [Google Scholar]
- 28.Ong SE, Blagoev B, Kratchmarova I, Kristensen DB, Steen H, Pandey A, Mann M. Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol Cell Proteomics. 2002;1(5):376–86. doi: 10.1074/mcp.m200025-mcp200. [DOI] [PubMed] [Google Scholar]
- 29.Gygi SP, Rist B, Gerber SA, Turecek F, Gelb MH, Aebersold R. Quantitative analysis of complex protein mixtures using isotope-coded affinity tags. Nat Biotechnol. 1999;17(10):994–9. doi: 10.1038/13690. [DOI] [PubMed] [Google Scholar]
- 30.Krijgsveld J, Ketting RF, Mahmoudi T, Johansen J, Artal-Sanz M, Verrijzer CP, Plasterk RH, Heck AJ. Metabolic labeling of C. elegans and D. melanogaster for quantitative proteomics. Nat Biotechnol. 2003;21(8):927–31. doi: 10.1038/nbt848. [DOI] [PubMed] [Google Scholar]
- 31.Munton RP, Tweedie-Cullen R, Livingstone-Zatchej M, Weinandy F, Waidelich M, Longo D, Gehrig P, Potthast F, Rutishauser D, Gerrits B, Panse C, Schlapbach R, Mansuy IM. Qualitative and quantitative analyses of protein phosphorylation in naive and stimulated mouse synaptosomal preparations. Mol Cell Proteomics. 2007;6(2):283–93. doi: 10.1074/mcp.M600046-MCP200. [DOI] [PubMed] [Google Scholar]
- 32.Trinidad JC, Thalhammer A, Specht CG, Lynn AJ, Baker PR, Schoepfer R, Burlingame AL. Quantitative analysis of synaptic phosphorylation and protein expression. Mol Cell Proteomics. 2007 doi: 10.1074/mcp.M700170-MCP200. [DOI] [PubMed] [Google Scholar]
- 33.Wu CC, MacCoss MJ, Howell KE, Matthews DE, Yates JR., 3rd Metabolic labeling of mammalian organisms with stable isotopes for quantitative proteomic analysis. Anal Chem. 2004;76(17):4951–9. doi: 10.1021/ac049208j. [DOI] [PubMed] [Google Scholar]
- 34.McClatchy DB, Dong MQ, Wu CC, Venable JD, Yates JR., 3rd 15N metabolic labeling of mammalian tissue with slow protein turnover. J Proteome Res. 2007;6(5):2005–10. doi: 10.1021/pr060599n. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.McClatchy DB, Liao L, Park SK, Venable JD, Yates JR. Quantification of the synaptosomal proteome of the rat cerebellum during post-natal development. Genome Res. 2007;17(9):1378–88. doi: 10.1101/gr.6375007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.MacCoss MJ, Wu CC, Matthews DE, Yates JR., 3rd Measurement of the isotope enrichment of stable isotope-labeled proteins using high-resolution mass spectra of peptides. Anal Chem. 2005;77(23):7646–53. doi: 10.1021/ac0508393. [DOI] [PubMed] [Google Scholar]
- 37.Dignam JD, Lebovitz RM, Roeder RG. Accurate transcription initiation by RNA polymerase II in a soluble extract from isolated mammalian nuclei. Nucleic Acids Res. 1983;11(5):1475–89. doi: 10.1093/nar/11.5.1475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Washburn MP, Wolters D, Yates JR., 3rd Large-scale analysis of the yeast proteome by multidimensional protein identification technology. Nat Biotechnol. 2001;19(3):242–7. doi: 10.1038/85686. [DOI] [PubMed] [Google Scholar]
- 39.Xu T, Venable JD, Park SK, Cociorva D, Lu B, Liao L, Wohlschlegel J, Hewel J, Yates JRI. ProLuCID, a fast and sensitive tandem mass spectra-based protein identification program. Mol Cell Proteomics. 2006:1. [Google Scholar]
- 40.Park SK, Venable JD, Xu T, Yates JR., 3rd A quantitative analysis software tool for mass spectrometry-based proteomics. Nat Methods. 2008;5(4):319–22. doi: 10.1038/nmeth.1195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Lu B, Ruse C, Xu T, Park SK, Yates J., 3rd Automatic validation of phosphopeptide identifications from tandem mass spectra. Anal Chem. 2007;79(4):1301–10. doi: 10.1021/ac061334v. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Tabb DL, McDonald WH, Yates JR., 3rd DTASelect and Contrast: tools for assembling and comparing protein identifications from shotgun proteomics. J Proteome Res. 2002;1(1):21–6. doi: 10.1021/pr015504q. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Park SK, Venable JD, Xu T, Yates JRI. Nat Methods. 2008. A Quantitative Analysis Tool via Isotope Labeling or Differential LC-MS. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ballif BA, Villen J, Beausoleil SA, Schwartz D, Gygi SP. Phosphoproteomic analysis of the developing mouse brain. Mol Cell Proteomics. 2004;3(11):1093–101. doi: 10.1074/mcp.M400085-MCP200. [DOI] [PubMed] [Google Scholar]
- 45.Bodenmiller B, Mueller LN, Mueller M, Domon B, Aebersold R. Reproducible isolation of distinct, overlapping segments of the phosphoproteome. Nat Methods. 2007;4(3):231–7. doi: 10.1038/nmeth1005. [DOI] [PubMed] [Google Scholar]
- 46.Cantin GT, Yi W, Lu B, Park SK, Xu T, Lee JD, Yates JR., 3rd Combining Protein-Based IMAC, Peptide-Based IMAC, and MudPIT for Efficient Phosphoproteomic Analysis. J Proteome Res. 2008 doi: 10.1021/pr0705441. [DOI] [PubMed] [Google Scholar]
- 47.Beausoleil SA, Villen J, Gerber SA, Rush J, Gygi SP. A probability-based approach for high-throughput protein phosphorylation analysis and site localization. Nat Biotechnol. 2006;24(10):1285–92. doi: 10.1038/nbt1240. [DOI] [PubMed] [Google Scholar]
- 48.Schwartz D, Gygi SP. An iterative statistical approach to the identification of protein phosphorylation motifs from large-scale data sets. Nat Biotechnol. 2005;23(11):1391–8. doi: 10.1038/nbt1146. [DOI] [PubMed] [Google Scholar]
- 49.Veeranna, Amin ND, Ahn NG, Jaffe H, Winters CA, Grant P, Pant HC. Mitogen-activated protein kinases (Erk1,2) phosphorylate Lys-Ser-Pro (KSP) repeats in neurofilament proteins NF-H and NF-M. J Neurosci. 1998;18(11):4008–21. doi: 10.1523/JNEUROSCI.18-11-04008.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Muchardt C, Yaniv M. ATP-dependent chromatin remodelling: SWI/SNF and Co. are on the job. J Mol Biol. 1999;293(2):187–98. doi: 10.1006/jmbi.1999.2999. [DOI] [PubMed] [Google Scholar]
- 51.Srinivasan R, Mager GM, Ward RM, Mayer J, Svaren J. NAB2 represses transcription by interacting with the CHD4 subunit of the nucleosome remodeling and deacetylase (NuRD) complex. J Biol Chem. 2006;281(22):15129–37. doi: 10.1074/jbc.M600775200. [DOI] [PubMed] [Google Scholar]
- 52.Liu Z, Mao L, Parelkar NK, Tang Q, Samdani S, Wang JQ. Distinct expression of phosphorylated N-methyl-D-aspartate receptor NR1 subunits by projection neurons and interneurons in the striatum of normal and amphetamine-treated rats. J Comp Neurol. 2004;474(3):393–406. doi: 10.1002/cne.20136. [DOI] [PubMed] [Google Scholar]
- 53.Carpenter B, MacKay C, Alnabulsi A, MacKay M, Telfer C, Melvin WT, Murray GI. The roles of heterogeneous nuclear ribonucleoproteins in tumour development and progression. Biochim Biophys Acta. 2006;1765(2):85–100. doi: 10.1016/j.bbcan.2005.10.002. [DOI] [PubMed] [Google Scholar]
- 54.Klose RJ, Bird AP. Genomic DNA methylation: the mark and its mediators. Trends Biochem Sci. 2006;31(2):89–97. doi: 10.1016/j.tibs.2005.12.008. [DOI] [PubMed] [Google Scholar]
- 55.Lewis JD, Meehan RR, Henzel WJ, Maurer-Fogy I, Jeppesen P, Klein F, Bird A. Purification, sequence, and cellular localization of a novel chromosomal protein that binds to methylated DNA. Cell. 1992;69(6):905–14. doi: 10.1016/0092-8674(92)90610-o. [DOI] [PubMed] [Google Scholar]
- 56.Zhou Z, Hong EJ, Cohen S, Zhao WN, Ho HY, Schmidt L, Chen WG, Lin Y, Savner E, Griffith EC, Hu L, Steen JA, Weitz CJ, Greenberg ME. Brain-specific phosphorylation of MeCP2 regulates activity-dependent Bdnf transcription, dendritic growth, and spine maturation. Neuron. 2006;52(2):255–69. doi: 10.1016/j.neuron.2006.09.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Seo S, Richardson GA, Kroll KL. The SWI/SNF chromatin remodeling protein Brg1 is required for vertebrate neurogenesis and mediates transactivation of Ngn and NeuroD. Development. 2005;132(1):105–15. doi: 10.1242/dev.01548. [DOI] [PubMed] [Google Scholar]
- 58.Elias JE, Gygi SP. Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry. Nat Methods. 2007;4(3):207–14. doi: 10.1038/nmeth1019. [DOI] [PubMed] [Google Scholar]
- 59.Medzihradszky KF, Darula Z, Perlson E, Fainzilber M, Chalkley RJ, Ball H, Greenbaum D, Bogyo M, Tyson DR, Bradshaw RA, Burlingame AL. O-sulfonation of serine and threonine: mass spectrometric detection and characterization of a new posttranslational modification in diverse proteins throughout the eukaryotes. Mol Cell Proteomics. 2004;3(5):429–40. doi: 10.1074/mcp.M300140-MCP200. [DOI] [PubMed] [Google Scholar]
- 60.Venable JD, Wohlschlegel J, McClatchy DB, Park SK, Yates JR., 3rd Relative quantification of stable isotope labeled peptides using a linear ion trap-Orbitrap hybrid mass spectrometer. Anal Chem. 2007;79(8):3056–64. doi: 10.1021/ac062054i. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Yu LR, Zhu Z, Chan KC, Issaq HJ, Dimitrov DS, Veenstra TD. Improved titanium dioxide enrichment of phosphopeptides from HeLa cells and high confident phosphopeptide identification by cross-validation of MS/MS and MS/MS/MS spectra. J Proteome Res. 2007;6(11):4150–62. doi: 10.1021/pr070152u. [DOI] [PubMed] [Google Scholar]
- 62.Erecinska M, Cherian S, Silver IA. Energy metabolism in mammalian brain during development. Prog Neurobiol. 2004;73(6):397–445. doi: 10.1016/j.pneurobio.2004.06.003. [DOI] [PubMed] [Google Scholar]
- 63.Evans GJ, Morgan A. Phosphorylation of cysteine string protein in the brain: developmental, regional and synaptic specificity. Eur J Neurosci. 2005;21(10):2671–80. doi: 10.1111/j.1460-9568.2005.04118.x. [DOI] [PubMed] [Google Scholar]
- 64.Kataoka M, Kuwahara R, Matsuo R, Sekiguchi M, Inokuchi K, Takahashi M. Development- and activity-dependent regulation of SNAP-25 phosphorylation in rat brain. Neurosci Lett. 2006;407(3):258–62. doi: 10.1016/j.neulet.2006.08.055. [DOI] [PubMed] [Google Scholar]
- 65.Putignano E, Lonetti G, Cancedda L, Ratto G, Costa M, Maffei L, Pizzorusso T. Developmental downregulation of histone posttranslational modifications regulates visual cortical plasticity. Neuron. 2007;53(5):747–59. doi: 10.1016/j.neuron.2007.02.007. [DOI] [PubMed] [Google Scholar]
- 66.Kaplan DR, Miller FD. Neurotrophin signal transduction in the nervous system. Curr Opin Neurobiol. 2000;10(3):381–91. doi: 10.1016/s0959-4388(00)00092-1. [DOI] [PubMed] [Google Scholar]
- 67.Malaterre J, Ramsay RG, Mantamadiotis T. Wnt-Frizzled signalling and the many paths to neural development and adult brain homeostasis. Front Biosci. 2007;12:492–506. doi: 10.2741/2077. [DOI] [PubMed] [Google Scholar]
- 68.Pitts AF, Miller MW. Expression of nerve growth factor, brain-derived neurotrophic factor, and neurotrophin-3 in the somatosensory cortex of the mature rat: coexpression with high-affinity neurotrophin receptors. J Comp Neurol. 2000;418(3):241–54. doi: 10.1002/(sici)1096-9861(20000313)418:3<241::aid-cne1>3.0.co;2-m. [DOI] [PubMed] [Google Scholar]
- 69.Khavari PA, Peterson CL, Tamkun JW, Mendel DB, Crabtree GR. BRG1 contains a conserved domain of the SWI2/SNF2 family necessary for normal mitotic growth and transcription. Nature. 1993;366(6451):170–4. doi: 10.1038/366170a0. [DOI] [PubMed] [Google Scholar]
- 70.Muchardt C, Reyes JC, Bourachot B, Leguoy E, Yaniv M. The hbrm and BRG-1 proteins, components of the human SNF/SWI complex, are phosphorylated and excluded from the condensed chromosomes during mitosis. EMBO J. 1996;15(13):3394–402. [PMC free article] [PubMed] [Google Scholar]
- 71.Hsieh J, Gage FH. Chromatin remodeling in neural development and plasticity. Curr Opin Cell Biol. 2005;17(6):664–71. doi: 10.1016/j.ceb.2005.09.002. [DOI] [PubMed] [Google Scholar]
- 72.Lessard J, Wu JI, Ranish JA, Wan M, Winslow MM, Staahl BT, Wu H, Aebersold R, Graef IA, Crabtree GR. An essential switch in subunit composition of a chromatin remodeling complex during neural development. Neuron. 2007;55(2):201–15. doi: 10.1016/j.neuron.2007.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Wu JI, Lessard J, Olave IA, Qiu Z, Ghosh A, Graef IA, Crabtree GR. Regulation of dendritic development by neuron-specific chromatin remodeling complexes. Neuron. 2007;56(1):94–108. doi: 10.1016/j.neuron.2007.08.021. [DOI] [PubMed] [Google Scholar]
- 74.Hagberg B, Aicardi J, Dias K, Ramos O. A progressive syndrome of autism, dementia, ataxia, and loss of purposeful hand use in girls: Rett’s syndrome: report of 35 cases. Ann Neurol. 1983;14(4):471–9. doi: 10.1002/ana.410140412. [DOI] [PubMed] [Google Scholar]
- 75.Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U, Zoghbi HY. Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat Genet. 1999;23(2):185–8. doi: 10.1038/13810. [DOI] [PubMed] [Google Scholar]
- 76.Arnold SE, Talbot K, Hahn CG. Neurodevelopment, neuroplasticity, and new genes for schizophrenia. Prog Brain Res. 2005;147:319–45. doi: 10.1016/S0079-6123(04)47023-X. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
There are four supplemental figures and five supplemental tables in this manuscript. Figure S1 shows the total number of phosphopeptides in each of the accumulating MudPIT runs. Figure S2 and S3 list the sequence motif of phosphopeptides from p1 and p45 cortical nuclear proteome. Figure S4 shows the annotated MS2 spectra of the phosphopeptides quantified and illustrated in Figure 6. Table S1 lists Ratios of phosphopeptides identified by MS2 from p1 or p45 cortex over a common internal standard (15N labeled rat brain). Table S2 lists ratios of phosphopeptides identified by MS3 from p1 or p45 cortex over a common internal standard (15N labeled rat brain). Table S3 lists phosphopeptides with large differences in phosphorylation ratios (singleton peptides). Table S4 lists phosphoprotein and total protein ratios between p1 and p45 cortex. Table S5 lists the ratios of total proteins from p1 or p45 cortex over a common internal standard (15N labeled rat brain).
Supporting Information Softwares ProLucid, Census and Debunker are available for download via the following website: http://fields.scripps.edu/download.php.
The MS2 and MS3 spectra of the reported phosphopeptides are available at the author’s website: http://fields.scripps.edu/yateslab/public/08_jpr_liao/
Supporting figures and tables are available free of charge via the Internet at http://pubs.acs.org
Ratios of phosphopeptides identified by MS2 from p1 or p45 cortex over a common internal standard (15N labeled rat brain). Column A lists peptide sequences with phosphorylation site indicated as (p). Column B and C list the protein index and name, respectively, from which the peptide sequence was derived. Column D lists the ratio of each phosphopeptide derived from p1 cortex over the same peptide derived from 15N brain. Column E, same as Column D except that the ratio was p45 cortex over 15N brain. Column F and G list the probability value (generated by Debunker) of each peptide listed in Column A, identified in p1 and p45, respectively. Column H lists the charge state of each peptide listed in Column A.
Ratios of phosphopeptides identified by MS3 from p1 or p45 cortex over a common internal standard (15N labeled rat brain). Same as Table S1 except that the phosphopeptides were identified by MS3.
List of phosphopeptides with large differences in phosphorylation ratios (singleton peptides). Column A lists peptide sequences with phosphorylation site indicated as (p). Column B and C list the protein index and name, respectively, from which the peptide sequence was derived. Column D lists the ratio of each phosphopeptide derived from p1 cortex over the same peptide derived from 15N brain. Column E, same as Column D except that the ratio was p45 cortex over 15N brain. Column F and G list the ratio of each protein from p1 or p45 cortex respectively over the protein from 15N brain. Column H and I, probability value (generated by Debunker) of each peptide listed in Column A, identified in p1 and p45, respectively. Column J, charge state of the phosphopeptide in Column A.
Phosphoprotein and total protein ratios between p1 and p45 cortex. Column A and B, protein index and name respectively. Column C, annotation of cellular localization for each protein.Column D and E, relative ratio between p1 and p45 cortex of phosphoproteins (D) and total proteins (E). Column F and G, labels of relative expression ratios of phosphoproteins (F) and total proteins (G) between p1 and p45, whereby, -1 indicates a ratio of <0.5, 0 indicates a ratio of between 0.5 and 2.0, and 1 indicates a ratio of >2.0. Column H list the cluster number of the proteins based on their relative expression ratio changes in phosphoprotein as well as total protein. Column I gives the color code used to plot the correlation graph in Figure 4A.
Ratios of total proteins from p1 or p45 cortex over a common internal standard (15N labeled rat brain). Column A and B, protein index and name respectively. Column C, the ratio of each protein derived from p1 cortex over the same protein derived from 15N brain. Column D, standard deviation (S.D.) of multiple peptide ratios used to derive the protein ratio. In cases where one peptide was used to identify the protein, S.D. was not available (n/a). Column E and F, same layout as Column C and D except that the protein ratios were from p45 cortex. Note that some proteins were quantified in either p1 or p45 cortex but not in both, in such cases the sample with the proteins not quantified labeled as “n/a”.
Total number of phosphopeptides increase as the function of the increasing number of MudPIT runs.
Sequence motif logo of phosphopeptides from p1 and p45 cortical nuclear proteome.
Sequence motif logo of phosphopeptides from p1 and p45 cortical nuclear proteome.
Annotated MS2 spectra of the phosphopeptides quantified and illustrated in Figure 6.




