Protein Modification
Mary Johnson (han at labome dot com)
Synatom Research, Princeton, New Jersey, United States
DOI
//dx.doi.org/10.13070/mm.en.2.118
Date
last modified : 2022-09-24; original version : 2012-04-09
Cite as
MATER METHODS 2012;2:118
Abstract

The article provides a summary of protein modifications from the UniProt database and an overview of research methods for major protein modifications: phosphorylation, acetylation and methylation, ubiquitination, glycosylation, and sumoylation.

Introduction

The human proteome consists of significantly more functional polypeptides than the collection of genes in the genome, due, in part, to co- and post-translational protein modifications (Fig 1A). One of the important areas of proteomic research is to identify those proteins that are post-translationally modified and their modification sites and to characterize the function of the modifications and the interaction of the modified protein within a functional cellular network.

Protein Modification figure 1
Figure 1. Protein modifications. A, Post-translational modifications, in conjunction with alternative splicing, play an important role in the generation of multiple functional proteins from a single gene. B, Post-translational modifications play important roles in diverse cell functions as intracellular signaling (phosphorylation), regulation of protein stability (ubiquitination), regulation of transcription (histone acetylation and methylation), and cell surface signaling (glycosylation).

The curated SWISS-Prot Protein Knowledgebase lists 695 unique modifications [6], as of September 24, 2022 (UniProt Release 2022_05). RESID database [7] by Dr. John S. Garavelli also provides comprehensive information for each modification.

Multiple methods to assay protein modifications have been developed over the past decades. Here we focus on the mass spectrometry as a general approach for detecting protein modifications, and specific methods for phosphorylation, ubiquitination, glycosylation, and sumoylation, as well as for analyzing histone acetylation and methylation in the context of chromatin remodeling (Table 1 and Fig 1B). Detailed protocols are not covered. Discussion about citrullination, using histone citrullination as an example, can be found at the review about histone modifications. Protein citrullination can also be investigated through site-specific incorporation of citrulline into proteins with a modified leucyl tRNA synthetase-tRNA pair [8]. The biotin-switch technique can be used to assay the degree of S-nitrosylation of protein of interest [9].

Modification Function Assay
PhosphorylationIntracellular signalingMass spectrometry
Radiometry
Western blotting
Phos-tag electrophoresis
UbiquitinationProtein degradationMass spectrometry
In vitro ubiquitination assays
Western blotting
TUBEs
Acetylation and methylationChromatin regulation
Transcriptional regulation
Mass spectrometry
CHIP
CHIP-on-CHIP
GlycosylationExtracellular signalingMass spectrometry
HPLC
Electrophoresis
SUMOylationIntracellular transport
Transcriptional regulation
Apoptosis
Protein stability
Stress response
Cell-cycle regulation
Mass spectrometry
SUBEs
PRISM
Western blotting [10]
Table 1. Types of protein modifications, their main cellular functions, and major research methods.

A major preliminary approach for identifying novel modification sites is the in silico analysis. Computer programs that identify putative modification sites based on sequence have been extensively utilized and are beyond the scope of this review. See Liu and Li, 2011 [11] for an excellent overview of computer programs.

Mass Spectrometry

Over the past 20 years, mass spectrometric analysis has become an essential tool in determining the types and sites of protein modifications. Mass spectrometric analysis can be performed on both purified proteins or a mixture of proteins, for example, cell lysates [12, 13] or tissue extracts [14, 15].

A mass spectrometer generates gas-phase ions from a sample, separates them according to their mass-to-charge ratio (m/z) and generates a record of their abundance. Mass spectrometry can be used for molecular weight determination of peptides and proteins, for determining the amino acid sequence of peptides, and for the detection of post-translational modifications, as well as the relative quantification of peptides and proteins. This method cannot be used for absolute quantification.

For the preparation of a sample for mass spectrometric analysis, the protein or lysate is digested into small peptide fragments using trypsin or, more recently, other proteases [16]. The fragments are then vaporized and analyzed to determine their m/z value. Since the sites of restriction digest are known, a computer program then determines the unique amino acid sequence of each peptide based on mass. The molecular weight and charge of a moiety such as a phosphate group are also known, the phosphorylation of specific amino acids within a peptide fragment can also be assessed.

A digested protein sample can be analyzed by matrix-assisted laser desorption/ionization (MALDI) peptide mapping or nanoelectrospray mass spectrometer. The use of these methods, however, can result in incomplete coverage, with some of the peptides clearly visible and others – not at all. This is often a major problem in analyzing complex mixtures, and for modified peptides and in the analysis of post-translational modifications, peptides are first separated by reversed-phase chromatography, with subsequent fraction collection and analysis by mass spectrometry (a method known as LC/MS) (Fig 2B) [17]. To more accurately determine the nature of the peptide modification, tandem mass spectrometry (MS/MS) experiments are often performed (Fig 2A). After the first MS step, the peptide ions collide with an inert gas, which leads to further fragmentation. These peptide fragments are then analyzed in the second MS step [18]. Some modified peptides will remain intact during this process, generating a fragmentation pattern similar to the unmodified peptide, while others will fragment significantly, generating a pattern that can provide further information about the nature and location of the modified amino acids. For example, Liu Y et al identified demethylation and methylation on differet residues of ALKBH5 with nano-ultra-performance liquid chromatography-electrospray inonization tandem mass spectrometry using Q-Exactive MS from Thermo Fisher Scientific, after immunoprecipitation and SDS-PAGE separation [19]. Hoxhaj G et al used LC-MS/MS to determine the sites on NAD kinase phosphorylated by akt kinase [20]. Black MH et al used LC-MS/MS to identify glutamylated peptides in SdeA by the Legionella meta-effector SidJ [21]. Bhogaraju S et al performed further analysis on the glutamylation of SdeA [22].

Protein Modification figure 2
Figure 2. Mass spectrometry as a tool for general protein modification analysis. A, Mass spectrometry (MS) analysis utilizes peptide ionization to analyze the mass to charge (m/z) ratio of protein fragments. In a further step (in MS/MS analysis), the products are further fragmented to obtain additional data. B, In LC-MS/MS, the complex mixture of proteins is separated via a liquid chromatography step (left panel), before being subjected to MS/MS (right panel) to identify the individual peptide fragments [1].

In addition to being used to determine the modification state of a single protein, mass spectrometry has been used to generate extensive datasets of proteins that carry a specific modification [23]. Some examples are Asn deamidation [14], phosphorylation [24], sumoylation [25], ubiquitination [26], non-histone [27] and histone methylation [28], glycosylation [29] or oxidized proteins [30-33]. This analysis usually involves affinity selection prior to the mass spectrometric analysis [34]. Such techniques are used to define ‘sub-proteomes’ while analyzing the activation state of entire signaling networks in cancer [35], neurodegenerative diseases [36, 37] or pathogen-host interactions [38, 39].

However, mass spectrometric analysis is costly and requires specific equipment and training. Because more quantitative data is often required, other biochemical methods are used in conjunction with mass spectrometry to analyze post-translational modifications. Moreover, MS-based analysis of PTMs has its own caveats that can lead to erroneous conclusions. Older [12], [13], and more recent [40-42] extensive reviews of the multiple MS methods, common errors and suggestions for accurate data collection and computational analysis are available [43, 44].

Phosphorylation

Phosphorylation, or the addition of a phosphate group to serine, threonine, or tyrosine residues, is one of most common forms of protein modification (Fig 3A). Protein phosphorylation plays an important role in intracellular signaling cascades and is reversible, fine-tuning the signal [45]. Several in vivo and in vitro methods are used to assess protein phosphorylation status, to assess phosphorylation of a specific amino acid, and to ascertain whether a specific kinase (or phosphatase) modifies a protein in question. In addition to the methods discussed below, Pro-Q® Diamond phosphoprotein gel stain can directly label phosphoproteins on a 2D PAGE gel, as in the case of SY5Y cells treated with Ab42 peptides [46].

Protein Modification figure 3
Figure 3. Protein phosphorylation analysis through antibody-based assays. A, During phosphorylation, a phosphate group is added to a serine, threonine, or tyrosine residue. This phosphate group can be used as a marker, either in using radioactive phosphate for radiometric assays or by utilizing antibodies raised against the phosphorylated amino acid. B, An in vivo phosphorylation assay demonstrates the phosphorylation of TH1 by c-Src. The two proteins are ectopically expressed, an immunoprecipitation against Myc-tagged TH1 is performed, and a general phosphotyrosine antibody is used to assess TH1 phosphorylation in the absence or presence of c-Src expression [2].
In vitro phosphorylation analysis

Radiometric kinase reactions using 32P-gamma-ATP can be used to assay phosphorylation status in vitro [47]. This is also the gold standard for assessing the role of specific kinases. Purified target protein, kinase, and 32P-gamma-ATP are incubated in a reaction buffer. The reaction solution is then passed through a filter to bind protein but wash away unbound ATP, and the level of phosphorylation (radioisotope retained on a filter) is detected by a scintillation counter. Alternatively, the reaction can be resolved by SDS-PAGE and visualized by exposure to X-ray film [48]. This method is less quantitative but can confirm the molecular weight of the phosphorylated protein.

Similarly, the reaction can include phosphatase instead of the kinase to determine whether a phosphorylated protein is a substrate of a specific phosphatase.

Radioactive pulse labeling

To assay phosphorylation in vivo, radioactive pulse labeling have been utilized [49]. Cells are grown in the presence 32P-orthophosphate. The protein of interest is then immunoprecipitated with a specific antibody and the retained radioisotope quantified by scintillation counter or resolved on an SDS-PAGE and exposed to X-ray film. This method can be used to assay phosphorylation under various physiological conditions. Furthermore, in conjunction with a protein knock-down step, this method can also be used to implicate a specific kinase or phosphatase in the modification status of the target protein.

Phospho-specific antibodies

There are two categories of phospho-specific antibodies. One category is the generic phosphotyrosine, phosphoserine, and phosphothreonine antibody, which will bind to any phospho-tyrosine, phospho-serine, and phospho-threonine moieties regardless of the neighboring amino acid residues. The second category includes the antibodies (or other affinity reagents [50] ) against phospho-specific amino acid epitopes (see a summary of phospho-specific antibodies in Labome product database).

Following identification of a (putative) phosphorylation site by an in silico method or by mass spectrometry, commercially available antibodies for the general class of phosphorylation sites can be used in conjunction with an immunoprecipitation step to determine if the protein of interest is phosphorylated at a specific type of site (for example a canonical cyclin/cdk site) [51]. Antibodies generated against a specific phosphorylated amino acid, such as phosphotyrosine, can also be used (Fig 3B). This can be followed up by generating a phospho-specific antibody to the specific epitope and assaying phosphorylation by simple Western blotting. To analyze a large number of samples, an ELISA can be performed, where the substrate is captured by the membrane and detected by the phospho-specific antibody [52]. Kenner LR et al confirmed phosphorylation of human eIF2alpha through western bloting with an eIF2alpha S51 phosphorylation-specific antibody from Cell Signaling (cat# 9721) [53]. However, the specificify of phosphorylation-specific antibodies is often questioned [54].

Anti-phosphoprotein antibody microarrays are powerful tools for investigating protein phosphorylation-mediated signaling systems. These microarrays facilitate the semiquantitative measurement of along the phosphorylation status of protein kinases and their substrates. A detailed protocol is reviewed here [55].

Detection of phosphoproteins with Phos-tag

Phos-tag is a selective phosphate-binding molecule, 1,3-bis[bis(pyridin-2-ylmethyl) amino]propan-2-olato dizinc(II), developed for MS-based proteomics [56], and can be used for separation of phosphorylated proteins in acrylamide and agarose gels [57]. By using Phos-tag SDS-PAGE various phosphorylated and unphosphorylated protein species can be visualized due to their noticeable mobility shifts [58-60]. The method is easy to use as it resembles the classical SDS-PAGE analysis. Biotinylated Phos-tag can also be used to facilitate Western blot analysis of samples [61, 62]. Protocols for Phos-tag SDS-PAGE with either alkaline pH [63] or neutral pH [64] buffer systems are now available. A review on recent advances of the Phos-tag based methodologies was published in 2015 [63]. Kenner LR et al confirmed phosphorylation of human eIF2alpha through Phos-Tag SDS-PAGE [53].

Apart from being useful tools for the identification of phosphoproteins, the Phos-tag SDS-PAGE technique, in combination with other non-denaturing gel electrophoresis methods proved to be powerful methods for the characterization of the phosphorylation-dependent activity of certain enzymes. For example, Meisrimler et al combined the Phos-tag technology with two-dimensional electrophoresis in which either non-reducing isoelectric focusing (IEF) PAGE or non-equilibrium pH gel electrophoresis (NEPHGE) was used in the first dimension and high resolution clear native electrophoresis (hrCNE) in the second dimension, to investigate phosphorylation-dependent changes in enzyme activity [65]. The authors discussed in detail the advantages and the disadvantages of these new techniques [65]. Kitchen P et al used Phos-tag reagent from NARD Chemicals, Kobe City, Japan to analyze the phosphorylation of AQP4 [66].

Ubiquitination

Protein ubiquitination, or the covalent attachment of ubiquitin to target proteins, has been best studied in the context of protein degradation. For example, the ubiquitination-mediated protein turnover has been shown to play an essential role in driving the cell cycle [67]. However, recent research has also revealed a role for ubiquitination in multiple protein-degradation-independent intracellular signaling pathways.

As an example, a protocol for the detection of linear polyubiquitin chains by an anti-linear ubiquitin antibody used by Sasaki et al in their studies can be found here [68]. Advantages and disadvantages of the commonly used immunoblotting-based methods, as well as the suggestion for their optimization, are highlighted in a recent review [69]. The review includes recommendations on sample preparation, separation, detection and identification of ubiquitin chains, as well as on the analysis of the ubiquitin chain topology.

Alternatively, ubiquitinated proteins can be detected by affinity purification. Two such approaches are becoming more popular: ligase trapping [70], and the TUBEs (Tandem Ubiquitin Binding Entity) technique [71]. In ligase trapping, ubiquitin ligases are fused to a polyubiquitin-binding domain, which allows the isolation of specific ubiquitinated substrates that can be further analyzed by mass spectrometry or western blotting. Similarly, TUBEs, which are engineered tagged sequences of four ubiquitin-binding domains linked together via flexible likers, are used to affinity purify polyubiquitinated proteins from cell extracts [71, 72],]. The TUBEs protect the ubiquitinated protein from degradation and de-ubiquitination, thus facilitating the identification of ubiquitination target sites as, for example, specific substrates of ubiquitin ligases [73]. TUBEs for protein purification (agarose-TUBEs, GST- and His6-TUBEs), immunohistochemistry (fluorescent TUBEs), or Far Western (ligand) blotting (biotin-TUBEs) are now available for researchers from LifeSciences.

The diversity of ubiquitin-mediated signaling depends on the multiple ways in which a target protein can be modified by the ubiquitin – monoubiquitination at one or several sites, and polyubiquitination via several possible linkages [74]. Substrate proteins are linked to ubiquitin via seven distinct ubiquitin lysine residues (Lys6, Lys11, Lys27, Lys29, Lys33, Lys48 and Lys63). Formation of a polyubiquitin chain happens when a lysine residue of ubiquitin is linked to the carboxy-terminal glycine of another ubiquitin. Thus the functional outcome of polyubiquitination depends on the lysine residue within ubiquitin that is used. This is, in turn, determined by the specific E2 (ubiquitin-conjugating enzyme) or E3 (ubiquitin ligase), but not the E1 (ubiquitin-activating enzyme) (Fig 4A). The generation of polyubiquitin chains with specific lysine linkages creates a unique binding surface for the specialized ubiquitin-binding domains of interacting proteins.

Protein Modification figure 4
Figure 4. Protein ubiquitination in vivo and in vitro can be analyzed by a Western blotting-based assay. A, In vivo and in vitro protein ubiquitination assays require the presence of the substrate protein, the E1, E2, and E3, ATP, and ubiquitin. The ubiquitinated substrate can then be visualized by SDS-PAGE and Western blotting. B, An in vivo ubiquitination assay reveals that RNF185 mediates the K63-linked polyubiquitination of BNIP1. The substrate (BNIP1), the E3 (RNF185), and the ubiquitin are all ectopically expressed, the Flag-tagged substrate is then immunoprecipitated, and the myc-tagged ubiquitin visualized by Western blotting [3].

Methods for assaying ubiquitination can reveal the ubiquitination at specific lysine residues or confirm the role of specific E3s (or E2s) in the modification of the target protein. The most straight-forward and commonly accepted way to assess ubiquitination in vivo is to immunoprecipitate the putatively ubiquitinated protein, and subject it to SDS-PAGE/Western blotting to observe the laddering typical of ubiquitinated proteins. The target protein, E3 ligase, and even ubiquitin are often ectopically expressed to determine the effects of mutation (Fig 4B). Inhibitors of proteasomal degradation, such as MG132, are frequently used to analyze the in vivo K48 (degradation-targeting) ubiquitination of target proteins.

In vitro ubiquitination assays (where the target protein, ubiquitin, and the E1, E2, and E3 are provided) can be used, in conjunction with SDS-PAGE followed by Western blotting to more directly assay the ubiquitination of the protein in question. Mutations of lysine residues within the protein in question can serve to determine the site of ubiquitin attachment. Conversely, in the case of poly-ubiquitination, utilizing ubiquitin with mutations of specific lysine residues can provide information about the specific ubiquitin linkage. To discover novel ubiquitinated proteins, ubiquitin-mediated protein purification followed by mass spectrometric analysis can be performed [75].

Acetylation and Methylation

Epigenetic modifications (such as phosphorylation, ubiquitination, acetylation, and methylation) of the conserved core histones H2A, H2B, H3, and H4 play an important regulatory role in gene expression [76]. For example, acetylation and methylation of lysine residues on the N-terminal histone tails mediate the formation of chromatin domains, such as euchromatin and heterochromatin, thus directly mediating gene silencing. Assaying the modification status of core histones has been an important technique in analyzing the regulation of gene expression [77]. Detailed histone modification research methods are discussed here.

Acetylation and methylation of other proteins have also been studied, for example, K49 acetylation of beta-catenin [78], or α-tubulin acetylation [79]. M Oginuma et al detected the acetylated beta-catenin in Western blots using a specific antibody ( 9534 from Cell Signaling Technology) [78].

Modified histone antibodies and ChIP

Extensive collections of antibodies are commercially available for all known histone modification sites. Using these antibodies, a chromatin immunoprecipitation (ChIP) experiment can be performed [80]. Briefly, the cell or tissue sample is treated with a cross-linking agent such as formaldehyde to cross-link histones to chromatin. The chromatin is sonicated or treated with a nuclease to generate short DNA fragments. An immunoprecipitation (IP) step is then performed using an antibody against the desired histone modification. If the putative histone modification site within a gene or promoter is known, PCR can be used to quantify the presence of the modification in question at a given site. (Fig 5A). Alternatively, this method can be used to identify the histone modification site within the general promoter region, as well as the kinetics of histone modification at that site (Fig 5B).

Protein Modification figure 5
Figure 5. Acetylation and methylation of core histones can be determined by chromatin immunoprecipitation. A, Chromatin immunoprecipitation (ChIP) can be used to assess core histone modification and, consequently, the epigenetic status of genes. B, ChIP analysis is utilized to narrow down sites of histone modification within a broader promoter region, as well as to assay modification kinetics for the fos gene following TCA treatment. Histone methylation within a 3kb region including the fos gene promoter is determined for up to 4 hours after TCA treatment [4].
ChIP-on-chip

If the site of histone modification is not known, a ChIP-on-chip can be performed to identify the regions in the genome carrying specific histone modifications [81]. In this setup, the DNA fragments in the IP input and the IP itself are labeled with different fluorophores. The samples are then passed over a chip with DNA probes, and the enrichment of a given DNA fragment in the IP fraction correlates with the localization of the modified histone in question to a given site in the genome.

ChIP and ChIP-on-chip experiments can provide information about histone modification at a promoter of interest, the kinetics of change in the modification status, as well as the location of histone modifications with respect to a gene or regulatory sequence, or throughout the genome. These epigenetic changes can shed light both on gene regulation in normal function and the dysregulation that occurs in disease.

Glycosylation

Glycosylation, or the attachment of one of a large number of glycan groups, is predicted to occur in close to 50% of all proteins and plays a role in biological processes as diverse as embryonic development, cell division, and regulation of protein structure. The glycosylation status of a wide array of proteins changes significantly during normal physiological events, and in diseases (for example inflammation, sepsis, and cancer). Thus, assays for protein glycosylation can be informative in research aimed both at disease prognosis and therapeutic purposes.

The two main types of protein glycosylation are N-glycosylation (in which the glycan is attached to an asparagine) and O-glycosylation (in which the glycan is attached to a serine or threonine). Glycosylation differs from protein modifications discussed above in that a vast array of modifying glycans has been described. Analysis of protein glycosylation is aimed at identifying the glycan group, the modified protein, or the site of attachment. In general, glycosylation analysis can be performed using three different approaches, as outlined in Fig 6A: the analysis of chemically or enzymatically released glycans themselves, the characterization of glycopeptides following a tryptic digest, or the characterization of glycans in intact glycoproteins.

Protein Modification figure 6
Figure 6. Glycosylation can be assayed by mass spectrometry to identify specific modifying glycans. A, Glycosylation can be assayed by mass spectrometry by three general methods: assaying the intact proteoglycan, assaying products of a tryptic digest, or assaying the released and isolated glycans. B, LC-MS analysis of fetal calf serum fetuin reveals a change in glycosylation when incubated with Cc5 bacteria (lower panel) as compared to untreated (upper panel). The fetuin was digested with trypsin and then subjected to LC-MS analysis. The blue squares represent GlcNAc, the red and green circles - Man and Gal, and the orange diamonds Sial [5].
Glycan analysis

For the analysis of glycans, mass spectrometry analysis, sometimes coupled with an HPLC step, can be utilized. The sugar groups are first released from the glycoprotein either enzymatically (peptide N glycosidase A or F) or chemically (hydrazinolysis). LC/MS analysis can then be performed directly on the released oligosaccharides (for example, [82] ). Alternatively, the reducing terminus produced by enzymatic or chemical cleavage can be tagged with a fluorophore label, and the fluorophore-labeled glycans can be analyzed by a number of possible HPLC methods, such as hydrophilic interaction HPLC [83].

An added fluorophore or chromophore at the glycan’s reducing end allows for their separation by electrophoresis or chromatography, and for their quantitation. Currently, solid-phase based approaches such as glycoprotein immobilization or on-tissue glycan mass spectrometry imaging are used alongside the more common solution-based analyses. A recent review of the review, methods for glycan isolation, identification, and quantitation can be found here [84].

Reagents with high affinity for glycans, like lectins and glycan-binding antibodies, have been used mostly to detect glycosylated proteins in clinical samples. Recent advances involve the use of glycan arrays, lectin engineering, and quantitative lectin measurements. Some of the challenges and advantages of these new tools for biological samples analysis have been reviewed recently [85].

Glycoprotein identification

While the methods described above provide information on the specific glycans, they are not informative when it comes to the identity of the protein carrying a specific modification or the site of attachment. To obtain this information, endoproteinase cleavage is performed (e.g., a tryptic digest), and the glycopeptides are analyzed by LC/MS or HILIC-MS/MS. This method can provide information on either all proteins carrying a specific glycan moiety or the site within a given protein where the glycan is attached [86]. For example, in Fig 6B, the effect of incubation with Cc5 bacteria on the glycosylation status of specific fetuin residues is assayed, following the tryptic digest, by LC-MS.

For some glycopeptides, subjecting the intact protein to mass spectrometric analysis can provide data about the protein identity and its glycosylation status. This method, however, is dependent on the biochemical properties of the glycopeptide and often provides a lower level of resolution than the two methods described above.

Multiple reviews of the major developments in the field of glycoproteomics that focus on the most recent advancements (2012-2015) that involve the use of MS-based approaches have been published [87-90].

Protein Modification figure 7
Figure 7. Main steps of the PRISM technique for SUMOylation analysis.
Sumoylation

Sumoylation, the addition of SUMOs (small ubiquitin-like modifiers) to proteins, is a post-translational modification similar to ubiquitination. As all other PTMs, sumoylation can affect a protein’s structure, subcellular localization, and function. As for the identification of other PTMs, mass-spectrometry is a useful tool despite its limitations. As of June 2016, a few recent reviews of the MS-based techniques for PTMs analysis have focused not only on the more studied types (phosphorylation, glycosylation, acetylation, methylation or ubiquitination) but also on the less common modifications, like sumoylation [41, 91].

Several other methods have been proposed. Among them, the use of SUMO-specific molecular traps, called SUBEs (SUMO Binding Entities), that is similar to the previously described TUBEs (see Ubiquitination above) and for which a methodological overview has been published [72] was important in the understanding of tumor-suppressing pathways in human cells [92].

Additionally, the Protease-Reliant Identification of SUMO Modification (PRISM) method, which allows for detection of SUMOylated proteins and the identification of SUMOylation sites, has been used [93]. The method involves the chemical blocking of all free lysines, with SUMO-specific proteases cleavage, and MS-based identification of the unblocked lysines (Figure 7).

References
  1. Vitek O. Getting started in computational mass spectrometry-based proteomics. PLoS Comput Biol. 2009;5:e1000366 pubmed publisher
  2. Wu W, Sun Z, Wu J, Peng X, Gan H, Zhang C, et al. Trihydrophobin 1 phosphorylation by c-Src regulates MAPK/ERK signaling and cell migration. PLoS ONE. 2012;7:e29920 pubmed publisher
  3. Tang F, Wang B, Li N, Wu Y, Jia J, Suo T, et al. RNF185, a novel mitochondrial ubiquitin E3 ligase, regulates autophagy through interaction with BNIP1. PLoS ONE. 2011;6:e24367 pubmed publisher
  4. Hazzalin C, Mahadevan L. Dynamic acetylation of all lysine 4-methylated histone H3 in the mouse nucleus: analysis at c-fos and c-jun. PLoS Biol. 2005;3:e393 pubmed
  5. Renzi F, Manfredi P, Mally M, Moes S, Jeno P, Cornelis G. The N-glycan glycoprotein deglycosylation complex (Gpd) from Capnocytophaga canimorsus deglycosylates human IgG. PLoS Pathog. 2011;7:e1002118 pubmed publisher
  6. Controlled vocabulary of posttranslational modifications. Available from: www.uniprot.org/help/controlled_vocabulary
  7. RESID Database at PIR. Available from: pir.georgetown.edu/resid/
  8. Mondal S, Wang S, Zheng Y, Sen S, Chatterjee A, Thompson P. Site-specific incorporation of citrulline into proteins in mammalian cells. Nat Commun. 2021;12:45 pubmed publisher
  9. Faraco G, Hochrainer K, Segarra S, Schaeffer S, Santisteban M, Menon A, et al. Dietary salt promotes cognitive impairment through tau phosphorylation. Nature. 2019;: pubmed publisher
  10. Bellail A, Jin H, Lo H, Jung S, Hamdouchi C, Kim D, et al. Ubiquitination and degradation of SUMO1 by small-molecule degraders extends survival of mice with patient-derived tumors. Sci Transl Med. 2021;13:eabh1486 pubmed publisher
  11. Liu C, Li H. In silico prediction of post-translational modifications. Methods Mol Biol. 2011;760:325-40 pubmed publisher
  12. Mann M, Hendrickson R, Pandey A. Analysis of proteins and proteomes by mass spectrometry. Annu Rev Biochem. 2001;70:437-73 pubmed
  13. Domon B, Aebersold R. Mass spectrometry and protein analysis. Science. 2006;312:212-7 pubmed
  14. Hsueh M, Onnerfjord P, Bolognesi M, Easley M, Kraus V. Analysis of "old" proteins unmasks dynamic gradient of cartilage turnover in human limbs. Sci Adv. 2019;5:eaax3203 pubmed publisher
  15. Adaikkan C, Middleton S, Marco A, Pao P, Mathys H, Kim D, et al. Gamma Entrainment Binds Higher-Order Brain Regions and Offers Neuroprotection. Neuron. 2019;102:929-943.e8 pubmed publisher
  16. Trevisiol S, Ayoub D, Lesur A, Ancheva L, Gallien S, Domon B. The use of proteases complementary to trypsin to probe isoforms and modifications. Proteomics. 2016;16:715-28 pubmed publisher
  17. Korfmacher W. Principles and applications of LC-MS in new drug discovery. Drug Discov Today. 2005;10:1357-67 pubmed
  18. Verbeck G, Ruotolo B, Sawyer H, Gillig K, Russell D. A fundamental introduction to ion mobility mass spectrometry applied to the analysis of biomolecules. J Biomol Tech. 2002;13:56-61 pubmed
  19. Liu Y, You Y, Lu Z, Yang J, Li P, Liu L, et al. N6-methyladenosine RNA modification-mediated cellular metabolism rewiring inhibits viral replication. Science. 2019;365:1171-1176 pubmed publisher
  20. Hoxhaj G, Ben Sahra I, Lockwood S, Timson R, Byles V, Henning G, et al. Direct stimulation of NADP+ synthesis through Akt-mediated phosphorylation of NAD kinase. Science. 2019;363:1088-1092 pubmed publisher
  21. Black M, Osinski A, Gradowski M, Servage K, Pawłowski K, Tomchick D, et al. Bacterial pseudokinase catalyzes protein polyglutamylation to inhibit the SidE-family ubiquitin ligases. Science. 2019;364:787-792 pubmed publisher
  22. Bhogaraju S, Bonn F, Mukherjee R, Adams M, Pfleiderer M, Galej W, et al. Inhibition of bacterial ubiquitin ligases by SidJ-calmodulin catalysed glutamylation. Nature. 2019;572:382-386 pubmed publisher
  23. Hennrich M, Gavin A. Quantitative mass spectrometry of posttranslational modifications: keys to confidence. Sci Signal. 2015;8:re5 pubmed publisher
  24. von Stechow L, Francavilla C, Olsen J. Recent findings and technological advances in phosphoproteomics for cells and tissues. Expert Rev Proteomics. 2015;12:469-87 pubmed publisher
  25. Eifler K, Vertegaal A. Mapping the SUMOylated landscape. FEBS J. 2015;282:3669-80 pubmed publisher
  26. Beaudette P, Popp O, Dittmar G. Proteomic techniques to probe the ubiquitin landscape. Proteomics. 2016;16:273-87 pubmed publisher
  27. Biggar K, Li S. Non-histone protein methylation as a regulator of cellular signalling and function. Nat Rev Mol Cell Biol. 2015;16:5-17 pubmed publisher
  28. Onder O, Sidoli S, Carroll M, Garcia B. Progress in epigenetic histone modification analysis by mass spectrometry for clinical investigations. Expert Rev Proteomics. 2015;12:499-517 pubmed publisher
  29. Goldman R, Sanda M. Targeted methods for quantitative analysis of protein glycosylation. Proteomics Clin Appl. 2015;9:17-32 pubmed publisher
  30. Baez N, Reisz J, Furdui C. Mass spectrometry in studies of protein thiol chemistry and signaling: opportunities and caveats. Free Radic Biol Med. 2015;80:191-211 pubmed publisher
  31. Artemenko K, Mi J, Bergquist J. Mass-spectrometry-based characterization of oxidations in proteins. Free Radic Res. 2015;49:477-93 pubmed publisher
  32. Verrastro I, Pasha S, Jensen K, Pitt A, Spickett C. Mass spectrometry-based methods for identifying oxidized proteins in disease: advances and challenges. Biomolecules. 2015;5:378-411 pubmed publisher
  33. Lennicke C, Rahn J, Heimer N, Lichtenfels R, Wessjohann L, Seliger B. Redox proteomics: Methods for the identification and enrichment of redox-modified proteins and their applications. Proteomics. 2016;16:197-213 pubmed publisher
  34. Rainer M, Bonn G. Enrichment of phosphorylated peptides and proteins by selective precipitation methods. Bioanalysis. 2015;7:243-52 pubmed publisher
  35. Lim Y. Mining the tumor phosphoproteome for cancer markers. Clin Cancer Res. 2005;11:3163-9 pubmed
  36. Conti A, Alessio M. Comparative Proteomics for the Evaluation of Protein Expression and Modifications in Neurodegenerative Diseases. Int Rev Neurobiol. 2015;121:117-52 pubmed publisher
  37. Gallart Palau X, Serra A, Sze S. Uncovering Neurodegenerative Protein Modifications via Proteomic Profiling. Int Rev Neurobiol. 2015;121:87-116 pubmed publisher
  38. Ravikumar V, Jers C, Mijakovic I. Elucidating Host-Pathogen Interactions Based on Post-Translational Modifications Using Proteomics Approaches. Front Microbiol. 2015;6:1313 pubmed publisher
  39. Yang Y, Hu M, Yu K, Zeng X, Liu X. Mass spectrometry-based proteomic approaches to study pathogenic bacteria-host interactions. Protein Cell. 2015;6:265-74 pubmed publisher
  40. Larance M, Lamond A. Multidimensional proteomics for cell biology. Nat Rev Mol Cell Biol. 2015;16:269-80 pubmed publisher
  41. Doll S, Burlingame A. Mass spectrometry-based detection and assignment of protein posttranslational modifications. ACS Chem Biol. 2015;10:63-71 pubmed publisher
  42. Na S, Paek E. Software eyes for protein post-translational modifications. Mass Spectrom Rev. 2015;34:133-47 pubmed publisher
  43. Kim M, Zhong J, Pandey A. Common errors in mass spectrometry-based analysis of post-translational modifications. Proteomics. 2016;16:700-14 pubmed publisher
  44. Schwämmle V, Verano Braga T, Roepstorff P. Computational and statistical methods for high-throughput analysis of post-translational modifications of proteins. J Proteomics. 2015;129:3-15 pubmed publisher
  45. Cohen P. The regulation of protein function by multisite phosphorylation--a 25 year update. Trends Biochem Sci. 2000;25:596-601 pubmed
  46. Zatsepina O, Kechko O, Mitkevich V, Kozin S, Yurinskaya M, Vinokurov M, et al. Amyloid-β with isomerized Asp7 cytotoxicity is coupled to protein phosphorylation. Sci Rep. 2018;8:3518 pubmed publisher
  47. Hastie C, McLauchlan H, Cohen P. Assay of protein kinases using radiolabeled ATP: a protocol. Nat Protoc. 2006;1:968-71 pubmed
  48. Yoon I, Nam M, Kim H, Moon H, Kim S, Jang J, et al. Glucose-dependent control of leucine metabolism by leucyl-tRNA synthetase 1. Science. 2019;: pubmed publisher
  49. Peck S. Analysis of protein phosphorylation: methods and strategies for studying kinases and substrates. Plant J. 2006;45:512-22 pubmed
  50. Meksiriporn B, Ludwicki M, Stephens E, Jiang A, Lee H, Waraho Zhmayev D, et al. A survival selection strategy for engineering synthetic binding proteins that specifically recognize post-translationally phosphorylated proteins. Nat Commun. 2019;10:1830 pubmed publisher
  51. Zhang H, Zha X, Tan Y, Hornbeck P, Mastrangelo A, Alessi D, et al. Phosphoprotein analysis using antibodies broadly reactive against phosphorylated motifs. J Biol Chem. 2002;277:39379-87 pubmed
  52. Blaydes J, Vojtesek B, Bloomberg G, Hupp T. The development and use of phospho-specific antibodies to study protein phosphorylation. Methods Mol Biol. 2000;99:177-89 pubmed
  53. Kenner L, Anand A, Nguyen H, Myasnikov A, Klose C, McGeever L, et al. eIF2B-catalyzed nucleotide exchange and phosphoregulation by the integrated stress response. Science. 2019;364:491-495 pubmed publisher
  54. Frohner I, Mudrak I, Schüchner S, Anrather D, Hartl M, Sontag J, et al. PP2AC Phospho-Tyr307 Antibodies Are Not Specific for this Modification but Are Sensitive to Other PP2AC Modifications Including Leu309 Methylation. Cell Rep. 2020;30:3171-3182.e6 pubmed publisher
  55. Zhang H, Shi X, Pelech S. Monitoring Protein Kinase Expression and Phosphorylation in Cell Lysates with Antibody Microarrays. Methods Mol Biol. 2016;1360:107-22 pubmed publisher
  56. Takeda H, Kawasaki A, Takahashi M, Yamada A, Koike T. Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry of phosphorylated compounds using a novel phosphate capture molecule. Rapid Commun Mass Spectrom. 2003;17:2075-81 pubmed
  57. Nagy Z, Comer S, Smolenski A. Analysis of Protein Phosphorylation Using Phos-Tag Gels. Curr Protoc Protein Sci. 2018;93:e64 pubmed publisher
  58. Kinoshita E, Kinoshita Kikuta E, Koike T. Phos-tag SDS-PAGE systems for phosphorylation profiling of proteins with a wide range of molecular masses under neutral pH conditions. Proteomics. 2012;12:192-202 pubmed publisher
  59. Kinoshita E, Kinoshita Kikuta E, Kubota Y, Takekawa M, Koike T. A Phos-tag SDS-PAGE method that effectively uses phosphoproteomic data for profiling the phosphorylation dynamics of MEK1. Proteomics. 2016;16:1825-36 pubmed publisher
  60. Sugiyama Y, Katayama S, Kameshita I, Morisawa K, Higuchi T, Todaka H, et al. Expression and phosphorylation state analysis of intracellular protein kinases using Multi-PK antibody and Phos-tag SDS-PAGE. MethodsX. 2015;2:469-74 pubmed publisher
  61. Horinouchi T, Terada K, Higashi T, Miwa S. Using Phos-Tag in Western Blotting Analysis to Evaluate Protein Phosphorylation. Methods Mol Biol. 2016;1397:267-277 pubmed publisher
  62. Kinoshita Kikuta E, Kinoshita E, Koike T. Phosphopeptide Detection with Biotin-Labeled Phos-tag. Methods Mol Biol. 2016;1355:17-29 pubmed publisher
  63. Kinoshita E, Kinoshita Kikuta E, Koike T. Separation and detection of large phosphoproteins using Phos-tag SDS-PAGE. Nat Protoc. 2009;4:1513-21 pubmed publisher
  64. Kinoshita Kikuta E, Kinoshita E, Koike T. Neutral Phosphate-Affinity SDS-PAGE system for profiling of protein phosphorylation. Methods Mol Biol. 2015;1295:323-54 pubmed publisher
  65. Meisrimler C, Schwendke A, Lüthje S. Two-dimensional phos-tag zymograms for tracing phosphoproteins by activity in-gel staining. Front Plant Sci. 2015;6:230 pubmed publisher
  66. Kitchen P, Salman M, Halsey A, Clarke Bland C, MacDonald J, Ishida H, et al. Targeting Aquaporin-4 Subcellular Localization to Treat Central Nervous System Edema. Cell. 2020;181:784-799.e19 pubmed publisher
  67. Tyers M, Jorgensen P. Proteolysis and the cell cycle: with this RING I do thee destroy. Curr Opin Genet Dev. 2000;10:54-64 pubmed
  68. Sasaki Y, Fujita H, Nakai M, Iwai K. Immunoblot analysis of linear polyubiquitination of NEMO. Methods Mol Biol. 2015;1280:297-309 pubmed publisher
  69. Emmerich C, Cohen P. Optimising methods for the preservation, capture and identification of ubiquitin chains and ubiquitylated proteins by immunoblotting. Biochem Biophys Res Commun. 2015;466:1-14 pubmed publisher
  70. Mark K, Loveless T, Toczyski D. Isolation of ubiquitinated substrates by tandem affinity purification of E3 ligase-polyubiquitin-binding domain fusions (ligase traps). Nat Protoc. 2016;11:291-301 pubmed publisher
  71. Hjerpe R, Aillet F, Lopitz Otsoa F, Lang V, England P, Rodriguez M. Efficient protection and isolation of ubiquitylated proteins using tandem ubiquitin-binding entities. EMBO Rep. 2009;10:1250-8 pubmed publisher
  72. Lang V, Aillet F, Da Silva Ferrada E, Xolalpa W, Zabaleta L, Rivas C, et al. Analysis of PTEN ubiquitylation and SUMOylation using molecular traps. Methods. 2015;77-78:112-8 pubmed publisher
  73. Yoshida Y, Saeki Y, Murakami A, Kawawaki J, Tsuchiya H, Yoshihara H, et al. A comprehensive method for detecting ubiquitinated substrates using TR-TUBE. Proc Natl Acad Sci U S A. 2015;112:4630-5 pubmed publisher
  74. Behrends C, Harper J. Constructing and decoding unconventional ubiquitin chains. Nat Struct Mol Biol. 2011;18:520-8 pubmed publisher
  75. Tomlinson E, Palaniyappan N, Tooth D, Layfield R. Methods for the purification of ubiquitinated proteins. Proteomics. 2007;7:1016-22 pubmed
  76. Grant P. A tale of histone modifications. Genome Biol. 2001;2:REVIEWS0003 pubmed
  77. Bartova E, Krejci J, Harnicarová A, Galiova G, Kozubek S. Histone modifications and nuclear architecture: a review. J Histochem Cytochem. 2008;56:711-21 pubmed publisher
  78. Oginuma M, Harima Y, Tarazona O, Diaz Cuadros M, Michaut A, Ishitani T, et al. Intracellular pH controls WNT downstream of glycolysis in amniote embryos. Nature. 2020;: pubmed publisher
  79. Magupalli V, Negro R, Tian Y, Hauenstein A, Di Caprio G, Skillern W, et al. HDAC6 mediates an aggresome-like mechanism for NLRP3 and pyrin inflammasome activation. Science. 2020;369: pubmed publisher
  80. Spencer V, Sun J, Li L, Davie J. Chromatin immunoprecipitation: a tool for studying histone acetylation and transcription factor binding. Methods. 2003;31:67-75 pubmed
  81. Buck M, Lieb J. ChIP-chip: considerations for the design, analysis, and application of genome-wide chromatin immunoprecipitation experiments. Genomics. 2004;83:349-60 pubmed
  82. Bangaru S, Ozorowski G, Turner H, Antanasijevic A, Huang D, Wang X, et al. Structural analysis of full-length SARS-CoV-2 spike protein from an advanced vaccine candidate. Science. 2020;: pubmed publisher
  83. Wuhrer M, Deelder A, Hokke C. Protein glycosylation analysis by liquid chromatography-mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci. 2005;825:124-33 pubmed
  84. Yang S, Rubin A, Eshghi S, Zhang H. Chemoenzymatic method for glycomics: Isolation, identification, and quantitation. Proteomics. 2016;16:241-56 pubmed publisher
  85. Tang H, Hsueh P, Kletter D, Bern M, Haab B. The detection and discovery of glycan motifs in biological samples using lectins and antibodies: new methods and opportunities. Adv Cancer Res. 2015;126:167-202 pubmed publisher
  86. Zauner G, Koeleman C, Deelder A, Wuhrer M. Protein glycosylation analysis by HILIC-LC-MS of Proteinase K-generated N- and O-glycopeptides. J Sep Sci. 2010;33:903-10 pubmed publisher
  87. Kleparnik K. Recent advances in combination of capillary electrophoresis with mass spectrometry: methodology and theory. Electrophoresis. 2015;36:159-78 pubmed publisher
  88. Feng X, Liu B, Li J, Liu X. Advances in coupling microfluidic chips to mass spectrometry. Mass Spectrom Rev. 2015;34:535-57 pubmed publisher
  89. Lazar I, Deng J, Ikenishi F, Lazar A. Exploring the glycoproteomics landscape with advanced MS technologies. Electrophoresis. 2015;36:225-37 pubmed publisher
  90. Hu H, Khatri K, Zaia J. Algorithms and design strategies towards automated glycoproteomics analysis. Mass Spectrom Rev. 2017;36:475-498 pubmed publisher
  91. Chicooree N, Unwin R, Griffiths J. The application of targeted mass spectrometry-based strategies to the detection and localization of post-translational modifications. Mass Spectrom Rev. 2015;34:595-626 pubmed publisher
  92. Ho J, Bassi C, Stambolic V. Characterization of nuclear PTEN and its post translational modifications. Methods. 2015;77-78:104-11 pubmed publisher
  93. Hendriks I, D Souza R, Chang J, Mann M, Vertegaal A. System-wide identification of wild-type SUMO-2 conjugation sites. Nat Commun. 2015;6:7289 pubmed publisher
ISSN : 2329-5139