Skip to main content
Philosophical transactions. Series A, Mathematical, physical, and engineering sciences logoLink to Philosophical transactions. Series A, Mathematical, physical, and engineering sciences
. 2016 Oct 28;374(2079):20150364. doi: 10.1098/rsta.2015.0364

Stable isotope dimethyl labelling for quantitative proteomics and beyond

Jue-Liang Hsu 1,, Shu-Hui Chen 2,
PMCID: PMC5031631  PMID: 27644970

Abstract

Stable-isotope reductive dimethylation, a cost-effective, simple, robust, reliable and easy-to- multiplex labelling method, is widely applied to quantitative proteomics using liquid chromatography-mass spectrometry. This review focuses on biological applications of stable-isotope dimethyl labelling for a large-scale comparative analysis of protein expression and post-translational modifications based on its unique properties of the labelling chemistry. Some other applications of the labelling method for sample preparation and mass spectrometry-based protein identification and characterization are also summarized.

This article is part of the themed issue ‘Quantitative mass spectrometry’.

Keywords: stable-isotope labelling, dimethyl labelling, quantitative proteomics, LC-MS/MS, post-translational modifications

1. Introduction

Comparative or quantitative analysis of protein expression in two biological states (e.g. normal versus disease or treated versus non-treated states) is a key focus of proteomics studies [1,2], which aim to discover new biomarkers or gain mechanistic understandings [3]. Two-dimensional gel electrophoresis [4], separating hundreds to thousands of proteins present in a highly complicated biological sample on a single gel, is traditionally used for protein profiling. However, time-consuming procedures such as spot-by-spot in-gel digestion followed by liquid chromatography-mass spectrometry (LC-MS) analysis and the lack of sensitivity for low-abundant proteins have limited its use in large-scale proteomics study [5].

Alternatively, as shown in figure 1, stable-isotope labelling coupled with LC-MS analysis is able to simultaneously provide quantitative and qualitative (protein identification) information in a single run and has become a popular method for large-scale analysis of protein expression [1]. The labelling methods can be divided into three categories, namely metabolic labelling [6,7], enzymatic labelling [8] and chemical labelling [911]. Isotopic elements are incorporated into the sample at the protein level (metabolic and chemical labelling) or at the peptide level during (enzymatic labelling) or after (chemical labelling) enzymatic digestion. Using shotgun proteomics approach, the digested peptides derived from two compared samples are combined and separated by LC or two-dimensional liquid chromatography (2D-LC), and then analysed by MS and tandem mass spectrometry (MS/MS) (figure 1). Proteins are identified based on peptide sequencing (MS/MS) via database search. Using the same set of data, the same proteins derived from two biological states are quantified based on the relative peak area of extracted ion chromatograms (XICs) of the precursor ion pair (MS) or based on the relative intensity of the tandem mass tag (MS/MS) of the isobaric ion pair. As summarized in table 1, metabolic labelling is referred to as stable-isotope labelling by amino acids in cell culture (SILAC) and enzymatic labelling is mainly referred to as H2O/H218O labelling.

Figure 1.

Figure 1.

Stable-isotope labelling methods for MS-based quantitative proteomics. (Online version in colour.)

Table 1.

Stable-isotope labelling methods for MS-based quantitative proteomics.

method labelling site ΔMw (isotopic pair) labelling performancea MS or MS/MS (quantification) sample type duplex/multiplex cost refs
metabolic labelling
 SILAC Lys and Arg 6 Da +++ MS cell culture duplex or triplex $$$ [6,7]
enzymatic labelling
 H2O/H218O C-termini 4 Da ++ MS all duplex $$ [8]
chemical labelling
 ICAT Cys 8 Da +++ MS all duplex $$$ [9]
 photocleavable ICAT Cys 7 Da +++ MS all duplex $$$ [10]
 cICAT Cys 9 Da +++ MS all duplex $$$ [11]
 methanol-d3 Asp, Glu, C-termini 3 Da + MS all duplex $ [12]
 acetylation N-termini and Lys 3 Da +++ MS all duplex $ [13]
 2-methoxy-4,5-dihydro-1H-imidazole Lys 4 Da + MS all duplex $$ [14]
 dimethyl labelling N-termini and Lys 4 Da +++ MS all duplex, triplex, four-plex or five-plex $ [1518]
 tandem mass tag N-termini and Lys isobaric ++ MS/MS all duplex $$$ [19]
 iTRAQ N-termini and Lys isobaric +++ MS/MS all four-plex or eight-plex $$$ [20,21]

aThe labelling performance is evaluated by the reaction rate and specificity of each labelling reagent. ‘+++’, ‘++’ and ‘+’ represent very good, good and fair, respectively.

For chemical labelling, dozens of stable-isotope reagents (table 1) mainly targeting amine or carboxylic acid groups have been reported [1221] and reviewed recently [2231]. Each chemical labelling method has advantages and disadvantages. For example, isotope-coded affinity tag (ICAT) was designed to incorporate selective affinity enrichment for cysteine residues but non-cysteine containing peptides were not labelled. Isobaric tags for relative and absolute quantitation (iTRAQ) are able to compare multiple samples (multiplex) up to eight-plex [20,21]. However, reactive N-hydroxysuccinimide (NHS) ester reagents contained in iTRAQ kits are not stable for long-term storage. In contrast with ICAT and iTRAQ, which are commercial kits, stable-isotope dimethyl labelling by reductive amination [1518] uses commercial reagents and is superior to others in high reaction yield, high accuracy and high reproducibility, as well as robustness, simplicity and low cost [32,33]. It was claimed that stable-isotope dimethyl labelling, SILAC and iTRAQ are three benchmarking labelling methods for quantitative proteomics [32]. In addition to a general review of quantitative proteomics using stable-isotope dimethyl labelling, the use of unique properties of dimethyl labelling for sample preparation and for MS-based protein characterization is also included in this review.

2. Labelling chemistry for liquid chromatography-mass spectrometry based quantitative proteomics

Reductive amination is a well-known organic reaction commonly used in protein chemistry [34,35]. As depicted in figure 2a, formaldehyde forms Schiff base with the N-terminus or ϵ-amino group of Lys residue of a peptide/protein; the aldimine intermediate is then reduced by sodium cyanoborohydride (NaBH3CN) to form a secondary amine which is more nucleophilic than the primary amine and immediately reacts with another formaldehyde molecule to form dimethylamino group. Reductive amination is a global labelling method for all peptides with one labelling site for peptides without Lys residue or 1 + N labelling sites for peptides with N Lys residues. All amino groups are dimethylated except the N-termini of Pro residue, which is a secondary amino group and is monomethylated by the reaction. It is particularly notable that dimethylated peptides exhibit remarkable a1 ion enhancement by collision induced dissociation (CID) (figure 2b), possibly via CO neutral loss from the b1 ion [36]. The strong a1 fragment ion is very useful for MS-based protein characterization and will be discussed later. Because formaldehyde (CH2O) is small and water-soluble, the labelling reaction is quick, specific and complete.

Figure 2.

Figure 2.

Reaction mechanism of (a) reductive dimethylation (dimethyl labelling) and (b) a1 ion formation of dimethylated peptides by CID.

Stable-isotope dimethyl labelling using reductive amination for MS-based quantitative proteomics was first reported in 2003 [15]. It is compatible with most chromatographic methods for peptide separation using reversed phase (RP) [15], strong cation exchange (SCX) [37], hydrophilic interaction chromatography (HILIC) [38], immobilized metal ion affinity chromatography (IMAC) [39] column or with multi-dimensional separation by combining two or three separation methods [37]. Because the number of positive charges borne by a peptide is normally increased by dimethylation, fragmentation efficiency of CID or electron transfer dissociation is likely to be enhanced [40]. This increases the success rate of protein identification using database search based on MS/MS spectra. There were some concerns when the method was first reported: (i) shift of the retention time caused by isotopic (deuterium) effect [41], (ii) overlapping of isotopic clusters due to the relatively small mass difference, and (iii) lack of quantification software [15,42]. The first problem was solved by using XICs for quantitative analysis instead of ion intensities [15,4345]. The second problem was simulated by MS-Isotope (embedded in the Protein-Prospector) software and found to be negligible in affecting accuracy when compared with the ratio calculated from ‘pure’ monoisotopic peaks [44] except for extremely large peptides (more than 3 kDa) bearing single labelling site [46]. The third concern is not a problem now because many available softwares such as Proteome Discoverer (Thermo Fisher Scientific), Mascot Distiller (Matrix Science), XPRESS [47], MaxQuant [48], PVIEW [49] and MSQuant [50] can be applied for any kinds of labelling methods including dimethyl labelling.

Because dimethyl labelling is relatively fast, specific and mild, it is perfectly suitable for online or on-column protocols via in situ labelling to improve the throughput and automation. Heck and co-workers standardized three types of protocols for stable-isotope dimethyl labelling: in-solution, online and on-column using solid-phase extraction (SPE) to meet the requirements of different sample amounts [18,43]. Using online labelling, a fully automated analysis method can be developed including the following procedures: (1) the peptide sample was loaded onto a trap column connected by a six-port valve; (2) the peptides retained on the column were chemically labelled by flushing the trapping column with the labelling reagents; (3) excess reagents were washed away by online desalting; (4) the second sample underwent steps (1), (2) and (3) sequentially; (5) the valve was switched to the analytical column connected to MS for a regular LC-MS analysis. The new protocols simplified several time-consuming handling steps including removal of amine-containing ingredients (e.g. NH4HCO3), addition of the labelling regents, incubation and quenching for the reaction, desalting and concentration of the product, leading to less sample loss and better reproducibility [51,52]. Raijmakers et al. applied this on-column labelling protocol to determine the differences in composition between bovine liver and spleen 20S core proteasome complexes [52]. Zou et al. integrated this on-column sequential labelling protocol with a RP-SCX biphasic column for multi-dimensional separation and high-throughput quantitative proteome analysis [51].

Although dimethyl labelling is commonly used for comparative quantification of two samples, it can be extended for multiplex analysis based on MS spectra. Different isotopic forms of formaldehyde such as CD2O or 13CH2O and sodium cyanoborohydride such as NaBD3CN or NaBH3CN are commercially available at low cost. As summarized in table 2, multiplex labelling can be easily achieved by different combinations of reagents to generate a panel of mass differences and used to compare multiple samples acquired from different time points or different dosages [20]. For example, four-plex analysis by dimethyl labelling can be achieved by using reagent combinations of CH2O/NaBH3CN, CH2O/NaBD3CN, CD2O/NaBH3CN and CH2O/NaBD3CN [16]. The multiplex can be further extended to five-plex using an additional isotopic reagent combination like 13CD2O/NaBD3CN [17]. These combinations were coupled with Lys-C enzyme to generate at least two labelling sites (N-termini and Lys) for each peptide, ensuring at least 4 Da of the mass shift between the nearest labelled forms in order to minimize overlapping of neighbouring isotopic clusters, increasing the accuracy of quantification. However, peptides generated by Lys-C are relatively long and may not be suitable for conventional MS/MS-based protein identification strategy. Therefore, other combinations like CH2O/NaBH3CN, CD2O/NaBH3CN and 13CD2O/NaBD3CN were used to enlarge the mass shift of the neighbouring isotopic peaks [18]. Tandem mass tags may also be generated by stable-isotope dimethyl labelling using mass defect-based pseudo-isobaric dimethyl labelling (pIDL) reagents, CD2O/NaBD3CN and 13CD2O/NaBH3CN, coupled with LysC digestion [53]. The precursor ion pair generated by the pIDL method have an almost identical mass (isobaric tag); the quantification ratios were calculated based on the intensity of a1 fragment ion pair which exhibit a small mass difference (0.00584 Da) resolved by high-resolution MS/MS [53].

Table 2.

Combinations of stable-isotope reagents for multiplex dimethyl labelling.

reagent
method CH2O + NaBH3CN + 28 Da per site CH2O + NaBD3CN + 30 Da per site CD2O + NaBH3CN + 32 Da per site CD2O + NaBD3CN + 34 Da per site 13CD2O + NaBD3CN + 36 Da per site 13CD2O + NaBH3CN + 34 Da per site enzyme refs
duplex trypsin [15]
triplex trypsin [18]
four-plex Lys-C [16]
five-plex Lys-C [17]
pIDL Lys-C [53]

3. Applications for quantitative proteomics

Stable-isotope dimethyl labelling has been widely used in quantitative proteomics since the method was first reported in 2003 [54]. In 2012, Heck et al. published a review on applications of stable-isotope dimethyl labelling mainly for global (or large-scale) expression profiling, quantitative analyses of post-translational modifications (PTMs) and protein interaction. In this review, we briefly update applications either not included in or reported after Heck's review (2012 until now).

(a). Global protein-expression profiling

Like most proteomics studies, many early applications of stable-isotope dimethyl labelling focused on finding biomarkers of diseases or key proteins involved in pathways of cell models [5559]. D'Aguanno et al. used stable-isotope dimethyl labelling to investigate the role p63 isoforms play in cancer stem cells (CSCs) [60]. Colon CSCs derived from primary tumour were transduced with lentiviral vectors carrying either p63 containing (TA) or lacking (ΔN) gene. The light and heavy labelled tryptic peptides from TAp63, ΔNp63, and control colon CSC cells were combined and separated using immobilized pH gradient isoelectric focusing followed by RP chromatographic separation and the separated peptides were analysed by LC-MS/MS. Proteins were identified and quantified using Mascot Distiller software. Differentially expressed proteins were functionally classified and annotated by bioinformatics tools. Their data showed that most modulated proteins were involved in metabolic processes and these proteins were confirmed by western blotting and targeted label-free quantitative analysis [60]. Swa et al. used stable-isotope dimethyl labelling coupled with integrative protein–protein interaction network analysis to reveal the role that annexin-1 may play in the process of developing mammary tumourigenesis [61]. Sato et al. applied triplex dimethyl labelling using CH2O/NaBH3CN (light), CD2O/NaBH3CN (medium) and 13CD2O/NaBD3CN (heavy) reagents to samples derived from the pooled, control and treated tissues, respectively. The combined samples were analysed by nanoLC-high-resolution LTQ Orbitrap Velos mass spectrometry. Among 6694 identified proteins, proteins belonging to the eukaryotic initiation factor (eIF) families were upregulated in the control compared with those in inhibitor-treated tissues which exhibited lung metastases. The data implied that eIF family members, especially eIF4A1 and eEF2, are highly correlated to the metastatic phenotype of advanced breast cancer [62]. Chiang et al. used duplex dimethyl labelling coupled with nanoLC-MS/MS to comparatively analyse urine proteins from patients with and without urothelial carcinoma [63]. Among 219 candidate proteins identified, SH3 domain binding glutamic acid-rich protein like 3 (SH3BGRL3) was identified to overexpress in patient urines. This finding positively correlated with the higher histological grading and muscle invasiveness of urothelial carcinoma, indicating SH3BGRL3 may be a potential biomarker for bladder cancer [63].

In addition to biomarker discovery and biomedical research, quantitative proteomics using stable-isotope dimethyl labelling was also applied to other fields such as food science and energy. Ho et al. used stable-isotope dimethyl labelling to quantitatively analyse signature tryptic peptide (SFMFGGLASGETR) derived from a marker protein, herbicide-resistantgene-related protein 5-enolpyruvylshikimate-3-phosphate synthase (CP4EPSPS), derived from genetically modified soya bean [64]. To enrich CP4EPSPS from other high abundant proteins such as glycinin and β-conglycinin, strong anion exchange and SDS-PAGE were used for sample preparation. The combined dimethylated tryptic peptides of CP4EPSPS were identified and quantified using matrix assisted laser desorption and ionization time of flight MS and the data showed excellent linear correlation with the content of genetic modification in soya bean samples [64]. Tolonen et al. applied duplex dimethyl labelling for quantitative proteomics of Clostridium phytofermentans in the medium of cellulosic substrates versus glucose. Their results indicated that secreted carbohydratases accompanied with glycolytic enzymes and alcohol dehydrogenases were upregulated upon the treatment of cellulosic substrates, promoting hemicellulose or cellulose degradation and ethanol production. They concluded that quantitative proteomics studies can provide valuable information to improve biomass fermentation for industrial applications [65].

(b). Quantitative analysis of protein post-translational modifications

Protein PTMs play crucial roles in modulating biological functions and more than 400 PTMs have been reported so far. However, identification and quantification of site-specific PTMs by MS are still challenging. Enrichment steps are normally required due to low abundance of the modified proteins and ion suppression effect inherent with electrospray ionization [66]. Many targeted enrichment methods, mainly for phospho/glyco peptides, have been developed [6769] but relatively large amounts of starting samples are still required for identifying low-abundant PTMs. Owing to the low cost, stable-isotope dimethyl labelling has been a better choice than other expensive labelling kits for these studies [54]. In table 3, we summarize applications of stable-isotope dimethyl labelling for quantitative analysis of phosphoproteomics and glycoproteomics in terms of sample type and amount of total protein used, labelling approach, enrichment method, quantification software and efficiency. These applications are briefly discussed in the following.

Table 3.

Applications of stable-isotope dimethyl labelling for quantitative analysis of protein phosphorylation and glycosylation.

sample multiplex enrichment method quantification software no. modified protein/ peptide identified (total protein amount used) refs
zebra fish embryos duplex SCX, online RP-TiO2 MSQuant 348 phosphoproteins (100 µg) [70]
phosphoproteomics
 MCF cells duplex IMAC-HILIC Mascot distiller 2857 unique phosphorylation sites (1 mg) [39]
 HeLa cells triplex immunoprecipitation (PY99-agarose beads) MSQuant 1100 unique phosphopeptides (4 mg) [71]
 membrane fraction of hESCs and NSCs duplex TiSH: TiO2-SIMAC-HILIC LOESS or DanteR package 10 087 phosphopeptides (200 µg) [72]
 porcine muscle triplex TiSH Proteome Discoverer 784 unique phosphopeptides (200 µg) [73]
 INS-1 cells duplex (CH2O/NaBH3CN and 13CH2O/NaBD3CN) TisH Proteome Discoverer 6600 phosphopeptides (300 µg) [74]
 human liver tissues duplex in situ sample processing approach, Ti4+-IMAC MaxQuant 8548 unique phosphopeptides (0.5 mg) [75]
 liver tissues from C57BL/6 mice duplex hydroxy acid-modified metal oxide chromatography (HAMMMOC) Mass Navigator 1090 unique phosphopeptides (25 µg) [76]
 heart tissues from C57BL/6 mice triplex SCX Proteome Discoverer 525 unique phosphopeptides [77]
 yeast mitochondria Triplex IMAC (Phos-Select Iron Affinity Gel) MassChroQ 670 unique phosphopeptides (1.3 mg) [78]
glycoproteomics
 plasma from C57/Bl6 mice duplex lectin affinity chromatography XPRESS 708 glycoproteins were identified (40 µl of plasma) [79]
 serum samples from patients of liver cirrhosis and HCC patients duplex hydrazide capture MSQuant 1179 glycopeptides were identified (100 µl of serum) [80]
 membrane protein fraction of rat myocardial tissue duplex TiO2 (for sialic acid-containing glycopeptides) + ZIC-HILIC (for neutral glycopeptides) MSQuant and StatQuant 590 non-redundant glycosylation sites were identified and quantified [81]

Quantitative phosphoproteomics is one of the hot topics in PTM studies and is also the most common applications of stable-isotope dimethyl labelling for PTM analysis. Common phosphopeptide enrichment methods include immobilized antibody [71], Fe2+-IMAC [82], TiO2 [70] and SCX [77]. In addition, Wakabayashi et al. developed hydroxy acid-modified metal oxide chromatography (HAMMMOC) to enhance the specificity of TiO2 [76]. Impressively, HAMMMOC method was able to identify 1090 unique phosphopeptides with as little as 25 µg of total protein contained in the sample. Larsen et al. developed a so-called TiSH three-stage purification approach, in which phosphopeptides were pre-fractionated using TiO2, followed by sequential elution from IMAC (SIMAC) to separate multi- and mono-phosphorylated peptides and the mono-phosphorylated peptides were further fractionated by HILIC [7274]. Phosphopeptide enrichment using single step enrichment may not provide sufficient specificity and many studies have applied multi-dimensional separation and enrichment for phosphopeptides.

Huang et al. reported the first application of dimethyl labelling for quantitative phosphoproteomics in which duplex dimethyl labelling combined with IMAC was used to analyse protein phosphorylation induced by 8-bromo-cGMP in pregnant rat uteri [82]. Lemeer et al. used SCX to fractionate dimethylated peptides followed by online affinity enrichment using TiO2 and LC-MS/MS analysis for comparative study of protein phosphorylation between Fyn/Yes knockdown zebrafish embryos and the wild-type [70]. Wu et al. combined IMAC and HILIC for the study of quantitative phosphoproteomics involved in estrogen-induced transcriptional regulation in MCF-7 breast cancer cells [39]. In their studies, a total of 2857 unique phosphorylation sites contained in 1338 phosphoproteins were identified and quantified. Boersema et al. reported an efficient quantitative profiling method using triplex dimethyl labelling coupled with peptide-level immunoprecipitation for comparative analysis of low-abundant tyrosine phosphorylated proteins in HeLa cells upon treatment with pervanadate or epidermal growth factor [71]. Song et al. compared conventional duplex with triplex dimethyl labelling and they found that the triplex labelling increased the number of quantified phosphopeptides by 50% and reduced the processing time by 50% [83]. We note that stable-isotope dimethyl labelling is able to couple with almost any phosphopeptide enrichment methods, yielding excellent quantitative results.

Protein glycosylation is another crucial PTM that controls many biological processes such as cell growth, cell migration, cell adhesiveness, immune response and tumour cell proliferation [84,85]. Stable-isotope dimethyl labelling has also been widely employed in quantitative glycoproteomics [7981,86]. As summarized in table 3, common glycopeptide enrichment methods include lectin affinity chromatography [79], hydrazide capture chemistry [80], and TiO2 combined with zwitterionic HILIC (ZIC-HILIC) [81,86]. Unlike specific recognition between lectin and carbohydrate epitopes, hydrazide capture method is able to catch all types of glycosylated protein/peptide and is becoming a popular method for quantitative glycoproteomics.

Her et al. demonstrated a comparative strategy to simultaneously analyse glycosylated and non-glycosylated proteins in which dimethylated glycopeptides were separated from the non-glycosylated peptides using microcrystalline cellulose. The relative abundances of the dimethylated peptide pairs, both non-glycosylated and glycosylated, were directly analysed using a LTQ linear ion trap mass spectrometer [87]. Zhang et al. developed an online method for quantitative N-glycoproteome analysis [88] by combining a HILIC column for glycopeptide enrichment, a hydrophilic PNGase F immobilized enzymatic reactor for deglycosylation, and a C18 trap column for dimethyl labelling. Zou et al. integrated glycopeptide enrichment using hydrazide capture and stable-isotope dimethyl labelling on beads in sequence to comparatively quantify N-linked glycoproteins in human serum of healthy volunteers (n = 16) and liver cancer patients (n = 11) [89]. Although hydrazide capture provides high specificity (more than 90%) for glycopeptide enrichment via aldehyde group exposed by oxidative cleavage of cis-vicinal diols on carbohydrates, oxidation of the N-terminal Ser/Thr residue of a peptide may concomitantly occur. This will lead to undesired covalent binding of glycopeptides to hydrazide beads via the oxidized Ser/Thr N-termini and hinder the release of N-terminal Ser/Thr-containing glycopeptides by PNGase F [90]. As depicted in figure 3, prior dimethyl labelling can block N-termini of the Ser/Thr residue to prevent oxidation (aldehyde formation), minimizing the sample loss due to undesired covalent bonding to the beads [91]. Xia et al. applied the revised protocol for comparative glycoproteomics study of triple-negative breast cancer (TNBC) tissues [92]. They identified 550 unique N-linked glycoproteins, among which 56 were upregulated and 16 were downregulated. Furthermore, several N-linked glycoproteins such as vascular endothelial growth factor receptor 1, insulin receptor and tissue factor pathway inhibitor were validated and regarded as potential biomarkers for TNBC diagnosis [92].

Figure 3.

Figure 3.

Flowchart of stable-isotope dimethyl labelling combined with hydrazide capture for quantitative glycoproteomics.

4. Applications beyond quantitative analysis

Heck et al. have previously demonstrated that dimethyl labelling is useful for de novo sequencing by combining the labelling with Lys-N enzyme to create a majority of Lys-N residue [93]. Dimethyl labelling is also useful for blocking active sites, increasing peptide basicity (or charge state), and enhancing peptide sequencing based on a1 fragment ion. As depicted in figure 4, many proteomics applications beyond quantification have been reported based on these characteristics. For example, dimethyl labelling can be used to detect protein N-termini [94] (figure 4a) generated after translocation/processing events which are crucial to understand the transformation of precursors to mature proteins. Chen et al. designed a flowchart to target protein N-termini via sequentially blocking active sites by dimethyl labelling. In their study, proteins were first dimethylated at N-termini and ϵ-amino group of Lys residue and then digested by trypsin to expose free N-termini of internal peptides. These peptides were captured by aldehyde-containing resin (POROS-AL). Peptides containing dimethylated or endogenously modified protein N-termini cannot be captured by the resin and the flow-through fractions are collected for LC-MS/MS analysis. By this manner, the N-terminal sequence or modifications (e.g. acetylation and formylation) were able to be unambiguously identified [95]. Based on the same concept, a so-called terminal amine isotopic labelling of substrates (TAILS) strategy was developed by Overall et al. using dendritic polyglycerol aldehyde polymers for internal peptide depletion [95,96]. Using TAILS strategy, 731 acetylated, 132 cyclized N-termini and 288 matrix metalloproteinase-2 cleavage sites in mouse fibroblast secretomes were identified and quantified for proteome-wide analysis of N terminome [95,96]. The authors further modified the TAILS strategy to analyse C terminome (C-TAILS) [97] in which dimethyl labelling was first used to block amino groups and then carboxylic acids including carboxyl groups of the C-termini and the side chain residue of aspartate or glutamate. The latter reaction was achieved by the combination of 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDC)/NHS and ethanolamine. After trypsin digestion, the newly generated N-terminal amino groups of internal peptides were protected using dimethyl labelling again and their free C-termini were captured by linear polyallylamine polymer via EDC-mediated condensation; the survived protein C-termini were identified and quantified using LC-MS/MS [97]. In addition to protein N-termini and C-termini, Guo et al. used isotope-coded dimethyl labelling and chemoprecipitation to identify and quantify low-abundant protein nitration [98]. In their study, the flowchart included the following steps: (i) dimethyl labelling was used to block N-terminal amine and Lys ϵ-amine of all tryptic peptides; (ii) Na2S2O4 was added to reduce nitrotyrosines in corresponding aminotyrosines; (iii) the dimethylated aminotyrosine-containing peptides were captured using solid-phase active ester reagent (SPAER) on glass beads and other peptides were washed away; and (iv) the dimethylated aminotyrosine-containing peptides were released from SPAER beads under acidic condition and analysed by LC-MS/MS [98].

Figure 4.

Figure 4.

Applications (beyond quantification) of stable-isotope dimethyl labelling for (a) identification of protein N-terminal sequence, (b) identification and quantification of Nα-acetylated peptide via SCX fractionation and (c) identification of disulfide linkages or ubiquitation tags via enhanced a1 ions. (Online version in colour.)

The second useful property of dimethyl labelling is based on the increased basicity of a peptide by the labelling [99], which can alter chromatographic behaviour during ionic separation (figure 4b). Based on this concept, Chen et al. compared the retention behaviour between dimethylated, non-modified and acetylated peptides (bearing the same peptide sequence) during SCX separation. They found that their retention order (acetylated peptide < non-modified peptide < dimethylated peptide) closely correlates to their basicities. Therefore, using dimethyl labelling and simple SCX SPE for sample preparation, the number of acetylated peptides identified from HepG2 cells was four times greater than that of direct analysis (without dimethyl labelling and SCX) and two times greater than that using merely SCX (without dimethyl labelling). Chen et al. further applied this approach (dimethyl labelling and SCX) to identify and quantify Nα-acetylated proteins in HepG2 cells with and without tert-butyl hydroperoxide (t-BHP) treatment [100]. In their study, 576 non-redundant Nα-acetylated peptides were identified from 50 µg of cytosolic proteins extracted from HepG2 cells. Notably, the whole process was completed in 24 h and Nα-acetylated peptides of some protein isoforms, such as β-actin/γ-actin, ERK1/ERK2, α-centractin/β-centractin and ADP/ATP translocase 2 and 3, were simultaneously detected from the SCX flow-through fraction [100].

The third useful property of dimethyl labelling is the enhanced a1 ion by CID fragmentation [36] as depicted in figure 2b. Hsu et al. found that a1 ion enhancement not only occurred to dimethylated peptides but also dimethylated proteins [101]. Based on multiple a1 fragments observed in a single MS/MS spectrum, Huang et al. developed a novel method to identify disulfide linkages [102]. As depicted in figure 4c (top), multiple a1 ions of a disulfide-linked precursor ion were detected by CID and they can be used to indicate the N-terminal residue of individual peptides that are linked by disulfide bonds. This approach was demonstrated using recombinant human pancreatitis-associated protein which contained three disulfide linkages formed by six cysteines. Once two or more a1 ions were matched, each peptide sequence was assigned based on matching the remaining b/y ion series in the MS/MS spectra. Home-made software named rapid assignment of disulfide linkage via a1 ion recognition (RADAR) was developed based on this algorithm [102]. Huang et al. applied RADAR to identify disulfide linkages of recombinant monoclonal antibody bevacizumab (Avastin) [103] and crude snake venom [104]. Such multiple a1 ion recognition approach was also applied to identify ubiquitinated peptides because ubiquitinated Lys normally contains a short diglycine (GG) tag. As shown in figure 4c (bottom), the main peptide skeleton and the diglycine side chain of an ubiquitinated peptide can be readily recognized by their individual a1 ion and the side chain's b2′ ion. As a proof of concept, Griffiths et al. demonstrated this approach by identifying a ubiquitinated protein, UbK48, spiked in a six-protein mixture [105]. Wu et al. reported that the a1 signal was suppressed for precursor ions with phosphorylated Ser/Thr N-termini (N-p*Ser/Thr) but no suppression was observed for the precursor ion with phosphorylated Tyr N-terminus [36]. Although somehow negative, this provides a useful hint for mapping phosphorylation sites at the N-termini [106]. Despite diverse usefulness, a1 ion is not detectable for ion trap instruments due to the ‘1/3 rule’, which also occurs for other tandem mass tags like iTRAQ. This limitation, however, can be overcome with the use of an Orbitrap instrument equipped with a higher-energy collisional dissociation chamber outside the trap.

5. Concluding remarks

Stable-isotope dimethyl labelling based on reductive dimethylation has been well accepted and widely used for quantitative proteomics analysis (460 citations up to now). It is a useful method for the ‘poor’. In addition, its usefulness is not limited to quantification; dimethyl labelling has been used to assist developing versatile enrichment or separation protocols, de novo sequencing, and characterization of protein PTMs based on MS analysis. With no doubt, we anticipate more quantitative applications and deeper explorations of its usefulness in MS-based analysis to be demonstrated in the future.

Competing interests

We declare we have no competing interests.

Funding

This work was financially supported by Taiwan MOST grants (MOST 104-2113-M-020–001) to J.-L.H.

References

  • 1.Aebersold R, Mann M. 2003. Mass spectrometry-based proteomics. Nature 422, 198–207. ( 10.1038/nature01511) [DOI] [PubMed] [Google Scholar]
  • 2.Yates JR, Washburn MP. 2013. Quantitative proteomics. Anal. Chem. 85, 8881 ( 10.1021/ac402745w) [DOI] [PubMed] [Google Scholar]
  • 3.Ezzell C. 2002. Proteins rule. Sci. Am. 286, 40–47. ( 10.1038/scientificamerican0402-40) [DOI] [PubMed] [Google Scholar]
  • 4.Görg A, Weiss W, Dunn MJ. 2004. Current two-dimensional electrophoresis technology for proteomics. Proteomics 4, 3665–3685. ( 10.1002/pmic.200401031) [DOI] [PubMed] [Google Scholar]
  • 5.Duncan MW, Aebersold R, Caprioli RM. 2010. The pros and cons of peptide-centric proteomics. Nat. Biotechnol. 28, 659–664. ( 10.1038/nbt0710-659) [DOI] [PubMed] [Google Scholar]
  • 6.Ong SE, Blagoev B, Kratchmarova I, Kristensen DB, Steen H, Pandey A, Mann M. 2002. Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol. Cell. Proteomics 1, 376–386. ( 10.1074/mcp.M200025-MCP200) [DOI] [PubMed] [Google Scholar]
  • 7.Seyfried NT, Gozal YM, Dammer EB, Xia Q, Duong DM, Cheng D, Lah JJ, Levey AI, Peng J. 2010. Multiplex SILAC analysis of a cellular TDP-43 proteinopathy model reveals protein inclusions associated with SUMOylation and diverse polyubiquitin chains. Mol. Cell. Proteomics 9, 705–718. ( 10.1074/mcp.M800390-MCP200) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Yao X, Freas A, Ramirez J, Demirev PA, Fenselau C. 2001. Proteolytic 18O labeling for comparative proteomics: model studies with two serotypes of adenovirus. Anal. Chem. 73, 2836–2842. ( 10.1021/ac001404c) [DOI] [PubMed] [Google Scholar]
  • 9.Gygi SP, Rist B, Gerber SA, Turecek F, Gelb MH, Aebersold R. 1999. Quantitative analysis of complex protein mixtures using isotope-coded affinity tags. Nat. Biotechnol. 17, 994–999. ( 10.1038/13690) [DOI] [PubMed] [Google Scholar]
  • 10.Zhou H, Ranish JA, Watts JD, Aebersold R. 2002. Quantitative proteome analysis by solid-phase isotope tagging and mass spectrometry. Nat. Biotechnol. 20, 512–515. ( 10.1038/nbt0502-512) [DOI] [PubMed] [Google Scholar]
  • 11.Li J, Steen H, Gygi SP. 2003. Protein profiling with cleavable isotope-coded affinity tag (cICAT) reagents: the yeast salinity stress response. Mol. Cell. Proteomics 2, 1198–1204. ( 10.1074/mcp.M300070-MCP200) [DOI] [PubMed] [Google Scholar]
  • 12.Goodlett DR, et al. 2001. Differential stable isotope labeling of peptides for quantitation and de novo sequence derivation. Rapid Commun. Mass Spectrom. 15, 1214–1221. ( 10.1002/rcm.362) [DOI] [PubMed] [Google Scholar]
  • 13.Chakraborty A, Regnier FE. 2002. Global internal standard technology for comparative proteomics. J. Chromatogr. A 949, 173–184. ( 10.1016/S0021-9673(02)00047-X) [DOI] [PubMed] [Google Scholar]
  • 14.Peters EC, Horn DM, Tully DC, Brock A. 2001. A novel multifunctional labeling reagent for enhanced protein characterization with mass spectrometry. Rapid Commun. Mass Spectrom. 15, 2387–2392. ( 10.1002/rcm.517) [DOI] [PubMed] [Google Scholar]
  • 15.Hsu JL, Huang SY, Chow NH, Chen SH. 2003. Stable-isotope dimethyl labeling for quantitative proteomics. Anal. Chem. 75, 6843–6852. ( 10.1021/ac0348625) [DOI] [PubMed] [Google Scholar]
  • 16.Hsu JL, Huang SY, Chen SH. 2006. Dimethyl multiplexed labeling combined with microcolumn separation and MS analysis for time course study in proteomics. Electrophoresis 27, 3652–3660. ( 10.1002/elps.200600147) [DOI] [PubMed] [Google Scholar]
  • 17.Wu Y, et al. 2014. Five-plex isotope dimethyl labeling for quantitative proteomics. Chem. Commun. 50, 1708–1710. ( 10.1039/c3cc47998f) [DOI] [PubMed] [Google Scholar]
  • 18.Boersema PJ, Aye TT, van Veen TA, Heck AJ, Mohammed S. 2008. Triplex protein quantification based on stable isotope labeling by peptide dimethylation applied to cell and tissue lysates. Proteomics 8, 4624–4632. ( 10.1002/pmic.200800297) [DOI] [PubMed] [Google Scholar]
  • 19.Thompson A, et al. 2003. Tandem mass tags: a novel quantification strategy for comparative analysis of complex protein mixtures by MS/MS. Anal. Chem. 75, 1895–1904. ( 10.1021/ac0262560) [DOI] [PubMed] [Google Scholar]
  • 20.Ross PL, et al. 2004. Multiplexed protein quantitation in Saccharomyces cerevisiae using amine-reactive isobaric tagging reagents. Mol. Cell. Proteomics 3, 1154–1169. ( 10.1074/mcp.M400129-MCP200) [DOI] [PubMed] [Google Scholar]
  • 21.Pierce A, et al. 2008. Eight-channel iTRAQ enables comparison of the activity of six leukemogenic tyrosine kinases. Mol. Cell. Proteomics 7, 853–863. ( 10.1074/mcp.M700251-MCP200) [DOI] [PubMed] [Google Scholar]
  • 22.Tao WA, Aebersold R. 2003. Advances in quantitative proteomics via stable isotope tagging and mass spectrometry. Curr. Opin. Biotechnol. 14, 110–118. ( 10.1016/S0958-1669(02)00018-6) [DOI] [PubMed] [Google Scholar]
  • 23.Sap KA, Demmers JAA.2012. Labeling methods in mass spectrometry based quantitative proteomics. In Integrative proteomics (ed. H-CE Leung), ch. 6. Rijeka, Croatia: InTech. ( ) [DOI]
  • 24.Kline KG, Sussman MR. 2010. Protein quantitation using isotope-assisted mass spectrometry. Annu. Rev. Biophys. 39, 291–308. ( 10.1146/annurev.biophys.093008.131339) [DOI] [PubMed] [Google Scholar]
  • 25.Schulze WX, Usadel B. 2010. Quantitation in mass-spectrometry-based proteomics. Annu. Rev. Plant Biol. 61, 491–516. ( 10.1146/annurev-arplant-042809-112132) [DOI] [PubMed] [Google Scholar]
  • 26.Bantscheff M, Schirle M, Sweetman G, Rick J, Kuster B. 2007. Quantitative mass spectrometry in proteomics: a critical review. Anal. Bioanal. Chem. 389, 1017–1031. ( 10.1007/s00216-007-1486-6) [DOI] [PubMed] [Google Scholar]
  • 27.Chaerkady R, Pandey A. 2007. Quantitative proteomics for identification of cancer biomarkers. Proteomics Clin. Appl. 1, 1080–1089. ( 10.1002/prca.200700284) [DOI] [PubMed] [Google Scholar]
  • 28.Zhou Y, Shan Y, Zhang L, Zhang Y. 2014. Recent advances in stable isotope labeling based techniques for proteome relative quantification. J. Chromatogr. A 1365, 1–11. ( 10.1016/j.chroma.2014.08.098) [DOI] [PubMed] [Google Scholar]
  • 29.Chen CH. 2008. Review of a current role of mass spectrometry for proteome research. Anal. Chim. Acta 624, 16–36. ( 10.1016/j.aca.2008.06.017) [DOI] [PubMed] [Google Scholar]
  • 30.Gevaert K, Impens F, Ghesquière B, Van Damme P, Lambrechts A, Vandekerckhove J. 2008. Stable isotopic labeling in proteomics. Proteomics 8, 4873–4885. ( 10.1002/pmic.200800421) [DOI] [PubMed] [Google Scholar]
  • 31.Chahrour O, Cobice D, Malone J. 2015. Stable isotope labeling methods in mass spectrometry-based quantitative proteomics. J. Pharm. Biomed. Anal. 113, 2–20. ( 10.1016/j.jpba.2015.04.013) [DOI] [PubMed] [Google Scholar]
  • 32.Altelaar AF, Frese CK, Preisinger C, Hennrich ML, Schram AW, Timmers HT, Heck AJ, Mohammed S. 2013. Benchmarking stable isotope labeling based quantitative proteomics. J. Proteomics 88, 14–26. ( 10.1016/j.jprot.2012.10.009) [DOI] [PubMed] [Google Scholar]
  • 33.Lau HT, Suh HW, Golkowski M, Ong SE. 2014. Comparing SILAC- and stable isotope dimethyl-labeling approaches for quantitative proteomics. J. Proteome Res. 13, 4164–4174. ( 10.1021/pr500630a) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Lundblad RL, Noyes CM. 1984. Chemical reagents for protein modification, vol. 1, ch. 10 Boca Raton, FL: CRC Press. [Google Scholar]
  • 35.Hermanson GT. 1996. Bioconjugate techniques. New York, NY: Academic Press. [Google Scholar]
  • 36.Hsu JL, Huang SY, Shiea JT, Huang WY, Chen SH. 2005. Beyond quantitative proteomics: signal enhancement of the a1 ion as a mass tag for peptide sequencing using dimethyl labeling. J. Proteome Res. 4, 101–108. ( 10.1021/pr049837%2B) [DOI] [PubMed] [Google Scholar]
  • 37.Xu B, Wang F, Song C, Sun Z, Cheng K, Tan Y, Wang H, Zou H. 2014. Large-scale proteome quantification of hepatocellular carcinoma tissues by a three-dimensional liquid chromatography strategy integrated with sample preparation. J Proteome Res. 13, 3645–3654. ( 10.1021/pr500200s) [DOI] [PubMed] [Google Scholar]
  • 38.Di Palma S, Raijmakers R, Heck AJ, Mohammed S. 2011. Evaluation of the deuterium isotope effect in zwitterionic hydrophilic interaction liquid chromatography separations for implementation in a quantitative proteomic approach. Anal. Chem. 83, 8352–8356. ( 10.1021/ac2018074) [DOI] [PubMed] [Google Scholar]
  • 39.Wu CJ, Chen YW, Tai JH, Chen SH. 2011. Quantitative phosphoproteomics studies using stable isotope dimethyl labeling coupled with IMAC-HILIC-nanoLC- MS/MS for estrogen-induced transcriptional regulation. J. Proteome Res. 10, 1088–1097. ( 10.1021/pr100864b) [DOI] [PubMed] [Google Scholar]
  • 40.Fu Q, Li L. 2005. De novo sequencing of neuropeptides using reductive isotopic methylation and investigation of ESI QTOF MS/MS fragmentation pattern of neuropeptides with N-terminal dimethylation. Anal. Chem. 77, 7783–7795. ( 10.1021/ac051324e) [DOI] [PubMed] [Google Scholar]
  • 41.Hansen KC, Schmitt-Ulms G, Chalkley RJ, Hirsch J, Baldwin MA, Burlingame AL. 2003. Mass spectrometric analysis of protein mixtures at low levels using cleavable 13C-isotope-coded affinity tag and multidimensional chromatography. Mol. Cell. Proteomics 2, 299–314. ( 10.1074/mcp.M300021-MCP200) [DOI] [PubMed] [Google Scholar]
  • 42.Ji C, Zhang N, Damaraju S, Damaraju VL, Carpenter P, Cass CE, Li L. 2007. A study of reproducibility of guanidination–dimethylation labeling and liquid chromatography matrix-assisted laser desorption ionization mass spectrometry for relative proteome quantification. Anal. Chim. Acta 585, 219–226. ( 10.1016/j.aca.2006.12.054) [DOI] [PubMed] [Google Scholar]
  • 43.Boersema PJ, Raijmakers R, Lemeer S, Mohammed S, Heck AJ. 2009. Multiplex peptide stable isotope dimethyl labeling for quantitative proteomics. Nat. Protoc. 4, 484–494. ( 10.1038/nprot.2009.21) [DOI] [PubMed] [Google Scholar]
  • 44.Ji C, Li L. 2005. Quantitative proteome analysis using differential stable isotopic labeling and microbore LC−MALDI MS and MS/MS. J. Proteome Res. 4, 734–742. ( 10.1021/pr049784w) [DOI] [PubMed] [Google Scholar]
  • 45.Boutilier JM, Warden H, Doucette AA, Wentzell PD. 2012. Chromatographic behaviour of peptides following dimethylation with H2/D2-formaldehyde: implications for comparative proteomics. J. Chromatogr. B 908, 59–66. ( 10.1016/j.jchromb.2012.09.035) [DOI] [PubMed] [Google Scholar]
  • 46.Cappadona S, Muñoz J, Spee WPE, Low TY, Mohammed S, van Breukelen B, Heck AJ. 2011. Deconvolution of overlapping isotopic clusters improves quantification of stable isotope-labeled peptides. J. Proteomics 74, 2204–2209. ( 10.1016/j.jprot.2011.04.022) [DOI] [PubMed] [Google Scholar]
  • 47.Han DK, Eng J, Zhou H, Aebersold R. 2001. Quantitative profiling of differentiation-induced microsomal proteins using isotope-coded affinity tags and mass spectrometry. Nat. Biotechnol. 19, 946–951. ( 10.1038/nbt1001-946) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Cox J, Mann M. 2008. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteomewide protein quantification. Nat. Biotechnol. 26, 1367–1372. ( 10.1038/nbt.1511) [DOI] [PubMed] [Google Scholar]
  • 49.Khan Z, Bloom JS, Garcia BA, Singh M, Kruglyak L. 2009. Protein quantification across hundreds of experimental conditions. Proc. Natl Acad. Sci. USA 106, 15 544–15 548. ( 10.1073/pnas.0904100106) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Mortensen P, et al. 2009. MSQuant, an open source platform for mass spectrometry-based quantitative proteomics. J. Proteome Res. 9, 393–403. ( 10.1021/pr900721e) [DOI] [PubMed] [Google Scholar]
  • 51.Wang F, Chen R, Zhu J, Sun D, Song C, Wu Y, Ye M, Wang L, Zou H. 2010. A fully automated system with online sample loading, isotope dimethyl labeling and multidimensional separation for high-throughput quantitative proteome analysis. Anal. Chem. 82, 3007–3015. ( 10.1021/ac100075y) [DOI] [PubMed] [Google Scholar]
  • 52.Raijmakers R, Berkers CR, de Jong A, Ovaa H, Heck AJ, Mohammed S. 2008. Automated online sequential isotope labeling for protein quantitation applied to proteasome tissue-specific diversity. Mol. Cell. Proteomics 7, 1755–1762. ( 10.1074/mcp.M800093-MCP200) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Zhou Y, Shan Y, Wu Q, Zhang S, Zhang L, Zhang Y. 2013. Mass defect-based pseudo-isobaric dimethyl labeling for proteome quantification. Anal. Chem. 85, 10 658–10 663. ( 10.1021/ac402834w) [DOI] [PubMed] [Google Scholar]
  • 54.Kovanich D, Cappadona S, Raijmakers R, Mohammed S, Scholten A, Heck AJ. 2012. Applications of stable isotope dimethyl labeling in quantitative proteomics. Anal. Bioanal. Chem. 404, 991–1009. ( 10.1007/s00216-012-6070-z) [DOI] [PubMed] [Google Scholar]
  • 55.Chen SH, et al. 2010. Nucleophosmin in the pathogenesis of arsenic-related bladder carcinogenesis revealed by quantitative proteomics. Toxicol. Appl. Pharmacol. 242, 126–135. ( 10.1016/j.taap.2009.09.016) [DOI] [PubMed] [Google Scholar]
  • 56.Munoz J, Low TY, Kok YJ, Chin A, Frese CK, Ding V, Choo A, Heck AJ. 2011. The quantitative proteomes of human-induced pluripotent stem cells and embryonic stem cells. Mol. Syst. Biol. 7, 550 ( 10.1038/msb.2011.84) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Ji C, Li L, Gebre M, Pasdar M, Li L. 2005. Identification and quantification of differentially expressed proteins in E-cadherin deficient SCC9 cells and SCC9 transfectants expressing E-cadherin by dimethyl isotope labeling, LC-MALDI MS and MS/MS. J. Proteome Res. 4, 1419–1426. ( 10.1021/pr050094h) [DOI] [PubMed] [Google Scholar]
  • 58.Sun T, et al. 2011. Activation of multiple proto-oncogenic tyrosine kinases in breast cancer via loss of the PTPN12 phosphatase. Cell 144, 703–718. ( 10.1016/j.cell.2011.02.003) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Yi CH, et al. 2011. Metabolic regulation of protein N-alpha-acetylation by Bcl-xL promotes cell survival. Cell 146, 607–620. ( 10.1016/j.cell.2011.06.050) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.D'Aguanno S, et al. 2014. p63 isoforms regulate metabolism of cancer stem cells. J. Proteome Res. 13, 2120–2136. ( 10.1021/pr4012574) [DOI] [PubMed] [Google Scholar]
  • 61.Swa HL, Shaik AA, Lim LH, Gunaratne J. 2015. Mass spectrometry based quantitative proteomics and integrative network analysis accentuates modulating roles of annexin-1 in mammary tumorigenesis. Proteomics 15, 408–418. ( 10.1002/pmic.201400175) [DOI] [PubMed] [Google Scholar]
  • 62.Sato M, Matsubara T, Adachi J, Hashimoto Y, Fukamizu K, Kishida M, Yang YA, Wakefield LM, Tomonaga T. 2015. Differential proteome analysis identifies TGF-β-related pro-metastatic proteins in a 4T1 murine breast cancer model. PLoS ONE 10, e0126483 ( 10.1371/journal.pone.0126483) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Chiang CY, et al. 2015. SH3BGRL3 protein as a potential prognostic biomarker for urothelial carcinoma: a novel binding partner of epidermal growth factor receptor. Clin. Cancer Res. 21, 5601–5611. ( 10.1158/1078-0432.CCR-14-3308) [DOI] [PubMed] [Google Scholar]
  • 64.Chang PC, Reddy PM, Ho YP. 2014. Quantification of genetically modified soya using strong anion exchange chromatography and time-of-flight mass spectrometry. Anal. Bioanal. Chem. 406, 5339–5346. ( 10.1007/s00216-014-7965-7) [DOI] [PubMed] [Google Scholar]
  • 65.Tolonen AC, Haas W, Chilaka AC, Aach J, Gygi SP, Church GM. 2011. Proteome-wide systems analysis of a cellulosic biofuel-producing microbe. Mol. Syst. Biol. 7, 461 ( 10.1038/msb.2010.116) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Mann M, Jensen ON. 2003. Proteomic analysis of post-translational modifications. Nat. Biotechnol. 21, 255–261. ( 10.1038/nbt0303-255) [DOI] [PubMed] [Google Scholar]
  • 67.Witze ES, Old WM, Resing KA, Ahn NG. 2007. Mapping protein posttranslational modifications with mass spectrometry. Nat. Methods 4, 798–806. ( 10.1038/nmeth1100) [DOI] [PubMed] [Google Scholar]
  • 68.Young NL, Plazas-Mayorca MD, Garcia BA. 2010. Systems-wide proteomic characterization of combinatorial posttranslational modification patterns. Expert Rev. Proteomics 7, 79–92. ( 10.1586/epr.09.100) [DOI] [PubMed] [Google Scholar]
  • 69.Choudhary C, Mann M. 2010. Decoding signalling networks by mass spectrometry-based proteomics. Nat. Rev. Mol. Cell Biol. 11, 427–439. ( 10.1038/nrm2900) [DOI] [PubMed] [Google Scholar]
  • 70.Lemeer S, Jopling C, Gouw J, Mohammed S, Heck AJ, Slijper M, den Hertog J. 2008. Comparative phosphoproteomics of zebrafish Fyn/Yes morpholino knockdown embryos. Mol. Cell. Proteomics 7, 2176–2187. ( 10.1074/mcp.M800081-MCP200) [DOI] [PubMed] [Google Scholar]
  • 71.Boersema PJ, et al. 2010. In-depth qualitative and quantitative profiling of tyrosine phosphorylation using a combination of phosphopeptide immunoaffinity purification and stable isotope dimethyl labeling. Mol. Cell. Proteomics 9, 84–99. ( 10.1074/mcp.M900291-MCP200) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Melo-Braga MN, Schulz M, Liu Q, Swistowski A, Palmisano G, Engholm-Keller K, Jakobsen L, Zeng X, Larsen MR. 2014. Comprehensive quantitative comparison of the membrane proteome, phosphoproteome, and sialiome of human embryonic and neural stem cells. Mol. Cell. Proteomics 13, 311–328. ( 10.1074/mcp.M112.026898) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Huang H, Larsen MR, Palmisano G, Dai J, Lametsch R. 2014. Quantitative phosphoproteomic analysis of porcine muscle within 24 h postmortem. J. Proteomics 106, 125–139. ( 10.1016/j.jprot.2014.04.020) [DOI] [PubMed] [Google Scholar]
  • 74.Engholm-Keller K, Birck P, Størling J, Pociot F, Mandrup-Poulsen T, Larsen MR. 2012. TiSH—a robust and sensitive global phosphoproteomics strategy employing a combination of TiO2, SIMAC, and HILIC. J. Proteomics 75, 5749–5761. ( 10.1016/j.jprot.2012.08.007) [DOI] [PubMed] [Google Scholar]
  • 75.Huang J, et al. 2014. In situ sample processing approach (iSPA) for comprehensive quantitative phosphoproteome analysis. J. Proteome Res. 13, 3896–3904. ( 10.1021/pr500454g) [DOI] [PubMed] [Google Scholar]
  • 76.Wakabayashi M, Yoshihara H, Masuda T, Tsukahara M, Sugiyama N, Ishihama Y. 2014. Phosphoproteome analysis of formalin-fixed and paraffin-embedded tissue sections mounted on microscope slides. J. Proteome Res. 13, 915–924. ( 10.1021/pr400960r) [DOI] [PubMed] [Google Scholar]
  • 77.Scholten A, et al. 2013. Phosphoproteomics study based on in vivo inhibition reveals sites of calmodulin-dependent protein kinase II regulation in the heart. J. Am. Heart Assoc. 2, e000318 ( 10.1161/JAHA.113.000318) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Renvoisé M, Bonhomme L, Davanture M, Valot B, Zivy M, Lemaire C. 2014. Quantitative variations of the mitochondrial proteome and phosphoproteome during fermentative and respiratory growth in Saccharomyces cerevisiae. J. Proteomics 106, 140–150. ( 10.1016/j.jprot.2014.04.022) [DOI] [PubMed] [Google Scholar]
  • 79.Wei X, Herbst A, Ma D, Aiken J, Li L. 2011. A quantitative proteomic approach to prion disease biomarker research: delving into the glycoproteome. J. Proteome Res. 10, 2687–2702. ( 10.1021/pr2000495) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Chen R, Wang F, Tan Y, Sun Z, Song C, Ye M, Wang H, Zou H. 2012. Development of a combined chemical and enzymatic approach for the mass spectrometric identification and quantification of aberrant N-glycosylation. J. Proteomics 75, 1666–1674. ( 10.1016/j.jprot.2011.12.015) [DOI] [PubMed] [Google Scholar]
  • 81.Parker BL, et al. 2011. Quantitative N-linked glycoproteomics of myocardial ischemia and reperfusion injury reveals early remodeling in the extracellular environment. Mol. Cell. Proteomics 10, M110.006833 ( 10.1074/mcp.M110.006833) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Huang SY, Tsai ML, Wu CJ, Hsu JL, Ho SH, Chen SH. 2006. Quantitation of protein phosphorylation in pregnant rat uteri using stable isotope dimethyl labeling coupled with IMAC. Proteomics 6, 1722–1734. ( 10.1002/pmic.200500507) [DOI] [PubMed] [Google Scholar]
  • 83.Song C, et al. 2011. Improvement of the quantification accuracy and throughput for phosphoproteome analysis by a pseudo triplex stable isotope dimethyl labeling approach. Anal. Chem. 83, 7755–7762. ( 10.1021/ac201299j) [DOI] [PubMed] [Google Scholar]
  • 84.Varki A. 1993. Biological roles of oligosaccharides: all of the theories are correct. Glycobiology 3, 97–130. ( 10.1093/glycob/3.2.97) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Peracaula R, Tabares G, Royle L, Harvey DJ, Dwek RA, Rudd PM, de Llorens R. 2003. Altered glycosylation pattern allows the distinction between prostate-specific antigen (PSA) from normal and tumor origins. Glycobiology 13, 457–470. ( 10.1093/glycob/cwg041) [DOI] [PubMed] [Google Scholar]
  • 86.Palmisano G, Lendal SE, Engholm-Keller K, Leth-Larsen R, Parker BL, Larsen MR. 2010. Selective enrichment of sialic acid-containing glycopeptides using titanium dioxide chromatography with analysis by HILIC and mass spectrometry. Nat. Protoc. 5, 1974–1982. ( 10.1038/nprot.2010.167) [DOI] [PubMed] [Google Scholar]
  • 87.Lin CY, Ma YC, Pai PJ, Her GR. 2012. A comparative study of glycoprotein concentration, glycoform profile and glycosylation site occupancy using isotope labeling and electrospray linear ion trap mass spectrometry. Anal. Chim. Acta 728, 49–56. ( 10.1016/j.aca.2012.03.058) [DOI] [PubMed] [Google Scholar]
  • 88.Weng Y, Qu Y, Jiang H, Wu Q, Zhang L, Yuan H, Zhou Y, Zhang X, Zhang Y. 2014. An integrated sample pretreatment platform for quantitative N-glycoproteome analysis with combination of on-line glycopeptide enrichment, deglycosylation and dimethyl labeling. Anal. Chim. Acta 833, 1–8. ( 10.1016/j.aca.2014.04.037) [DOI] [PubMed] [Google Scholar]
  • 89.Sun Z, Qin H, Wang F, Cheng K, Dong M, Ye M, Zou H. 2012. Capture and dimethyl labeling of glycopeptides on hydrazide beads for quantitative glycoproteomics analysis. Anal. Chem. 84, 8452–8456. ( 10.1021/ac302130r) [DOI] [PubMed] [Google Scholar]
  • 90.Zhang H, et al. 2005. High throughput quantitative analysis of serum proteins using glycopeptide capture and liquid chromatography mass spectrometry. Mol. Cell. Proteomics 4, 144–155. ( 10.1074/mcp.M400090-MCP200) [DOI] [PubMed] [Google Scholar]
  • 91.Huang J, et al. 2015. A peptide N-terminal protection strategy for comprehensive glycoproteome analysis using hydrazide chemistry based method. Sci. Rep. 5, 10164 ( 10.1038/srep10164) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Chen X, et al. 2016. Comparative profiling of triple-negative breast carcinomas tissue glycoproteome by sequential purification of glycoproteins and stable isotope labeling. Cell. Physiol. Biochem. 38, 110–121. ( 10.1159/000438613) [DOI] [PubMed] [Google Scholar]
  • 93.Hennrich ML, Mohammed S, Altelaar AF, Heck AJ. 2010. Dimethyl isotope labeling assisted de novo peptide sequencing. J. Am. Soc. Mass Spectrom. 21, 1957–1965. ( 10.1016/j.jasms.2010.08.007) [DOI] [PubMed] [Google Scholar]
  • 94.Shen PT, Hsu JL, Chen SH. 2007. Dimethyl isotope-coded affinity selection for the analysis of free and blocked N-termini of proteins using LC-MS/MS. Anal. Chem. 79, 9520–9530. ( 10.1021/ac701678h) [DOI] [PubMed] [Google Scholar]
  • 95.Kleifeld O, et al. 2010. Isotopic labeling of terminal amines in complex samples identifies protein N-termini and protease cleavage products. Nat. Biotechnol. 28, 281–288. ( 10.1038/nbt.1611) [DOI] [PubMed] [Google Scholar]
  • 96.Kleifeld O, Doucet A, Prudova A, Auf dem Keller U, Gioia M, Kizhakkedathu JN, Overall CM. 2011. Identifying and quantifying proteolytic events and the natural N terminome by terminal amine isotopic labeling of substrates. Nat. Protoc. 6, 1578–1611. ( 10.1038/nprot.2011.382) [DOI] [PubMed] [Google Scholar]
  • 97.Schilling O, Barré O, Huesgen PF, Overall CM. 2010. Proteomewide analysis of protein carboxy termini: C terminomics. Nat. Methods 7, 508–511. ( 10.1038/nmeth.1467) [DOI] [PubMed] [Google Scholar]
  • 98.Guo J, Prokai-Tatrai K, Prokai L. 2012. Relative quantitation of protein nitration by liquid chromatography-mass spectrometry using isotope-coded dimethyl labeling and chemoprecipitation. J. Chromatogr. A 1232, 266–275. ( 10.1016/j.chroma.2011.12.100) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Rice JC, Allis CD. 2001. Histone methylation versus histone acetylation: new insights into epigenetic regulation. Curr. Opin. Cell Biol. 13, 263–273. ( 10.1016/S0955-0674(00)00208-8) [DOI] [PubMed] [Google Scholar]
  • 100.Chen SH, Chen CR, Chen SH, Li DT, Hsu JL. 2013. Improved Nα-acetylated peptide enrichment following dimethyl labeling and SCX. J. Proteome Res. 12, 3277–3287. ( 10.1021/pr400127j) [DOI] [PubMed] [Google Scholar]
  • 101.Hsu JL, Chen SH, Li DT, Shi FK. 2007. Enhanced a1 fragmentation for dimethylated proteins and its applications for N-terminal identification and comparative protein quantitation. J. Proteome Res. 6, 2376–2383. ( 10.1021/pr060639n) [DOI] [PubMed] [Google Scholar]
  • 102.Huang SY, Wen CH, Li DT, Hsu JL, Chen C, Shi FK, Lin YY. 2008. Assignment of disulfide-linked peptides using automatic a1 ion recognition. Anal. Chem. 80, 9135–9140. ( 10.1021/ac8013725) [DOI] [PubMed] [Google Scholar]
  • 103.Huang SY, Hsieh YT, Chen CH, Chen CC, Sung WC, Chou MY, Chen SF. 2012. Automatic disulfide bond assignment using a1 ion screening by mass spectrometry for structural characterization of protein pharmaceuticals. Anal. Chem. 84, 4900–4906. ( 10.1021/ac3005007) [DOI] [PubMed] [Google Scholar]
  • 104.Huang SY, Chen SF, Chen CH, Huang HW, Wu WG, Sung WC. 2014. Global disulfide bond profiling for crude snake venom using dimethyl labeling coupled with mass spectrometry and RADAR algorithm. Anal. Chem. 86, 8742–8750. ( 10.1021/ac501931t) [DOI] [PubMed] [Google Scholar]
  • 105.Chicooree N, Connolly Y, Tan CT, Malliri A, Li Y, Smith DL, Griffiths JR. 2013. Enhanced detection of ubiquitin isopeptides using reductive methylation. J. Am. Soc. Mass Spectrom. 24, 421–430. ( 10.1007/s13361-012-0538-0) [DOI] [PubMed] [Google Scholar]
  • 106.Wu CJ, Hsu JL, Huang SY, Chen SH. 2010. Mapping N-terminus phosphorylation sites and quantitation by stable isotope dimethyl labeling. J. Am. Soc. Mass Spectrom. 21, 460–471. ( 10.1016/j.jasms.2009.12.001) [DOI] [PubMed] [Google Scholar]

Articles from Philosophical transactions. Series A, Mathematical, physical, and engineering sciences are provided here courtesy of The Royal Society

RESOURCES