ACS Publications. Most Trusted. Most Cited. Most Read
My Activity
CONTENT TYPES

Figure 1Loading Img
RETURN TO ISSUEPREVTechnical Note

Unambiguous Phosphosite Localization using Electron-Transfer/Higher-Energy Collision Dissociation (EThcD)

View Author Information
Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands.
Netherlands Proteomics Centre, Padualaan 8, 3584 CH Utrecht, The Netherlands
§ Research Institute of Molecular Pathology (IMP), Dr. Bohrgasse 7, A-1030 Vienna, Austria
Institute of Molecular Biotechnology (IMBA), Vienna, Austria
Cite this: J. Proteome Res. 2013, 12, 3, 1520–1525
Publication Date (Web):January 24, 2013
https://doi.org/10.1021/pr301130k

Copyright © 2013 American Chemical Society. This publication is licensed under these Terms of Use.

  • Open Access

Article Views

4641

Altmetric

-

Citations

136
LEARN ABOUT THESE METRICS
PDF (705 KB)
Supporting Info (1)»

Abstract

We recently introduced a novel scheme combining electron-transfer and higher-energy collision dissociation (termed EThcD), for improved peptide ion fragmentation and identification. We reasoned that phosphosite localization, one of the major hurdles in high-throughput phosphoproteomics, could also highly benefit from the generation of such EThcD spectra. Here, we systematically assessed the impact on phosphosite localization utilizing EThcD in comparison to methods employing either ETD or HCD, respectively, using a defined synthetic phosphopeptide mixture and also using a larger data set of Ti4+-IMAC enriched phosphopeptides from a tryptic human cell line digest. In combination with a modified version of phosphoRS, we observed that in the majority of cases EThcD generated richer and more confidently identified spectra, resulting in superior phosphosite localization scores. Our data demonstrates the distinctive potential of EThcD for PTM localization, also beyond protein phosphorylation.

Introduction

ARTICLE SECTIONS
Jump To

Reversible phosphorylation of proteins is a key regulatory mechanism in living cells. (1) Protein phosphorylation can modulate protein activity, turnover, subcellular localization, complex formation, folding and degradation. Dynamic phosphorylation plays a pivotal role in almost all biological processes including cell division, differentiation, polarization and apoptosis. (2) Moreover, it is an important switch in cellular signal transduction. (3) The importance of this post-translational modification (PTM) for cell biology has driven the development of novel mass spectrometric tools for sensitive and global detection of phosphorylation. (4, 5) However, the analysis of phosphorylated peptides by mass spectrometry is still not as straightforward as for “regular”, unmodified peptides. One of the major challenges in phosphoproteomics is to improve MS level representation since phosphopeptides are usually present at substoichiometric levels. Hence, an enrichment step is necessary to enable deeper penetration of the phosphoproteome. Enrichment is typically performed by chromatography, (6) antibodies (7) or metal-ion/metal oxide affinity-based (8, 9) techniques. Two other main challenges are the identification of phosphopeptides and confident localization of the corresponding phosphosite. (10) The challenge is caused by the higher lability of the phosphate group when compared to the amide bond. A number of strategies have been proposed to circumvent poor fragmentation and improve sequence and site diagnostic fragmentation, including the use of neutral loss-triggered MS/MS/MS (11) and multistage activation (MSA) (12) in ion traps, the use of beam type CID fragmentation, (13) and electron capture/transfer dissociation (14) or a combination of some of these approaches. (9, 15)
Once phosphopeptide identification is feasible through sufficient peptide backbone fragments, it can still be challenging to pinpoint the true phosphosite. This becomes more difficult as the number of potential phosphorylation sites within the peptide sequence increases. In principle, unambiguous phosphosite localization requires site-determining fragment ions. (16) Direct validation is feasible through detection of a fragment ion that carries the phosphate group. Neutral loss fragment ions can be used as well; however, since they exhibit the same mass as a water loss from an unmodified residue they do not directly confirm the correct site. (17) Diagnostic phosphosite-specific fragments facilitate pinpointing the correct phosphosite. (18-20) Several algorithms and programs have been developed to enable automatic phosphosite localization. (3, 16, 21-26) These software tools are based on distinct but similar approaches and they all aim to provide a metric that allows for assessment of the confidence in phosphosite localization. Recently, Taus et al. have reported on a new algorithm, coined phosphoRS, (27) which presently is uniquely compatible with CID, HCD and ETD fragmentation and was optimized for both low- and high-resolution MS/MS spectra. phosphoRS provides individual localization probabilities for all potential phosphosites in a given peptide.
Generally, all scoring tools depend on the quality of the MS/MS spectra. The more site-determining ions are detected, the higher the confidence in phosphosite localization. We have recently introduced a novel fragmentation scheme combining electron-transfer and higher-energy collision dissociation, termed EThcD. (28) This method employs dual fragmentation to generate both b/y and c/z ions which leads to very fragment ion- and thus data-rich MS/MS spectra. Compared to HCD and ETD, we found a substantial increase in peptide backbone fragmentation, which translated into a remarkable average peptide sequence coverage of ∼94% for tryptic peptides. We reasoned that localization of post-translational modifications could also highly benefit from EThcD spectra. Here, we systematically assessed the impact on phosphosite localization using EThcD. In this work we evaluate the performance of EThcD in comparison to ETD and HCD using a defined synthetic phosphopeptide mixture and also on a larger data set of Ti4+-IMAC enriched phosphopeptides, all in combination with a modified version of phosphoRS.

Experimental Section

ARTICLE SECTIONS
Jump To

Materials

All chemicals were purchased from Sigma-Aldrich (Steinheim, Germany) unless otherwise stated. Formic acid and ammonia were obtained from Merck (Darmstadt, Germany). Acetonitrile was purchased from Biosolve (Valkenswaard, The Netherlands).

Sample Preparation

Protein from HeLa cells was harvested and digested with trypsin, as previously described. (29) Ti4+-IMAC beads were prepared as reported elsewhere. (30, 31) Phosphopeptides were enriched as previously described. (32) Briefly, Gel-loader tips that were plugged with C8 material (3M, Zoeterwoude, The Netherlands) were filled up to 1 cm with Ti4+-IMAC beads. Columns were equilibrated with loading buffer (80% ACN, 6% TFA). Peptides were reconstituted in loading buffer, loaded onto the columns and washed with washing buffer 1 (50% ACN, 0.5% TFA, 200 mM NaCl) and subsequently washing buffer 2 (50% ACN, 0.1% TFA). Phosphopeptides were eluted with elution buffer 1 (10% NH3 in H20) followed by elution buffer 2 (80% ACN, 2% FA). Eluate was acidified and diluted with formic acid to a final acetonitrile concentration of <5%, split into three equal amounts and directly analyzed by single run LC–MS/MS utilizing ETD, HCD and EThcD, respectively.

Mass Spectrometry

All data was acquired on an ETD enabled Thermo Scientific LTQ Orbitrap Velos mass spectrometer (Thermo Fisher Scientific, Bremen, Germany). A Thermo Scientific EASY-nLC 1000 (Thermo Fisher Scientific, Odense, Denmark) was connected to the LTQ Orbitrap Velos mass spectrometer. ETD, HCD and EThcD methods were set up as previously described. (28) Briefly, all spectra were acquired in the Orbitrap at a resolution of 7500. For HCD the normalized collision energy was set to 40%. The ETD reaction time was set to 50 ms for ETD and EThcD. Supplemental activation was enabled for ETD. HCD normalized collision energy was set to 30% for EThcD (calculation based on precursor m/z and charge state). The anion AGC target was set to 4e5 for both ETD and EThcD.

Data Analysis

Peak lists were generated using Thermo Scientific Proteome Discoverer 1.3 software (Thermo Fisher Scientific, Bremen, Germany). The nonfragment filter was used to simplify ETD spectra with the following settings: the precursor peak was removed within a 4 Da window, charged reduced precursors were removed within a 2 Da window, and neutral losses from charge reduced precursors were removed within a 2 Da window (the maximum neutral loss mass was set to 120 Da). MS/MS spectra were searched against a database containing the synthetic phosphopeptide sequences and the human Uniprot database (version v2010–12), respectively, including a list of common contaminants using SEQUEST or Mascot (Matrix Science, UK). The precursor mass tolerance was set to 10 ppm, the fragment ion mass tolerance was set to 0.02 Da. Enzyme specificity was set to Trypsin with 2 missed cleavages allowed. Data from the synthetic phosphopeptide mixture was searched with no enzyme specificity. Oxidation of methionine and phosphorylation (S,T,Y) were used as variable modification and carbamidomethylation of cysteines was set as fixed modification. Percolator (33) was used to filter the PSMs for <1% false-discovery-rate. Phosphorylation sites were localized by applying a custom version of phosphoRS (27) (v3.0 – EThcD enabled) that has been expanded to allow analysis of EThcD data. (28) Briefly, the algorithm considers both HCD- and ETD-type fragment ions at the same time. While singly and doubly charged b- and y-type fragment ions including neutral loss of phosphoric acid (H3PO4) are considered for site localization, only singly charged c-, z-radical and z-prime ions are scored.

Results and Discussion

ARTICLE SECTIONS
Jump To

Increasing the confidence in phosphosite localization is a key challenge in phosphoproteomics. Site-determining fragment ions are required to unambiguously pinpoint the correct phosphosite. Observing all possible peptide backbone cleavages in a single MS/MS spectrum substantially simplifies phosphosite localization. Recently, we showed that EThcD enables complete peptide sequencing through dual fragmentation. (28) In EThcD, the peptide precursor is initially subjected to an ion/ion reaction with fluoranthene anions in a linear ion trap, which generates c- and z-ions. However, the unreacted precursor and the charge-reduced precursor remain highly abundant after ETD. In the second step HCD all-ion fragmentation is applied to all ETD derived ions. This generates b- and y-ions from the unreacted precursor and simultaneously increases the yield of c- and z-ions by fragmentation of the charge reduced precursor. Since the remaining unreacted precursor population is higher charged than the ETD-derived fragment ions one can apply a level of energy that fragments the precursor but does not induce secondary fragmentation of c- and z-ions. Here, we continue to explore the benefits of this novel fragmentation mode for the analysis of phosphopeptides.

Evaluation of Phosphosite Localization by EThcD using a Defined Phosphopeptide Mixture

To evaluate the potential added value of phosphopeptide analysis by EThcD we initially used a defined mixture of well-characterized synthetic phosphopeptides. This mixture consists of 30 phosphopeptides of varying length with up to four phosphorylated residues (see Supplementary Table 1 for a complete list, Supporting Information). We analyzed this mixture by LC–MS/MS employing ETD, HCD and EThcD fragmentation, respectively. We used identical instrument settings with the only exception being the parameters for peptide dissociation, which were set to the for each method optimized values. The data was searched with SEQUEST and the PSMs were manually validated and filtered (7 ppm peptide mass tolerance, search engine rank 1, absolute Xcorr threshold 0.4). Additionally, we considered only PSMs for which the injection time did not max out (<500 ms), that is, the target number of ions was reached. Note that this precaution was taken to exclude the number of ions as a variable that might impair the quality of fragmentation. We calculated the average precursor ion purity (PIP) (34) for each data set and found similar values, which were approximately 95% for all three techniques. Together, these stringent criteria ensure that the activation technique is the only variable that controls the fragmentation behavior. A summary of the data from this direct comparison is given in Table 1. Similar numbers of PSMs were identified for all three fragmentation techniques. We found that EThcD provided 248 PSMs while these numbers were 237 and 216 for HCD and ETD, respectively. Out of the 30 unique synthetic phosphopeptides injected ETD, HCD and EThCD identified 21, 22 and 24, respectively. We found the average SEQUEST Xcorr being highest for EThcD (2.5) followed by HCD (1.9) and ETD (1.5), which is in line with our previous results for nonmodified peptides. (28) The SEQUEST algorithm correctly annotated the known phosphosites in 79% of ETD and 78% of HCD data. Significantly, for EThcD this was over 95% (of all PSMs), which directly reflects the higher spectral quality, due to the generation of both b/y and c/z ions. This initial data suggests that EThcD provides even more extensive backbone fragmentation of phosphorylated peptides than ETD or HCD alone, facilitating sensitive phosphosite localization with very high confidence. It should be noted that the application of a site localization algorithm would be prudent for real-life samples since the true phosphorylation sites are unknown.
Table 1. Analysis of 30 Synthetic Phosphopeptides
  ETD HCD EThcD
#PSM 216 237 248
# unique peptides 21/30 22/30 24/30
average Xcorr 1.5 1.9 2.5
% PSM with correctly localized phosphosite (SEQUEST) 79% 78% 95%
# phosphosites with phosphoRS site probability >99% 478 410 423
% phosphosites with phosphoRS site probability >99% 96% 95% 97%
Recently, Taus et al. described phosphoRS, a novel tool to improve confident localization of phosphosites. (27) The software is based on validated peptide identifications provided by database search engines and calculates site probabilities for each potential phosphosite in the peptide sequence. For this study we used a modified version of phosphoRS that also enables assessment of individual phosphosite probabilities for EThcD fragmentation. We analyzed each data set using phosphoRS and found that it performs equally well for all three fragmentation techniques. Of all true phosphosites, 96% (ETD), 95% (HCD) and 97% (EThcD) were assigned a site probability >99%, which corresponds to a very high confidence in site localization (Table 1). Together, these findings suggest that EThcD generates MS/MS spectra that contain sufficient fragment ions for the unambiguous and sensitive phosphorylation site localization.

Phosphosite Localization of Ti4+-IMAC Enriched Phosphopeptides by EThcD

Next, we assessed the performance of EThcD for phosphosite localization on a larger data set. We used Ti4+-IMAC material for the enrichment of phosphopeptides from a tryptic digest of HeLa cells and analyzed equal amounts (corresponding to enriched phosphopeptides from 100 μg of protein) by LC–MS/MS with ETD, HCD and EThcD, respectively (Supplementary Figure 1A, Supporting Information). All three methods generated a similar number of MS/MS spectra. All spectra were searched with SEQUEST. The ETD data was also searched with Mascot because we found SEQUEST to perform poorly for doubly charged phosphopeptides. Note that other search engines such as OMSSA or SpectrumMill might provide larger number of identifications for ETD data. (35) However, these algorithms are currently not compatible with EThcD data and phosphoRS analysis within the Proteome Discoverer software environment. All identified PSMs were then filtered for <1% FDR using percolator to ensure consistency. In total we identified 2217 (ETD), 4179 (HCD) and 3594 (EThcD) phospho-PSMs (Table 2). Our initial analysis of a defined synthetic phosphopeptide mixture demonstrated that EThcD performs at least on the same level as HCD in terms of peptide identification. However, the overall identification success rate in the Ti4+-IMAC data set was slightly lower for EThcD compared to HCD. This can be attributed to the rigid automatic FDR filtering. The MS/MS spectra from the synthetic phosphopeptide mixture were manually validated whereas the Ti4+-IMAC data set was computationally filtered to <1% FDR. The application of EThcD, in comparison to ETD or HCD alone, significantly increases the number of fragment ions observed in the MS/MS scans. On the one hand EThcD spectra contain more sequence information, which is beneficial for inferring the peptide sequence and PTM localization. On the other hand, these additional fragment ions may also match to random peptide sequences, increasing their score and hampering the differentiation between correct and incorrect matches. Consequently, the chance for a high scoring random match will be elevated. Similar to the increased average score of decoy hits also the true hits are likely to provide on average higher scores. Depending on whether the distance between the two score distributions decreases or increases, the identification success rate will be higher or lower. Since the ID success rate is slightly lower for EThcD compared to HCD alone, the negative effect of higher-scoring random matches might be more pronounced. Thus, higher score cut-offs need to be applied in order to reach the desired FDR. A standard target-decoy approach (36) against a reversed concatenated database revealed the FDR for EThcD (2.6%) being almost twice as high compared to HCD (1.4%), which provides further evidence for this hypothesis.
Table 2. LC–MS/MS Analysis of Ti4+-IMAC Enriched Tryptic Phosphopeptides Originating from a Cellular Lysate using ETD, HCD and EThcD
  ETD HCD EThcD
#PSM 2266 4282 3679
ID success rate 25% 51% 44%
average Xcorr 1.9 2.5 3.2
% average peptide sequence coverage 83% 81% 92%
# phospho-PSM 2217 4179 3594
# phospho-sites >99% pRS probability 2002 4291 3942
% phospho-sites >99% pRS probability 81% 89% 95%
Next, we calculated the average peptide sequence coverage for all PSM. As expected, EThcD provided a substantial increase in sequence coverage (92%) compared to HCD (81%) and ETD (83%). Obtaining near-complete peptide sequence coverage tremendously simplifies phosphosite localization. We used the extended phosphoRS algorithm to validate our assumption. Remarkably, EThcD provided for 95% of all phosphosites a confident site localization probability of >99%. In the HCD data set we found that 89% of all phosphosites were assigned with a confident site localization probability >99%, while this was only 81% for ETD data set. We recalculated these number for all peptides that contain >2 residues that can be phosphorylated because singly phosphorylated peptides with only one potential phosphorylation site could bias the results toward HCD. Of all phosphosites from this subset of peptides 97% (ETcaD), 93% (EThcD) and 87% (HCD), respectively, were assigned a localization probability >99%.
For multiply phosphorylated peptides site localization becomes more challenging. Figure 1 shows an MS/MS spectrum of a doubly phosphorylated peptide upon EThcD fragmentation. The overall sequence coverage is 89% taking b/y- and c/z-ions into account. Six out of 18 amino acid bond cleavages are represented by c- and b-ions (referred to as “golden pairs” (37)). Additionally, we observed 11 z/y-ion pairs, which strengthens the argument that EThcD provides extensive sequence information that facilitates pinpointing the correct phosphorylation site. More than 95% of the phosphosites from all doubly phosphorylated peptides were assigned with a site localization probability >99%, highlighting that EThcD performs equally well with singly and doubly phosphorylated peptides. A known limitation of ETD is its inability to cleave the N–Cα bond N-terminal to proline. (38, 39) This can hamper phosphosite localization for proline-rich peptides. Generation of dual ion series in EThcD can overcome this issue. Figure 2 shows the EThcD spectrum of a singly phosphorylated peptide that contains four serine residues. The c- and z-ions derived from the ETD step cover only the N-terminal part of the peptide and the site probability is only 50%. The additional y-ions derived from the subsequent HCD activation provide supporting sequence information and cover also the two serine residues next to the prolines which enables unambiguous phosphosite localization.

Figure 1

Figure 1. EThcD MS/MS spectrum of a doubly phosphorylated peptide. RGTGQSDDSDIWDDTALIK is doubly phosphorylated and contains in total four potential phosphorylation sites. EThcD generates dual ion series that enable phosphorylation site localization with very high confidence (phosphoRS site probabilities: T(3), 0.0%; S(6), 100.0%; S(9), 100.0%; T(15), 0.0%). SEQUEST Xcorr 7.79.

Figure 2

Figure 2. EThcD spectrum of a proline-containing phosphopeptide. This EThcD spectrum of a doubly charged peptide that contains four serine residues, one of which is phosphorylated. ETD does not cleave the N–Cα bond N-terminal to proline and the phosphorylation site probability is only 50% based on c- and z-ions alone. Dual fragmentation by EThcD generates complementary sequence information from c/z- and b/y-ions (SEQUEST Xcorr 4.10). Here, the exact phosphosite is revealed by y-ions that cover the corresponding phosphosite (phosphoRS site probabilitis: S(1): 0.0; S(3): 0.0; S(8): 99.5; S(10): 0.5). SEQUEST Xcorr 4.10.

Conclusions

ARTICLE SECTIONS
Jump To

Here we have evaluated the potential of EThcD in improving the analysis of phosphopeptides. Our data highlights the benefit of dual ion series as generated by EThcD fragmentation. We observed for a defined phosphopeptide mixture average higher SEQUEST Xcorr values, higher peptide sequence coverage and more confident phosphosite localization in EThcD compared to ETD and HCD. This finding was confirmed when we analyzed a complex phosphopeptide sample resulting from a Ti4+-IMAC enrichment of peptides from a cellular lysate. This is in line with recent reports that showed that confidence in phosphorylation site localization increases when multiple separately acquired MS/MS spectra (e.g., ETD/CID or MSA/ETD) are combined for scoring. (25, 26) For this larger data set, we observed that the identification success rate was slightly lower for EThcD compared to HCD. This can be attributed to the use of conventional database search engines that are not optimized for spectra that contain dual ion series. (40) However, the fact that both peptide sequence coverage and the percentage of localized phosphosites are higher for EThcD than for HCD suggests that once a peptide was identified, further analyses such as site localization benefit from the more data-rich EThcD spectra. In EThcD often c/b- and z/y-ion pairs are observed that increase the confidence in a particular peptide backbone cleavage. (41) We speculate that the identification success rate of EThcD for phosphopeptides can be improved by novel or optimized data analysis tools. Finally, we reason that EThcD can also be beneficial and used to improve the localization of other post-translational modifications such as ubiquitination, glycosylation or acetylation.

Supporting Information

ARTICLE SECTIONS
Jump To

Additional information as noted in the text. This material is available free of charge via the Internet at http://pubs.acs.org.

Terms & Conditions

Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Author Information

ARTICLE SECTIONS
Jump To

  • Corresponding Authors
    • Albert J. R. Heck - Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands.Netherlands Proteomics Centre, Padualaan 8, 3584 CH Utrecht, The Netherlands Email: [email protected] [email protected]
    • Shabaz Mohammed - Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands.Netherlands Proteomics Centre, Padualaan 8, 3584 CH Utrecht, The Netherlands Email: [email protected] [email protected]
  • Authors
    • Christian K. Frese - Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands.Netherlands Proteomics Centre, Padualaan 8, 3584 CH Utrecht, The Netherlands
    • Houjiang Zhou - Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands.Netherlands Proteomics Centre, Padualaan 8, 3584 CH Utrecht, The Netherlands
    • Thomas Taus - Research Institute of Molecular Pathology (IMP), Dr. Bohrgasse 7, A-1030 Vienna, Austria
    • A. F. Maarten Altelaar - Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands.Netherlands Proteomics Centre, Padualaan 8, 3584 CH Utrecht, The Netherlands
    • Karl Mechtler - Research Institute of Molecular Pathology (IMP), Dr. Bohrgasse 7, A-1030 Vienna, AustriaInstitute of Molecular Biotechnology (IMBA), Vienna, Austria
  • Notes
    The authors declare no competing financial interest.

Acknowledgment

ARTICLE SECTIONS
Jump To

We thank Mathias Madalinski for peptide synthesis of the primeXS phosphopeptide mixture. This research was performed within the framework of the PRIME-XS project, grant number 262067, funded by the European Union 7th Framework Program. Additionally, The Netherlands Proteomics Centre, a program embedded in The Netherlands Genomics Initiative, is kindly acknowledged for financial support as well as The Netherlands Organization for Scientific Research (NWO) with the VIDI grant (700.10.429). Work in the Mechtler lab was supported by the European Commission via the FP7 projects MeioSys and PRIME-XS, the Austrian Science Fund via the Special Research Program Chromosome Dynamics (SFB-F3402).

References

ARTICLE SECTIONS
Jump To

This article references 41 other publications.

  1. 1
    Cohen, P. The regulation of protein function by multisite phosphorylation--a 25 year update Trends Biochem. Sci. 2000, 25 (12) 596 601
  2. 2
    Hunter, T. Signaling--2000 and beyond Cell 2000, 100 (1) 113 27
  3. 3
    Olsen, J. V.; Blagoev, B.; Gnad, F.; Macek, B.; Kumar, C.; Mortensen, P.; Mann, M. Global, in vivo, and site-specific phosphorylation dynamics in signaling networks Cell 2006, 127 (3) 635 48
  4. 4
    Eyrich, B.; Sickmann, A.; Zahedi, R. P. Catch me if you can: mass spectrometry-based phosphoproteomics and quantification strategies Proteomics 2011, 11 (4) 554 70
  5. 5
    Mann, M.; Jensen, O. N. Proteomic analysis of post-translational modifications Nat. Biotechnol. 2003, 21 (3) 255 61
  6. 6
    Di Palma, S.; Hennrich, M. L.; Heck, A. J.; Mohammed, S. Recent advances in peptide separation by multidimensional liquid chromatography for proteome analysis J. Proteomics 2012, 75 (13) 3791 813
  7. 7
    Rush, J.; Moritz, A.; Lee, K. A.; Guo, A.; Goss, V. L.; Spek, E. J.; Zhang, H.; Zha, X. M.; Polakiewicz, R. D.; Comb, M. J. Immunoaffinity profiling of tyrosine phosphorylation in cancer cells Nat. Biotechnol. 2005, 23 (1) 94 101
  8. 8
    Ficarro, S. B.; McCleland, M. L.; Stukenberg, P. T.; Burke, D. J.; Ross, M. M.; Shabanowitz, J.; Hunt, D. F.; White, F. M. Phosphoproteome analysis by mass spectrometry and its application to Saccharomyces cerevisiae Nat. Biotechnol. 2002, 20 (3) 301 5
  9. 9
    Zhou, H.; Low, T. Y.; Hennrich, M. L.; van der Toorn, H.; Schwend, T.; Zou, H.; Mohammed, S.; Heck, A. J. Enhancing the identification of phosphopeptides from putative basophilic kinase substrates using Ti (IV) based IMAC enrichment Mol. Cell. Proteomics 2011, 10 (10) M110 006452
  10. 10
    Boersema, P. J.; Mohammed, S.; Heck, A. J. Phosphopeptide fragmentation and analysis by mass spectrometry J. Mass Spectrom. 2009, 44 (6) 861 78
  11. 11
    Beausoleil, S. A.; Jedrychowski, M.; Schwartz, D.; Elias, J. E.; Villen, J.; Li, J.; Cohn, M. A.; Cantley, L. C.; Gygi, S. P. Large-scale characterization of HeLa cell nuclear phosphoproteins Proc. Natl. Acad. Sci. U.S.A. 2004, 101 (33) 12130 5
  12. 12
    Schroeder, M. J.; Shabanowitz, J.; Schwartz, J. C.; Hunt, D. F.; Coon, J. J. A neutral loss activation method for improved phosphopeptide sequence analysis by quadrupole ion trap mass spectrometry Anal. Chem. 2004, 76 (13) 3590 8
  13. 13
    Olsen, J. V.; Macek, B.; Lange, O.; Makarov, A.; Horning, S.; Mann, M. Higher-energy C-trap dissociation for peptide modification analysis Nat. Methods 2007, 4 (9) 709 12
  14. 14
    Chi, A.; Huttenhower, C.; Geer, L. Y.; Coon, J. J.; Syka, J. E.; Bai, D. L.; Shabanowitz, J.; Burke, D. J.; Troyanskaya, O. G.; Hunt, D. F. Analysis of phosphorylation sites on proteins from Saccharomyces cerevisiae by electron transfer dissociation (ETD) mass spectrometry Proc. Natl. Acad. Sci. U.S.A. 2007, 104 (7) 2193 8
  15. 15
    Molina, H.; Matthiesen, R.; Kandasamy, K.; Pandey, A. Comprehensive comparison of collision induced dissociation and electron transfer dissociation Anal. Chem. 2008, 80 (13) 4825 35
  16. 16
    Beausoleil, S. A.; Villen, J.; Gerber, S. A.; Rush, J.; Gygi, S. P. A probability-based approach for high-throughput protein phosphorylation analysis and site localization Nat. Biotechnol. 2006, 24 (10) 1285 92
  17. 17
    Stensballe, A.; Jensen, O. N.; Olsen, J. V.; Haselmann, K. F.; Zubarev, R. A. Electron capture dissociation of singly and multiply phosphorylated peptides Rapid Commun. Mass Spectrom. 2000, 14 (19) 1793 800
  18. 18
    Kelstrup, C. D.; Hekmat, O.; Francavilla, C.; Olsen, J. V. Pinpointing phosphorylation sites: Quantitative filtering and a novel site-specific x-ion fragment J. Proteome Res. 2011, 10 (7) 2937 48
  19. 19
    Shin, Y. S.; Moon, J. H.; Kim, M. S. Observation of phosphorylation site-specific dissociation of singly protonated phosphopeptides J. Am. Soc. Mass Spectrom. 2010, 21 (1) 53 9
  20. 20
    Gehrig, P. M.; Roschitzki, B.; Rutishauser, D.; Reiland, S.; Schlapbach, R. Phosphorylated serine and threonine residues promote site-specific fragmentation of singly charged, arginine-containing peptide ions Rapid Commun. Mass Spectrom. 2009, 23 (10) 1435 45
  21. 21
    Lu, B.; Ruse, C.; Xu, T.; Park, S. K.; Yates, J. R., 3rd Automatic validation of phosphopeptide identifications from tandem mass spectra Anal. Chem. 2007, 79 (4) 1301 10
  22. 22
    Bailey, C. M.; Sweet, S. M.; Cunningham, D. L.; Zeller, M.; Heath, J. K.; Cooper, H. J. SLoMo: automated site localization of modifications from ETD/ECD mass spectra J. Proteome Res. 2009, 8 (4) 1965 71
  23. 23
    Savitski, M. M.; Mathieson, T.; Becher, I.; Bantscheff, M. H-score, a mass accuracy driven rescoring approach for improved peptide identification in modification rich samples J. Proteome Res. 2010, 9 (11) 5511 6
  24. 24
    Ruttenberg, B. E.; Pisitkun, T.; Knepper, M. A.; Hoffert, J. D. PhosphoScore: an open-source phosphorylation site assignment tool for MSn data Journal of Proteome Research 2008, 7 (7) 3054 9
  25. 25
    Hansen, T. A.; Sylvester, M.; Jensen, O. N.; Kjeldsen, F. Automated and high confidence protein phosphorylation site localization using complementary collision-activated dissociation and electron transfer dissociation tandem mass spectrometry Anal. Chem. 2012, 84 (22) 9694 9
  26. 26
    Vandenbogaert, M.; Hourdel, V.; Jardin-Mathe, O.; Bigeard, J.; Bonhomme, L.; Legros, V.; Hirt, H.; Schwikowski, B.; Pflieger, D. Automated phosphopeptide identification using multiple MS/MS fragmentation modes J. Proteome Res. 2012, 11 (12) 5695 703
  27. 27
    Taus, T.; Kocher, T.; Pichler, P.; Paschke, C.; Schmidt, A.; Henrich, C.; Mechtler, K. Universal and confident phosphorylation site localization using phosphoRS J. Proteome Res. 2011, 10 (12) 5354 62
  28. 28
    Frese, C. K.; Altelaar, M.; van den Toorn, H. W.; Nolting, D.; Griep-Raming, J.; Heck, A. J.; Mohammed, S. Toward full peptide sequence coverage by dual fragmentation combining electron-transfer and higher-energy collision dissociation tandem mass spectrometry Anal. Chem. 2012, 84 (22) 9668 73
  29. 29
    Altelaar, A. F.; Frese, C. K.; Preisinger, C.; Hennrich, M. L.; Schram, A. W.; Timmers, H. T.; Heck, A. J.; Mohammed, S. Benchmarking stable isotope labeling based quantitative proteomics J. Proteomics 2012,  DOI: 10.1016/j.jprot.2012.10.009
  30. 30
    Zhou, H.; Ye, M.; Dong, J.; Han, G.; Jiang, X.; Wu, R.; Zou, H. Specific phosphopeptide enrichment with immobilized titanium ion affinity chromatography adsorbent for phosphoproteome analysis J. Proteome Res. 2008, 7 (9) 3957 67
  31. 31
    Yu, Z.; Han, G.; Sun, S.; Jiang, X.; Chen, R.; Wang, F.; Wu, R.; Ye, M.; Zou, H. Preparation of monodisperse immobilized Ti(4+) affinity chromatography microspheres for specific enrichment of phosphopeptides Anal. Chim. Acta 2009, 636 (1) 34 41
  32. 32
    Zhou, H.; Ye, M.; Dong, J.; Corradini, E.; Cristobal, A.; Heck, A. J. R.; Zou, H.; Mohammed, S. Robust phosphoproteome enrichment using monodisperse microspheres-based immobilized titanium (IV) ion affinity chromatography. Nat. Protoc. 2012, accepted
  33. 33
    Kall, L.; Canterbury, J. D.; Weston, J.; Noble, W. S.; MacCoss, M. J. Semi-supervised learning for peptide identification from shotgun proteomics datasets Nat. Methods 2007, 4 (11) 923 5
  34. 34
    Mertins, P.; Udeshi, N. D.; Clauser, K. R.; Mani, D. R.; Patel, J.; Ong, S. E.; Jaffe, J. D.; Carr, S. A. iTRAQ labeling is superior to mTRAQ for quantitative global proteomics and phosphoproteomics Mol. Cell. Proteomics 2012, 11 (6) M111 014423
  35. 35
    Kandasamy, K.; Pandey, A.; Molina, H. Evaluation of Several MS/MS Search Algorithms for Analysis of Spectra Derived from Electron Transfer Dissociation Experiments Anal. Chem. 2009, 81 (17) 7170 80
  36. 36
    Elias, J. E.; Gygi, S. P. Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry Nat. Methods 2007, 4 (3) 207 14
  37. 37
    Horn, D. M.; Zubarev, R. A.; McLafferty, F. W. Automated de novo sequencing of proteins by tandem high-resolution mass spectrometry Proc. Natl. Acad. Sci. U.S.A. 2000, 97 (19) 10313 7
  38. 38
    Cooper, H. J.; Hudgins, R. R.; Hakansson, K.; Marshall, A. G. Secondary fragmentation of linear peptides in electron capture dissociation Int. J. Mass Spectrom. 2003, 228 (2–3) 723 8
  39. 39
    Li, W.; Song, C.; Bailey, D. J.; Tseng, G. C.; Coon, J. J.; Wysocki, V. H. Statistical analysis of electron transfer dissociation pairwise fragmentation patterns Anal. Chem. 2011, 83 (24) 9540 5
  40. 40
    Kim, M. S.; Zhong, J.; Kandasamy, K.; Delanghe, B.; Pandey, A. Systematic evaluation of alternating CID and ETD fragmentation for phosphorylated peptides Proteomics 2011, 11 (12) 2568 72
  41. 41
    Nielsen, M. L.; Savitski, M. M.; Zubarev, R. A. Improving protein identification using complementary fragmentation techniques in fourier transform mass spectrometry Mol. Cell. Proteomics 2005, 4 (6) 835 45

Cited By

ARTICLE SECTIONS
Jump To

This article is cited by 136 publications.

  1. Yuming Jiang, Devasahayam Arokia Balaya Rex, Dina Schuster, Benjamin A. Neely, Germán L. Rosano, Norbert Volkmar, Amanda Momenzadeh, Trenton M. Peters-Clarke, Susan B. Egbert, Simion Kreimer, Emma H. Doud, Oliver M. Crook, Amit Kumar Yadav, Muralidharan Vanuopadath, Adrian D. Hegeman, Martín L. Mayta, Anna G. Duboff, Nicholas M. Riley, Robert L. Moritz, Jesse G. Meyer. Comprehensive Overview of Bottom-Up Proteomics Using Mass Spectrometry. ACS Measurement Science Au 2024, Article ASAP.
  2. Trenton M. Peters-Clarke, Joshua J. Coon, Nicholas M. Riley. Instrumentation at the Leading Edge of Proteomics. Analytical Chemistry 2024, 96 (20) , 7976-8010. https://doi.org/10.1021/acs.analchem.3c04497
  3. Allen Po, Claire E. Eyers. Top-Down Proteomics and the Challenges of True Proteoform Characterization. Journal of Proteome Research 2023, 22 (12) , 3663-3675. https://doi.org/10.1021/acs.jproteome.3c00416
  4. Xiaojuan Li, Robert Wilmanowski, Xinliu Gao, Zachary L. VanAernum, Daniel P. Donnelly, Brent Kochert, Hillary A. Schuessler, Douglas Richardson. Precise O-Glycosylation Site Localization of CD24Fc by LC-MS Workflows. Analytical Chemistry 2022, 94 (23) , 8416-8425. https://doi.org/10.1021/acs.analchem.2c01137
  5. Trenton M. Peters-Clarke, Nicholas M. Riley, Michael S. Westphall, Joshua J. Coon. Practical Effects of Intramolecular Hydrogen Rearrangement in Electron Transfer Dissociation-Based Proteomics. Journal of the American Society for Mass Spectrometry 2022, 33 (1) , 100-110. https://doi.org/10.1021/jasms.1c00284
  6. Kailin Zhang, Yingying Shi, Mengying Du, Yicheng Xu, Yan Wang, Xianglei Kong. Versatile Double-Beam Confocal Laser System Combined with a Fourier Transform Ion Cyclotron Resonance Mass Spectrometer for Photodissociation Mass Spectrometry and Spectroscopy. Analytical Chemistry 2021, 93 (26) , 9056-9063. https://doi.org/10.1021/acs.analchem.1c00248
  7. Weiwei Peng, Matti F. Pronker, Joost Snijder. Mass Spectrometry-Based De Novo Sequencing of Monoclonal Antibodies Using Multiple Proteases and a Dual Fragmentation Scheme. Journal of Proteome Research 2021, 20 (7) , 3559-3566. https://doi.org/10.1021/acs.jproteome.1c00169
  8. Tobias P. Wörner, Tatiana M. Shamorkina, Joost Snijder, Albert J. R. Heck. Mass Spectrometry-Based Structural Virology. Analytical Chemistry 2021, 93 (1) , 620-640. https://doi.org/10.1021/acs.analchem.0c04339
  9. Dongxia Wang, Jakub Baudys, Jonathan L. Bundy, Maria Solano, Theodore Keppel, John R. Barr. Comprehensive Analysis of the Glycan Complement of SARS-CoV-2 Spike Proteins Using Signature Ions-Triggered Electron-Transfer/Higher-Energy Collisional Dissociation (EThcD) Mass Spectrometry. Analytical Chemistry 2020, 92 (21) , 14730-14739. https://doi.org/10.1021/acs.analchem.0c03301
  10. Jean M. Lodge, Kevin L. Schauer, Dain R. Brademan, Nicholas M. Riley, Evgenia Shishkova, Michael S. Westphall, Joshua J. Coon. Top-Down Characterization of an Intact Monoclonal Antibody Using Activated Ion Electron Transfer Dissociation. Analytical Chemistry 2020, 92 (15) , 10246-10251. https://doi.org/10.1021/acs.analchem.0c00705
  11. Jennifer S. Brodbelt, Lindsay J. Morrison, Inês Santos. Ultraviolet Photodissociation Mass Spectrometry for Analysis of Biological Molecules. Chemical Reviews 2020, 120 (7) , 3328-3380. https://doi.org/10.1021/acs.chemrev.9b00440
  12. Sean R. Kundinger, Isaac Bishof, Eric B. Dammer, Duc M. Duong, Nicholas T. Seyfried. Middle-Down Proteomics Reveals Dense Sites of Methylation and Phosphorylation in Arginine-Rich RNA-Binding Proteins. Journal of Proteome Research 2020, 19 (4) , 1574-1591. https://doi.org/10.1021/acs.jproteome.9b00633
  13. Arnold Steckel, Katalin Uray, Gergo Kalló, Éva Csosz, Gitta Schlosser. Investigation of Neutral Losses and the Citrulline Effect for Modified H4 N-Terminal Pentapeptides. Journal of the American Society for Mass Spectrometry 2020, 31 (3) , 565-573. https://doi.org/10.1021/jasms.9b00036
  14. Suttipong Suttapitugsakul, Fangxu Sun, Ronghu Wu. Recent Advances in Glycoproteomic Analysis by Mass Spectrometry. Analytical Chemistry 2020, 92 (1) , 267-291. https://doi.org/10.1021/acs.analchem.9b04651
  15. Martin Penkert, Anett Hauser, Robert Harmel, Dorothea Fiedler, Christian P. R. Hackenberger, Eberhard Krause. Electron Transfer/Higher Energy Collisional Dissociation of Doubly Charged Peptide Ions: Identification of Labile Protein Phosphorylations. Journal of the American Society for Mass Spectrometry 2019, 30 (9) , 1578-1585. https://doi.org/10.1007/s13361-019-02240-4
  16. Clement M. Potel, Miao-Hsia Lin, Nadine Prust, Henk W. P. van den Toorn, Albert J. R. Heck, Simone Lemeer. Gaining Confidence in the Elusive Histidine Phosphoproteome. Analytical Chemistry 2019, 91 (9) , 5542-5547. https://doi.org/10.1021/acs.analchem.9b00734
  17. Marialaura Dilillo, Erik L. de Graaf, Avinash Yadav, Mikhail E. Belov, Liam A. McDonnell. Ultraviolet Photodissociation of ESI- and MALDI-Generated Protein Ions on a Q-Exactive Mass Spectrometer. Journal of Proteome Research 2019, 18 (1) , 557-564. https://doi.org/10.1021/acs.jproteome.8b00896
  18. Clement M. Potel, Simone Lemeer, Albert J. R. Heck. Phosphopeptide Fragmentation and Site Localization by Mass Spectrometry: An Update. Analytical Chemistry 2019, 91 (1) , 126-141. https://doi.org/10.1021/acs.analchem.8b04746
  19. Jared B. Shaw, Neha Malhan, Yury V. Vasil’ev, Nathan I. Lopez, Alexander Makarov, Joseph S. Beckman, Valery G. Voinov. Sequencing Grade Tandem Mass Spectrometry for Top–Down Proteomics Using Hybrid Electron Capture Dissociation Methods in a Benchtop Orbitrap Mass Spectrometer. Analytical Chemistry 2018, 90 (18) , 10819-10827. https://doi.org/10.1021/acs.analchem.8b01901
  20. M. Montana Quick, Christopher M. Crittenden, Jake A. Rosenberg, Jennifer S. Brodbelt. Characterization of Disulfide Linkages in Proteins by 193 nm Ultraviolet Photodissociation (UVPD) Mass Spectrometry. Analytical Chemistry 2018, 90 (14) , 8523-8530. https://doi.org/10.1021/acs.analchem.8b01556
  21. Kshitij Khatri, Yi Pu, Joshua A. Klein, Juan Wei, Catherine E. Costello, Cheng Lin, Joseph Zaia. Comparison of Collisional and Electron-Based Dissociation Modes for Middle-Down Analysis of Multiply Glycosylated Peptides. Journal of the American Society for Mass Spectrometry 2018, 29 (6) , 1075-1085. https://doi.org/10.1007/s13361-018-1909-y
  22. Adam Pap, Eva Klement, Eva Hunyadi-Gulyas, Zsuzsanna Darula, Katalin F. Medzihradszky. Status Report on the High-Throughput Characterization of Complex Intact O-Glycopeptide Mixtures. Journal of the American Society for Mass Spectrometry 2018, 29 (6) , 1210-1220. https://doi.org/10.1007/s13361-018-1945-7
  23. Fengfei Ma, Ruixiang Sun, Daniel M. Tremmel, Sara Dutton Sackett, Jon Odorico, Lingjun Li. Large-Scale Differentiation and Site Specific Discrimination of Hydroxyproline Isomers by Electron Transfer/Higher-Energy Collision Dissociation (EThcD) Mass Spectrometry. Analytical Chemistry 2018, 90 (9) , 5857-5864. https://doi.org/10.1021/acs.analchem.8b00413
  24. Matthew J. Berberich, Joao A. Paulo, Robert A. Everley. MS3-IDQ: Utilizing MS3 Spectra beyond Quantification Yields Increased Coverage of the Phosphoproteome in Isobaric Tag Experiments. Journal of Proteome Research 2018, 17 (4) , 1741-1747. https://doi.org/10.1021/acs.jproteome.8b00006
  25. Alyssa Garabedian, Matthew A. Baird, Jacob Porter, Kevin Jeanne Dit Fouque, Pavel V. Shliaha, Ole N. Jensen, Todd D. Williams, Francisco Fernandez-Lima, and Alexandre A. Shvartsburg . Linear and Differential Ion Mobility Separations of Middle-Down Proteoforms. Analytical Chemistry 2018, 90 (4) , 2918-2925. https://doi.org/10.1021/acs.analchem.7b05224
  26. Caiping Tian, Keke Liu, Rui Sun, Ling Fu, and Jing Yang . Chemoproteomics Reveals Unexpected Lysine/Arginine-Specific Cleavage of Peptide Chains as a Potential Protein Degradation Machinery. Analytical Chemistry 2018, 90 (1) , 794-800. https://doi.org/10.1021/acs.analchem.7b03237
  27. Nicholas M. Riley and Joshua J. Coon . The Role of Electron Transfer Dissociation in Modern Proteomics. Analytical Chemistry 2018, 90 (1) , 40-64. https://doi.org/10.1021/acs.analchem.7b04810
  28. Daniel S. Ziemianowicz, Ryan Bomgarden, Chris Etienne, David C. Schriemer. Amino Acid Insertion Frequencies Arising from Photoproducts Generated Using Aliphatic Diazirines. Journal of the American Society for Mass Spectrometry 2017, 28 (10) , 2011-2021. https://doi.org/10.1007/s13361-017-1730-z
  29. Qing Yu, Bowen Wang, Zhengwei Chen, Go Urabe, Matthew S. Glover, Xudong Shi, Lian-Wang Guo, K. Craig Kent, Lingjun Li. Electron-Transfer/Higher-Energy Collision Dissociation (EThcD)-Enabled Intact Glycopeptide/Glycoproteome Characterization. Journal of the American Society for Mass Spectrometry 2017, 28 (9) , 1751-1764. https://doi.org/10.1007/s13361-017-1701-4
  30. Samantha Ferries, Simon Perkins, Philip J. Brownridge, Amy Campbell, Patrick A. Eyers, Andrew R. Jones, and Claire E. Eyers . Evaluation of Parameters for Confident Phosphorylation Site Localization Using an Orbitrap Fusion Tribrid Mass Spectrometer. Journal of Proteome Research 2017, 16 (9) , 3448-3459. https://doi.org/10.1021/acs.jproteome.7b00337
  31. Anett Hauser, Martin Penkert, and Christian P. R. Hackenberger . Chemical Approaches to Investigate Labile Peptide and Protein Phosphorylation. Accounts of Chemical Research 2017, 50 (8) , 1883-1893. https://doi.org/10.1021/acs.accounts.7b00170
  32. Nicholas M. Riley, Alexander S. Hebert, Gerhard Dürnberger, Florian Stanek, Karl Mechtler, Michael S. Westphall, and Joshua J. Coon . Phosphoproteomics with Activated Ion Electron Transfer Dissociation. Analytical Chemistry 2017, 89 (12) , 6367-6376. https://doi.org/10.1021/acs.analchem.7b00212
  33. Nicholas M. Riley, Michael S. Westphall, Alexander S. Hebert, and Joshua J. Coon . Implementation of Activated Ion Electron Transfer Dissociation on a Quadrupole-Orbitrap-Linear Ion Trap Hybrid Mass Spectrometer. Analytical Chemistry 2017, 89 (12) , 6358-6366. https://doi.org/10.1021/acs.analchem.7b00213
  34. Alba Cristobal, Fabio Marino, Harm Post, Henk W. P. van den Toorn, Shabaz Mohammed, and Albert J. R. Heck . Toward an Optimized Workflow for Middle-Down Proteomics. Analytical Chemistry 2017, 89 (6) , 3318-3325. https://doi.org/10.1021/acs.analchem.6b03756
  35. Martin Penkert, Lisa M. Yates, Michael Schümann, David Perlman, Dorothea Fiedler, and Eberhard Krause . Unambiguous Identification of Serine and Threonine Pyrophosphorylation Using Neutral-Loss-Triggered Electron-Transfer/Higher-Energy Collision Dissociation. Analytical Chemistry 2017, 89 (6) , 3672-3680. https://doi.org/10.1021/acs.analchem.6b05095
  36. Vera Bilan, Mario Leutert, Paolo Nanni, Christian Panse, and Michael O. Hottiger . Combining Higher-Energy Collision Dissociation and Electron-Transfer/Higher-Energy Collision Dissociation Fragmentation in a Product-Dependent Manner Confidently Assigns Proteomewide ADP-Ribose Acceptor Sites. Analytical Chemistry 2017, 89 (3) , 1523-1530. https://doi.org/10.1021/acs.analchem.6b03365
  37. Rijing Liao, Dan Zheng, Aiying Nie, Shaolian Zhou, Haibing Deng, Yuan Gao, Pengyuan Yang, Yanyan Yu, Lin Tan, Wei Qi, Jiaxi Wu, En Li, and Wei Yi . Sensitive and Precise Characterization of Combinatorial Histone Modifications by Selective Derivatization Coupled with RPLC-EThcD-MS/MS. Journal of Proteome Research 2017, 16 (2) , 780-787. https://doi.org/10.1021/acs.jproteome.6b00788
  38. Fabio Marino, Geert P. M. Mommen, Anita Jeko, Hugo D. Meiring, Jacqueline A. M. van Gaans-van den Brink, Richard A. Scheltema, Cécile A. C. M. van Els, and Albert J. R. Heck . Arginine (Di)methylated Human Leukocyte Antigen Class I Peptides Are Favorably Presented by HLA-B*07. Journal of Proteome Research 2017, 16 (1) , 34-44. https://doi.org/10.1021/acs.jproteome.6b00528
  39. Victoria C. Cotham, William M. McGee, and Jennifer S. Brodbelt . Modulation of Phosphopeptide Fragmentation via Dual Spray Ion/Ion Reactions Using a Sulfonate-Incorporating Reagent. Analytical Chemistry 2016, 88 (16) , 8158-8165. https://doi.org/10.1021/acs.analchem.6b01901
  40. Sven H. Giese, Adam Belsom, and Juri Rappsilber . Optimized Fragmentation Regime for Diazirine Photo-Cross-Linked Peptides. Analytical Chemistry 2016, 88 (16) , 8239-8247. https://doi.org/10.1021/acs.analchem.6b02082
  41. Michelle R. Robinson, Juliana M. Taliaferro, Kevin N. Dalby, and Jennifer S. Brodbelt . 193 nm Ultraviolet Photodissociation Mass Spectrometry for Phosphopeptide Characterization in the Positive and Negative Ion Modes. Journal of Proteome Research 2016, 15 (8) , 2739-2748. https://doi.org/10.1021/acs.jproteome.6b00289
  42. Kyle L. Fort, Andrey Dyachenko, Clement M. Potel, Eleonora Corradini, Fabio Marino, Arjan Barendregt, Alexander A. Makarov, Richard A. Scheltema, Albert J. R. Heck. Implementation of Ultraviolet Photodissociation on a Benchtop Q Exactive Mass Spectrometer and Its Application to Phosphoproteomics. Analytical Chemistry 2016, 88 (4) , 2303-2310. https://doi.org/10.1021/acs.analchem.5b04162
  43. Nicholas M. Riley and Joshua J. Coon . Phosphoproteomics in the Age of Rapid and Deep Proteome Profiling. Analytical Chemistry 2016, 88 (1) , 74-94. https://doi.org/10.1021/acs.analchem.5b04123
  44. Jennifer S. Brodbelt . Ion Activation Methods for Peptides and Proteins. Analytical Chemistry 2016, 88 (1) , 30-51. https://doi.org/10.1021/acs.analchem.5b04563
  45. Christopher M. Rose, Matthew J. P. Rush, Nicholas M. Riley, Anna E. Merrill, Nicholas W. Kwiecien, Dustin D. Holden, Christopher Mullen, Michael S. Westphall, Joshua J. Coon. A Calibration Routine for Efficient ETD in Large-Scale Proteomics. Journal of the American Society for Mass Spectrometry 2015, 26 (11) , 1848-1857. https://doi.org/10.1007/s13361-015-1183-1
  46. Fabio Marino, Marshall Bern, Geert P. M. Mommen, Aneika C. Leney, Jacqueline A. M. van Gaans-van den Brink, Alexandre M. J. J. Bonvin, Christopher Becker, Cécile A. C. M. van Els, and Albert J. R. Heck . Extended O-GlcNAc on HLA Class-I-Bound Peptides. Journal of the American Chemical Society 2015, 137 (34) , 10922-10925. https://doi.org/10.1021/jacs.5b06586
  47. Scott A. Robotham, Jennifer S. Brodbelt. Comparison of Ultraviolet Photodissociation and Collision Induced Dissociation of Adrenocorticotropic Hormone Peptides. Journal of the American Society for Mass Spectrometry 2015, 26 (9) , 1570-1579. https://doi.org/10.1007/s13361-015-1186-y
  48. Andrea M. Brunner, Philip Lössl, Fan Liu, Romain Huguet, Christopher Mullen, Masami Yamashita, Vlad Zabrouskov, Alexander Makarov, A. F. Maarten Altelaar, and Albert J. R. Heck . Benchmarking Multiple Fragmentation Methods on an Orbitrap Fusion for Top-down Phospho-Proteoform Characterization. Analytical Chemistry 2015, 87 (8) , 4152-4158. https://doi.org/10.1021/acs.analchem.5b00162
  49. Nadine A. Binai, Fabio Marino, Peter Soendergaard, Nicolai Bache, Shabaz Mohammed, and Albert J. R. Heck . Rapid Analyses of Proteomes and Interactomes Using an Integrated Solid-Phase Extraction–Liquid Chromatography–MS/MS System. Journal of Proteome Research 2015, 14 (2) , 977-985. https://doi.org/10.1021/pr501011z
  50. Joe R. Cannon, Dustin D. Holden, and Jennifer S. Brodbelt . Hybridizing Ultraviolet Photodissociation with Electron Transfer Dissociation for Intact Protein Characterization. Analytical Chemistry 2014, 86 (21) , 10970-10977. https://doi.org/10.1021/ac5036082
  51. Mostafa Zarei, Adrian Sprenger, Christine Gretzmeier, and Joern Dengjel . Rapid Combinatorial ERLIC–SCX Solid-Phase Extraction for In-Depth Phosphoproteome Analysis. Journal of Proteome Research 2013, 12 (12) , 5989-5995. https://doi.org/10.1021/pr4007969
  52. Ruben Shrestha, Sumudu Karunadasa, TaraBryn S. Grismer, Andres V. Reyes, Shou-Ling Xu. SECRET AGENT O-GlcNAcylates Hundreds of Proteins Involved in Diverse Cellular Processes in Arabidopsis. Molecular & Cellular Proteomics 2024, 23 (4) , 100732. https://doi.org/10.1016/j.mcpro.2024.100732
  53. Irina D. Vasileva, Tatiana Yu Samgina, Albert T. Lebedev. Mass Spectrometric De Novo Sequencing of Natural Peptides. 2024, 61-75. https://doi.org/10.1007/978-1-0716-3646-6_3
  54. Leonard A. Daly, Christopher J. Clarke, Allen Po, Sally O. Oswald, Claire E. Eyers. Considerations for defining +80 Da mass shifts in mass spectrometry-based proteomics: phosphorylation and beyond. Chemical Communications 2023, 59 (77) , 11484-11499. https://doi.org/10.1039/D3CC02909C
  55. Martin Toul, Veronika Slonkova, Jan Mican, Adam Urminsky, Maria Tomkova, Erik Sedlak, David Bednar, Jiri Damborsky, Lenka Hernychova, Zbynek Prokop. Identification, characterization, and engineering of glycosylation in thrombolytics. Biotechnology Advances 2023, 66 , 108174. https://doi.org/10.1016/j.biotechadv.2023.108174
  56. Ruijie Liu, Shujun Xia, Huilin Li. Native top‐down mass spectrometry for higher‐order structural characterization of proteins and complexes. Mass Spectrometry Reviews 2023, 42 (5) , 1876-1926. https://doi.org/10.1002/mas.21793
  57. Yi Yang, Liang Qiao. Data‐independent acquisition proteomics methods for analyzing post‐translational modifications. PROTEOMICS 2023, 23 (7-8) https://doi.org/10.1002/pmic.202200046
  58. Yuanli Song, Jing Gao, Qian Meng, Feng Tang, Yuqiu Wang, Yue Zeng, Wei Huang, Hong Shao, Hu Zhou. Conjugation site characterization of antibody–drug conjugates using electron-transfer/higher-energy collision dissociation (EThcD). Analytica Chimica Acta 2023, 1251 , 340978. https://doi.org/10.1016/j.aca.2023.340978
  59. Jasmin Adriana Schäfer, F.X. Reymond Sutandy, Christian Münch. Omics-based approaches for the systematic profiling of mitochondrial biology. Molecular Cell 2023, 83 (6) , 911-926. https://doi.org/10.1016/j.molcel.2023.02.015
  60. Taewook Kang, Santosh Bhosale, Alistair Edwards, Martin R. Larsen. Phosphoproteomics: Methods and Challenges. 2023, 417-429. https://doi.org/10.1016/B978-0-12-821618-7.00031-6
  61. Xiaoyin Zeng, Yanting Lan, Jing Xiao, Longbo Hu, Long Tan, Mengdi Liang, Xufei Wang, Shaohua Lu, Tao Peng, Fei Long. Advances in phosphoproteomics and its application to COPD. Expert Review of Proteomics 2022, 19 (7-12) , 311-324. https://doi.org/10.1080/14789450.2023.2176756
  62. Sander R. Piersma, Andrea Valles‐Marti, Frank Rolfs, Thang V. Pham, Alex A. Henneman, Connie R. Jiménez. Inferring kinase activity from phosphoproteomic data: Tool comparison and recent applications. Mass Spectrometry Reviews 2022, 48 https://doi.org/10.1002/mas.21808
  63. Yujie Gan, Huanhuan Sha, Renrui Zou, Miao Xu, Yuan Zhang, Jifeng Feng, Jianzhong Wu. Research Progress on Mono-ADP-Ribosyltransferases in Human Cell Biology. Frontiers in Cell and Developmental Biology 2022, 10 https://doi.org/10.3389/fcell.2022.864101
  64. Min Zhou, Luyang Jiao, Shiyin Xu, Yicheng Xu, Mengying Du, Xianyi Zhang, Xianglei Kong. A novel method for photon unfolding spectroscopy of protein ions in the gas phase. Review of Scientific Instruments 2022, 93 (4) https://doi.org/10.1063/5.0080040
  65. Paul J. Hensbergen, Arnoud H. de Ru, Annemieke H. Friggen, Jeroen Corver, Wiep Klaas Smits, Peter A. van Veelen. New insights into the type A glycan modification of Clostridioides difficile flagellar protein flagellin C by phosphoproteomics analysis. Journal of Biological Chemistry 2022, 298 (3) , 101622. https://doi.org/10.1016/j.jbc.2022.101622
  66. Wiep Klaas Smits, Yassene Mohammed, Arnoud H. de Ru, Valentina Cordo', Annemieke H. Friggen, Peter A. van Veelen, Paul J. Hensbergen, . Clostridioides difficile Phosphoproteomics Shows an Expansion of Phosphorylated Proteins in Stationary Growth Phase. mSphere 2022, 7 (1) https://doi.org/10.1128/msphere.00911-21
  67. Luyao LIU, Hongqiang QIN, Mingliang YE. Recent advances in glycopeptide enrichment and mass spectrometry data interpretation approaches for glycoproteomics analyses. Chinese Journal of Chromatography 2021, 39 (10) , 1045-1054. https://doi.org/10.3724/SP.J.1123.2021.06011
  68. Xinyue Liu, Rose Fields, Devin K. Schweppe, Joao A. Paulo. Strategies for mass spectrometry-based phosphoproteomics using isobaric tagging. Expert Review of Proteomics 2021, 18 (9) , 795-807. https://doi.org/10.1080/14789450.2021.1994390
  69. Joao A. Paulo, Devin K. Schweppe. Advances in quantitative high‐throughput phosphoproteomics with sample multiplexing. PROTEOMICS 2021, 21 (9) https://doi.org/10.1002/pmic.202000140
  70. Katie Dunphy, Paul Dowling, Despina Bazou, Peter O’Gorman. Current Methods of Post-Translational Modification Analysis and Their Applications in Blood Cancers. Cancers 2021, 13 (8) , 1930. https://doi.org/10.3390/cancers13081930
  71. Jun Sun, Yaoyao Mu, Tengmei Liu, Hui Jing, Mohammed Obadi, Yanjun Yang, Shijian Dong, Bin Xu. Evaluation of glycation reaction of ovalbumin with dextran: Glycation sites identification by capillary liquid chromatography coupled with tandem mass spectrometry. Food Chemistry 2021, 341 , 128066. https://doi.org/10.1016/j.foodchem.2020.128066
  72. Adina Borbély, Lilla Pethő, Ildikó Szabó, Mohammed Al-Majidi, Arnold Steckel, Tibor Nagy, Sándor Kéki, Gergő Kalló, Éva Csősz, Gábor Mező, Gitta Schlosser. Structural Characterization of Daunomycin-Peptide Conjugates by Various Tandem Mass Spectrometric Techniques. International Journal of Molecular Sciences 2021, 22 (4) , 1648. https://doi.org/10.3390/ijms22041648
  73. Suruchi Aggarwal, Priya Tolani, Srishti Gupta, Amit Kumar Yadav. Posttranslational modifications in systems biology. 2021, 93-126. https://doi.org/10.1016/bs.apcsb.2021.03.005
  74. Zhengwei Chen, Qinying Yu, Qing Yu, Jillian Johnson, Richard Shipman, Xiaofang Zhong, Junfeng Huang, Sanjay Asthana, Cynthia Carlsson, Ozioma Okonkwo, Lingjun Li. In-depth Site-specific Analysis of N-glycoproteome in Human Cerebrospinal Fluid and Glycosylation Landscape Changes in Alzheimer's Disease. Molecular & Cellular Proteomics 2021, 20 , 100081. https://doi.org/10.1016/j.mcpro.2021.100081
  75. Daniel L. Hurdiss, Ieva Drulyte, Yifei Lang, Tatiana M. Shamorkina, Matti F. Pronker, Frank J. M. van Kuppeveld, Joost Snijder, Raoul J. de Groot. Cryo-EM structure of coronavirus-HKU1 haemagglutinin esterase reveals architectural changes arising from prolonged circulation in humans. Nature Communications 2020, 11 (1) https://doi.org/10.1038/s41467-020-18440-6
  76. Adam M. Hawkridge, Sven Hackbusch. Ultraviolet photodissociation of fondaparinux generates signature antithrombin-like 3-O-sulfated -GlcNS3S6S- monosaccharide fragment (Y3/C3). Analytical and Bioanalytical Chemistry 2020, 412 (28) , 7925-7935. https://doi.org/10.1007/s00216-020-02925-w
  77. Kirti Pandey, Nicole A. Mifsud, Terry C.C. Lim Kam Sian, Rochelle Ayala, Nicola Ternette, Sri H. Ramarathinam, Anthony W. Purcell. In-depth mining of the immunopeptidome of an acute myeloid leukemia cell line using complementary ligand enrichment and data acquisition strategies. Molecular Immunology 2020, 123 , 7-17. https://doi.org/10.1016/j.molimm.2020.04.008
  78. Saar A. M. Laarse, Charlotte A. G. H. Gelder, Marshall Bern, Michiel Akeroyd, Maurien M. A. Olsthoorn, Albert J. R. Heck. Targeting proline in (phospho)proteomics. The FEBS Journal 2020, 287 (14) , 2979-2997. https://doi.org/10.1111/febs.15190
  79. Thierry Schmidlin, Maarten Altelaar. Effects of electron-transfer/higher-energy collisional dissociation (EThcD) on phosphopeptide analysis by data-independent acquisition. International Journal of Mass Spectrometry 2020, 452 , 116336. https://doi.org/10.1016/j.ijms.2020.116336
  80. James A. Wilkins, Krista Kaasik, Robert J. Chalkley, Alma L. Burlingame. Characterization of Prenylated C-terminal Peptides Using a Thiopropyl-based Capture Technique and LC-MS/MS. Molecular & Cellular Proteomics 2020, 19 (6) , 1005-1016. https://doi.org/10.1074/mcp.RA120.001944
  81. Biling Huang, Yan Liu, Hongwei Yao, Yufen Zhao. NMR-based investigation into protein phosphorylation. International Journal of Biological Macromolecules 2020, 145 , 53-63. https://doi.org/10.1016/j.ijbiomac.2019.12.171
  82. K. Rachael Parks, Anna J. MacCamy, Josephine Trichka, Matthew Gray, Connor Weidle, Andrew J. Borst, Arineh Khechaduri, Brittany Takushi, Parul Agrawal, Javier Guenaga, Richard T. Wyatt, Rhea Coler, Michael Seaman, Celia LaBranche, David C. Montefiori, David Veesler, Marie Pancera, Andrew McGuire, Leonidas Stamatatos. Overcoming Steric Restrictions of VRC01 HIV-1 Neutralizing Antibodies through Immunization. Cell Reports 2019, 29 (10) , 3060-3072.e7. https://doi.org/10.1016/j.celrep.2019.10.071
  83. Gemma Hardman, Simon Perkins, Philip J Brownridge, Christopher J Clarke, Dominic P Byrne, Amy E Campbell, Anton Kalyuzhnyy, Ashleigh Myall, Patrick A Eyers, Andrew R Jones, Claire E Eyers. Strong anion exchange‐mediated phosphoproteomics reveals extensive human non‐canonical phosphorylation. The EMBO Journal 2019, 38 (21) https://doi.org/10.15252/embj.2018100847
  84. Janaína Capelli‐Peixoto, Simon Ngao Mule, Fabia Tomie Tano, Giuseppe Palmisano, Beatriz Simonsen Stolf. Proteomics and Leishmaniasis: Potential Clinical Applications. PROTEOMICS – Clinical Applications 2019, 13 (6) https://doi.org/10.1002/prca.201800136
  85. Oksana Tyshchuk, Christoph Gstöttner, Dennis Funk, Simone Nicolardi, Stefan Frost, Stefan Klostermann, Tim Becker, Elena Jolkver, Felix Schumacher, Claudia Ferrara Koller, Hans Rainer Völger, Manfred Wuhrer, Patrick Bulau, Michael Mølhøj. Characterization and prediction of positional 4-hydroxyproline and sulfotyrosine, two post-translational modifications that can occur at substantial levels in CHO cells-expressed biotherapeutics. mAbs 2019, 11 (7) , 1219-1232. https://doi.org/10.1080/19420862.2019.1635865
  86. Elizabeth S. Hecht, Michaela Scigelova, Shannon Eliuk, Alexander Makarov. Fundamentals and Advances of Orbitrap Mass Spectrometry. 2019, 1-40. https://doi.org/10.1002/9780470027318.a9309.pub2
  87. Haopeng Xiao, Fangxu Sun, Suttipong Suttapitugsakul, Ronghu Wu. Global and site‐specific analysis of protein glycosylation in complex biological systems with Mass Spectrometry. Mass Spectrometry Reviews 2019, 38 (4-5) , 356-379. https://doi.org/10.1002/mas.21586
  88. John R. Yates. Large‐Scale Phosphoproteomics. 2019, 291-309. https://doi.org/10.1002/9781119081661.ch12
  89. Parviz Ghezellou, Vannuruswamy Garikapati, Seyed Mahdi Kazemi, Kerstin Strupat, Alireza Ghassempour, Bernhard Spengler. A perspective view of top‐down proteomics in snake venom research. Rapid Communications in Mass Spectrometry 2019, 33 (S1) , 20-27. https://doi.org/10.1002/rcm.8255
  90. Ivo A. Hendriks, Sara C. Larsen, Michael L. Nielsen. An Advanced Strategy for Comprehensive Profiling of ADP-ribosylation Sites Using Mass Spectrometry-based Proteomics*. Molecular & Cellular Proteomics 2019, 18 (5) , 1010-1026. https://doi.org/10.1074/mcp.TIR119.001315
  91. Noritaka Hashii, Junya Suzuki, Hisatoshi Hanamatsu, Jun-ichi Furukawa, Akiko Ishii-Watabe. In-depth site-specific O-Glycosylation analysis of therapeutic Fc-fusion protein by electron-transfer/higher-energy collisional dissociation mass spectrometry. Biologicals 2019, 58 , 35-43. https://doi.org/10.1016/j.biologicals.2019.01.005
  92. Alexandra C. Walls, Xiaoli Xiong, Young-Jun Park, M. Alejandra Tortorici, Joost Snijder, Joel Quispe, Elisabetta Cameroni, Robin Gopal, Mian Dai, Antonio Lanzavecchia, Maria Zambon, Félix A. Rey, Davide Corti, David Veesler. Unexpected Receptor Functional Mimicry Elucidates Activation of Coronavirus Fusion. Cell 2019, 176 (5) , 1026-1039.e15. https://doi.org/10.1016/j.cell.2018.12.028
  93. Elise J. Needham, Benjamin L. Parker, Timur Burykin, David E. James, Sean J. Humphrey. Illuminating the dark phosphoproteome. Science Signaling 2019, 12 (565) https://doi.org/10.1126/scisignal.aau8645
  94. P. Boomathi Pandeswari, Varatharajan Sabareesh. Middle-down approach: a choice to sequence and characterize proteins/proteomes by mass spectrometry. RSC Advances 2019, 9 (1) , 313-344. https://doi.org/10.1039/C8RA07200K
  95. Stanislav Naryzhny. Inventory of proteoforms as a current challenge of proteomics: Some technical aspects. Journal of Proteomics 2019, 191 , 22-28. https://doi.org/10.1016/j.jprot.2018.05.008
  96. Shasha Li, Yue Zhou, Kaijie Xiao, Jing Li, Zhixin Tian. Selective fragmentation of the N‐glycan moiety and protein backbone of ribonuclease B on an Orbitrap Fusion Lumos Tribrid mass spectrometer. Rapid Communications in Mass Spectrometry 2018, 32 (23) , 2031-2039. https://doi.org/10.1002/rcm.8273
  97. Andrew J Borst, Connor E Weidle, Matthew D Gray, Brandon Frenz, Joost Snijder, M Gordon Joyce, Ivelin S Georgiev, Guillaume BE Stewart-Jones, Peter D Kwong, Andrew T McGuire, Frank DiMaio, Leonidas Stamatatos, Marie Pancera, David Veesler. Germline VRC01 antibody recognition of a modified clade C HIV-1 envelope trimer and a glycosylated HIV-1 gp120 core. eLife 2018, 7 https://doi.org/10.7554/eLife.37688
  98. Karli R. Reiding, Albert Bondt, Vojtech Franc, Albert J.R. Heck. The benefits of hybrid fragmentation methods for glycoproteomics. TrAC Trends in Analytical Chemistry 2018, 108 , 260-268. https://doi.org/10.1016/j.trac.2018.09.007
  99. Charlotte Gaviard, Thierry Jouenne, Julie Hardouin. Proteomics of Pseudomonas aeruginosa : the increasing role of post-translational modifications. Expert Review of Proteomics 2018, 15 (9) , 757-772. https://doi.org/10.1080/14789450.2018.1516550
  100. Hilda Garay, Luis Ariel Espinosa, Yasser Perera, Aniel Sánchez, David Diago, Silvio E. Perea, Vladimir Besada, Osvaldo Reyes, Luis Javier González. Characterization of low‐abundance species in the active pharmaceutical ingredient of CIGB‐300: A clinical‐grade anticancer synthetic peptide. Journal of Peptide Science 2018, 24 (6) https://doi.org/10.1002/psc.3081
Load all citations
  • Abstract

    Figure 1

    Figure 1. EThcD MS/MS spectrum of a doubly phosphorylated peptide. RGTGQSDDSDIWDDTALIK is doubly phosphorylated and contains in total four potential phosphorylation sites. EThcD generates dual ion series that enable phosphorylation site localization with very high confidence (phosphoRS site probabilities: T(3), 0.0%; S(6), 100.0%; S(9), 100.0%; T(15), 0.0%). SEQUEST Xcorr 7.79.

    Figure 2

    Figure 2. EThcD spectrum of a proline-containing phosphopeptide. This EThcD spectrum of a doubly charged peptide that contains four serine residues, one of which is phosphorylated. ETD does not cleave the N–Cα bond N-terminal to proline and the phosphorylation site probability is only 50% based on c- and z-ions alone. Dual fragmentation by EThcD generates complementary sequence information from c/z- and b/y-ions (SEQUEST Xcorr 4.10). Here, the exact phosphosite is revealed by y-ions that cover the corresponding phosphosite (phosphoRS site probabilitis: S(1): 0.0; S(3): 0.0; S(8): 99.5; S(10): 0.5). SEQUEST Xcorr 4.10.

  • References

    ARTICLE SECTIONS
    Jump To

    This article references 41 other publications.

    1. 1
      Cohen, P. The regulation of protein function by multisite phosphorylation--a 25 year update Trends Biochem. Sci. 2000, 25 (12) 596 601
    2. 2
      Hunter, T. Signaling--2000 and beyond Cell 2000, 100 (1) 113 27
    3. 3
      Olsen, J. V.; Blagoev, B.; Gnad, F.; Macek, B.; Kumar, C.; Mortensen, P.; Mann, M. Global, in vivo, and site-specific phosphorylation dynamics in signaling networks Cell 2006, 127 (3) 635 48
    4. 4
      Eyrich, B.; Sickmann, A.; Zahedi, R. P. Catch me if you can: mass spectrometry-based phosphoproteomics and quantification strategies Proteomics 2011, 11 (4) 554 70
    5. 5
      Mann, M.; Jensen, O. N. Proteomic analysis of post-translational modifications Nat. Biotechnol. 2003, 21 (3) 255 61
    6. 6
      Di Palma, S.; Hennrich, M. L.; Heck, A. J.; Mohammed, S. Recent advances in peptide separation by multidimensional liquid chromatography for proteome analysis J. Proteomics 2012, 75 (13) 3791 813
    7. 7
      Rush, J.; Moritz, A.; Lee, K. A.; Guo, A.; Goss, V. L.; Spek, E. J.; Zhang, H.; Zha, X. M.; Polakiewicz, R. D.; Comb, M. J. Immunoaffinity profiling of tyrosine phosphorylation in cancer cells Nat. Biotechnol. 2005, 23 (1) 94 101
    8. 8
      Ficarro, S. B.; McCleland, M. L.; Stukenberg, P. T.; Burke, D. J.; Ross, M. M.; Shabanowitz, J.; Hunt, D. F.; White, F. M. Phosphoproteome analysis by mass spectrometry and its application to Saccharomyces cerevisiae Nat. Biotechnol. 2002, 20 (3) 301 5
    9. 9
      Zhou, H.; Low, T. Y.; Hennrich, M. L.; van der Toorn, H.; Schwend, T.; Zou, H.; Mohammed, S.; Heck, A. J. Enhancing the identification of phosphopeptides from putative basophilic kinase substrates using Ti (IV) based IMAC enrichment Mol. Cell. Proteomics 2011, 10 (10) M110 006452
    10. 10
      Boersema, P. J.; Mohammed, S.; Heck, A. J. Phosphopeptide fragmentation and analysis by mass spectrometry J. Mass Spectrom. 2009, 44 (6) 861 78
    11. 11
      Beausoleil, S. A.; Jedrychowski, M.; Schwartz, D.; Elias, J. E.; Villen, J.; Li, J.; Cohn, M. A.; Cantley, L. C.; Gygi, S. P. Large-scale characterization of HeLa cell nuclear phosphoproteins Proc. Natl. Acad. Sci. U.S.A. 2004, 101 (33) 12130 5
    12. 12
      Schroeder, M. J.; Shabanowitz, J.; Schwartz, J. C.; Hunt, D. F.; Coon, J. J. A neutral loss activation method for improved phosphopeptide sequence analysis by quadrupole ion trap mass spectrometry Anal. Chem. 2004, 76 (13) 3590 8
    13. 13
      Olsen, J. V.; Macek, B.; Lange, O.; Makarov, A.; Horning, S.; Mann, M. Higher-energy C-trap dissociation for peptide modification analysis Nat. Methods 2007, 4 (9) 709 12
    14. 14
      Chi, A.; Huttenhower, C.; Geer, L. Y.; Coon, J. J.; Syka, J. E.; Bai, D. L.; Shabanowitz, J.; Burke, D. J.; Troyanskaya, O. G.; Hunt, D. F. Analysis of phosphorylation sites on proteins from Saccharomyces cerevisiae by electron transfer dissociation (ETD) mass spectrometry Proc. Natl. Acad. Sci. U.S.A. 2007, 104 (7) 2193 8
    15. 15
      Molina, H.; Matthiesen, R.; Kandasamy, K.; Pandey, A. Comprehensive comparison of collision induced dissociation and electron transfer dissociation Anal. Chem. 2008, 80 (13) 4825 35
    16. 16
      Beausoleil, S. A.; Villen, J.; Gerber, S. A.; Rush, J.; Gygi, S. P. A probability-based approach for high-throughput protein phosphorylation analysis and site localization Nat. Biotechnol. 2006, 24 (10) 1285 92
    17. 17
      Stensballe, A.; Jensen, O. N.; Olsen, J. V.; Haselmann, K. F.; Zubarev, R. A. Electron capture dissociation of singly and multiply phosphorylated peptides Rapid Commun. Mass Spectrom. 2000, 14 (19) 1793 800
    18. 18
      Kelstrup, C. D.; Hekmat, O.; Francavilla, C.; Olsen, J. V. Pinpointing phosphorylation sites: Quantitative filtering and a novel site-specific x-ion fragment J. Proteome Res. 2011, 10 (7) 2937 48
    19. 19
      Shin, Y. S.; Moon, J. H.; Kim, M. S. Observation of phosphorylation site-specific dissociation of singly protonated phosphopeptides J. Am. Soc. Mass Spectrom. 2010, 21 (1) 53 9
    20. 20
      Gehrig, P. M.; Roschitzki, B.; Rutishauser, D.; Reiland, S.; Schlapbach, R. Phosphorylated serine and threonine residues promote site-specific fragmentation of singly charged, arginine-containing peptide ions Rapid Commun. Mass Spectrom. 2009, 23 (10) 1435 45
    21. 21
      Lu, B.; Ruse, C.; Xu, T.; Park, S. K.; Yates, J. R., 3rd Automatic validation of phosphopeptide identifications from tandem mass spectra Anal. Chem. 2007, 79 (4) 1301 10
    22. 22
      Bailey, C. M.; Sweet, S. M.; Cunningham, D. L.; Zeller, M.; Heath, J. K.; Cooper, H. J. SLoMo: automated site localization of modifications from ETD/ECD mass spectra J. Proteome Res. 2009, 8 (4) 1965 71
    23. 23
      Savitski, M. M.; Mathieson, T.; Becher, I.; Bantscheff, M. H-score, a mass accuracy driven rescoring approach for improved peptide identification in modification rich samples J. Proteome Res. 2010, 9 (11) 5511 6
    24. 24
      Ruttenberg, B. E.; Pisitkun, T.; Knepper, M. A.; Hoffert, J. D. PhosphoScore: an open-source phosphorylation site assignment tool for MSn data Journal of Proteome Research 2008, 7 (7) 3054 9
    25. 25
      Hansen, T. A.; Sylvester, M.; Jensen, O. N.; Kjeldsen, F. Automated and high confidence protein phosphorylation site localization using complementary collision-activated dissociation and electron transfer dissociation tandem mass spectrometry Anal. Chem. 2012, 84 (22) 9694 9
    26. 26
      Vandenbogaert, M.; Hourdel, V.; Jardin-Mathe, O.; Bigeard, J.; Bonhomme, L.; Legros, V.; Hirt, H.; Schwikowski, B.; Pflieger, D. Automated phosphopeptide identification using multiple MS/MS fragmentation modes J. Proteome Res. 2012, 11 (12) 5695 703
    27. 27
      Taus, T.; Kocher, T.; Pichler, P.; Paschke, C.; Schmidt, A.; Henrich, C.; Mechtler, K. Universal and confident phosphorylation site localization using phosphoRS J. Proteome Res. 2011, 10 (12) 5354 62
    28. 28
      Frese, C. K.; Altelaar, M.; van den Toorn, H. W.; Nolting, D.; Griep-Raming, J.; Heck, A. J.; Mohammed, S. Toward full peptide sequence coverage by dual fragmentation combining electron-transfer and higher-energy collision dissociation tandem mass spectrometry Anal. Chem. 2012, 84 (22) 9668 73
    29. 29
      Altelaar, A. F.; Frese, C. K.; Preisinger, C.; Hennrich, M. L.; Schram, A. W.; Timmers, H. T.; Heck, A. J.; Mohammed, S. Benchmarking stable isotope labeling based quantitative proteomics J. Proteomics 2012,  DOI: 10.1016/j.jprot.2012.10.009
    30. 30
      Zhou, H.; Ye, M.; Dong, J.; Han, G.; Jiang, X.; Wu, R.; Zou, H. Specific phosphopeptide enrichment with immobilized titanium ion affinity chromatography adsorbent for phosphoproteome analysis J. Proteome Res. 2008, 7 (9) 3957 67
    31. 31
      Yu, Z.; Han, G.; Sun, S.; Jiang, X.; Chen, R.; Wang, F.; Wu, R.; Ye, M.; Zou, H. Preparation of monodisperse immobilized Ti(4+) affinity chromatography microspheres for specific enrichment of phosphopeptides Anal. Chim. Acta 2009, 636 (1) 34 41
    32. 32
      Zhou, H.; Ye, M.; Dong, J.; Corradini, E.; Cristobal, A.; Heck, A. J. R.; Zou, H.; Mohammed, S. Robust phosphoproteome enrichment using monodisperse microspheres-based immobilized titanium (IV) ion affinity chromatography. Nat. Protoc. 2012, accepted
    33. 33
      Kall, L.; Canterbury, J. D.; Weston, J.; Noble, W. S.; MacCoss, M. J. Semi-supervised learning for peptide identification from shotgun proteomics datasets Nat. Methods 2007, 4 (11) 923 5
    34. 34
      Mertins, P.; Udeshi, N. D.; Clauser, K. R.; Mani, D. R.; Patel, J.; Ong, S. E.; Jaffe, J. D.; Carr, S. A. iTRAQ labeling is superior to mTRAQ for quantitative global proteomics and phosphoproteomics Mol. Cell. Proteomics 2012, 11 (6) M111 014423
    35. 35
      Kandasamy, K.; Pandey, A.; Molina, H. Evaluation of Several MS/MS Search Algorithms for Analysis of Spectra Derived from Electron Transfer Dissociation Experiments Anal. Chem. 2009, 81 (17) 7170 80
    36. 36
      Elias, J. E.; Gygi, S. P. Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry Nat. Methods 2007, 4 (3) 207 14
    37. 37
      Horn, D. M.; Zubarev, R. A.; McLafferty, F. W. Automated de novo sequencing of proteins by tandem high-resolution mass spectrometry Proc. Natl. Acad. Sci. U.S.A. 2000, 97 (19) 10313 7
    38. 38
      Cooper, H. J.; Hudgins, R. R.; Hakansson, K.; Marshall, A. G. Secondary fragmentation of linear peptides in electron capture dissociation Int. J. Mass Spectrom. 2003, 228 (2–3) 723 8
    39. 39
      Li, W.; Song, C.; Bailey, D. J.; Tseng, G. C.; Coon, J. J.; Wysocki, V. H. Statistical analysis of electron transfer dissociation pairwise fragmentation patterns Anal. Chem. 2011, 83 (24) 9540 5
    40. 40
      Kim, M. S.; Zhong, J.; Kandasamy, K.; Delanghe, B.; Pandey, A. Systematic evaluation of alternating CID and ETD fragmentation for phosphorylated peptides Proteomics 2011, 11 (12) 2568 72
    41. 41
      Nielsen, M. L.; Savitski, M. M.; Zubarev, R. A. Improving protein identification using complementary fragmentation techniques in fourier transform mass spectrometry Mol. Cell. Proteomics 2005, 4 (6) 835 45
  • Supporting Information

    Supporting Information

    ARTICLE SECTIONS
    Jump To

    Additional information as noted in the text. This material is available free of charge via the Internet at http://pubs.acs.org.


    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect