You are on page 1of 22

Expert Review of Proteomics

ISSN: 1478-9450 (Print) 1744-8387 (Online) Journal homepage: https://www.tandfonline.com/loi/ieru20

Detecting post-translational modification


signatures as potential biomarkers in clinical mass
spectrometry

Ruzanna Mnatsakanyan, Gerta Shema, Mark Basik, Gerald Batist, Christoph


H. Borchers, Albert Sickmann & René P. Zahedi

To cite this article: Ruzanna Mnatsakanyan, Gerta Shema, Mark Basik, Gerald Batist, Christoph
H. Borchers, Albert Sickmann & René P. Zahedi (2018) Detecting post-translational modification
signatures as potential biomarkers in clinical mass spectrometry, Expert Review of Proteomics,
15:6, 515-535, DOI: 10.1080/14789450.2018.1483340

To link to this article: https://doi.org/10.1080/14789450.2018.1483340

© 2018 The Author(s). Published by Informa View supplementary material


UK Limited, trading as Taylor & Francis
Group.

Published online: 03 Jul 2018. Submit your article to this journal

Article views: 9299 View related articles

View Crossmark data Citing articles: 39 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ieru20
EXPERT REVIEW OF PROTEOMICS
2018, VOL. 15, NO. 6, 515–535
https://doi.org/10.1080/14789450.2018.1483340

REVIEW

Detecting post-translational modification signatures as potential biomarkers in


clinical mass spectrometry
Ruzanna Mnatsakanyana, Gerta Shemaa, Mark Basikb, Gerald Batistb, Christoph H. Borchers b,c,d,e
,
Albert Sickmann a,f,g and René P. Zahedi a,b,e
a
Protein Dynamics, Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V, Dortmund, 44227, Germany; bGerald Bronfman Department of
Oncology, Jewish General Hospital, McGill University, Montreal, Quebec H4A 3T2, Canada; cUniversity of Victoria-Genome British Columbia
Proteomics Centre, University of Victoria, Victoria, British Columbia V8Z 7X8, Canada; dDepartment of Biochemistry and Microbiology, University of
Victoria, Victoria, British Columbia, V8P 5C2, Canada; eSegal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill
University, Montreal, Quebec H3T 1E2, Canada; fMedizinische Fakultät, Medizinische Proteom-Center (MPC), Ruhr-Universität Bochum, 44801
Bochum, Germany; gDepartment of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen AB24 3FX, Scotland, United Kingdom

ABSTRACT ARTICLE HISTORY


Introduction: Numerous diseases are caused by changes in post-translational modifications (PTMs). Received 23 October 2017
Therefore, the number of clinical proteomics studies that include the analysis of PTMs is increasing. Accepted 29 May 2018
Combining complementary information—for example changes in protein abundance, PTM levels, with KEYWORDS
the genome and transcriptome (proteogenomics)—holds great promise for discovering important Clinical proteomics;
drivers and markers of disease, as variations in copy number, expression levels, or mutations without biomarkers;
spatial/functional/isoform information is often insufficient or even misleading. post-translational
Areas covered: We discuss general considerations, requirements, pitfalls, and future perspectives in modification (PTM); LC-MS;
applying PTM-centric proteomics to clinical samples. This includes samples obtained from a human phosphorylation;
subject, for instance (i) bodily fluids such as plasma, urine, or cerebrospinal fluid, (ii) primary cells such glycosylation; proteolysis
as reproductive cells, blood cells, and (iii) tissue samples/biopsies.
Expert commentary: PTM-centric discovery proteomics can substantially contribute to the under-
standing of disease mechanisms by identifying signatures with potential diagnostic or even therapeutic
relevance but may require coordinated efforts of interdisciplinary and eventually multi-national con-
sortia, such as initiated in the cancer moonshot program. Additionally, robust and standardized mass
spectrometry (MS) assays—particularly targeted MS, MALDI imaging, and immuno-MALDI—may be
transferred to the clinic to improve patient stratification for precision medicine, and guide therapies.

1. PTMs and their role in health and disease archaea [1]) but also in their prevalence. Every human protein has
the potential to be post-translationally modified at least once
Cellular homeostasis is primarily maintained through adaptive
during its life span. The fundamental role of PTMs as switches of
changes of the proteome and metabolome. On the proteome
cellular homeostasis therefore implies that they represent critical
level, this mainly involves changes in (i) protein abundance levels,
and sensitive nodes for pathogenesis, both as drivers and markers,
for example through altered expression, degradation, or both, (ii)
and thus aberrant PTM patterns have been associated with numer-
protein localization, (iii) protein–protein interactions, (iv) protein
ous diseases.
function, and (v) protein activity—all of which can be modulated
Mass spectrometry (MS) is an unrivaled tool for detecting
by post-translational modifications (PTMs). PTM involves the
and quantifying PTMs and their changes in a broad variety of
attachment, removal, exchange, and rearrangement of functional
samples, including purified proteins and protein complexes,
groups to amino acid side chains and protein N-termini, but also
organelles, cells, tissues, or biofluids. Consequently, more than
the hydrolysis of peptide bonds (proteolysis). Indeed, PTMs are a
95% of current data on PTMs is derived from MS-based proteo-
most elegant, energetically efficient, and rapid strategy to modu-
mic studies [2] and—with the ongoing advances in instrumen-
late and maximize the functionality of a single gene product—
tation, methods, and bioinformatics—the number of known
depending on the type of PTM, this even can occur in a highly
PTMs is likely to increase in the future [3,4]. The largest and
reversible manner, such as in case of phosphorylation or redox
manually curated PTM database available, PhosphoSitePlus
modifications. Hence, throughout all species, PTMs are important
(www.phosphosite.org) contains more than 400,000 non-
for regulating fundamental biochemical processes. This is reflected
redundant PTMs on more than 20,000 proteins, mainly from
in both the high number of known PTMs (>400 reported in the
human and mouse [2] (Figure 1).
Uniprot database, 326 in eukaryotes, 250 in bacteria, and 80 in

CONTACT René P. Zahedi rene.zahedi@ladydavis.ca Lady Davis Institute - Jewish General Hospital, JGH Proteomics Centre, E-615, 3755 Côte Ste-Catherine
Road, Montreal H3T 1E2Quebec
*These authors contributed to this paper equally
supplementary material can be accessed here
© 2018 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License (http://creativecommons.org/licenses/by-nc-nd/4.0/),
which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited, and is not altered, transformed, or built upon in any way.
516 R. MNATSAKANYAN ET AL.

Figure 1. Distribution of the >400,000 PTM sites reported at www.phosphosite.org, October 2017. Due to historical and technical reasons five PTMs—Ser/Thr/Tyr
phosphorylation, ubiquitination, and acetylation—account for 92% of these modifications, but this may not necessarily mirror the biological relevance and
abundance of individual PTMs.

Figure 2. Frequently reported post-translational protein modifications (adapted from Pagel et al., Expert Rev Proteomics; 12(3):235-53; with permission).

Importantly, a protein sequence can have many different that particular protein. Even for a specific residue, there can be
modifiable amino acids (Figure 2), but not necessarily all will a competition between different modifications, as shown, for
be modified at the same time point or within the same copy of example, for Lys acetylation and ubiquitination [5], for different
EXPERT REVIEW OF PROTEOMICS 517

lipid modifications [6], and also for O-phosphorylation and In this review, we discuss general considerations, require-
O-GlcNac [7,8]. For each modifiable amino acid, there is an ments, pitfalls, and future perspectives in applying PTM-centric
equilibrium between the free and different modified versions proteomics to clinical samples—from experimental design to
within a biological system, which can be shifted to precisely data analysis. We focus on four classes of PTMs: phosphorylation,
drive cellular response in a specific direction. Particularly for glycosylation, proteolytic cleavage and oxidative Cysteine mod-
p53, it has been demonstrated that function and activity ifications and provide examples of relevant studies and methods.
depend on the complete PTM pattern rather individual PTMs,
as they can synergistically modulate protein structure (PTM
2. Can PTMs be used as biomarkers?
crosstalk and PTM code) [9-11]. Analyzing complete protein
PTM patterns is a major challenge for MS, as currently PTM Studying disease mechanisms has been mainly done with the
research is mainly restricted to the identification of peptides help of models, such as cell culture and animal models.
after proteolytic digestion (‘bottom-up’ proteomics) [12]. Thus, However, more and more, omics studies involve the analysis
information about the full modification state of individual pro- of clinical samples. For precision medicine, PTM signatures may
tein molecules is typically lost and would require the analysis of either be identified in disease models and then validated in
intact proteins (‘top-down’ proteomics) which, unfortunately, clinical samples or directly identified using clinical samples.
might not be applicable to the analysis of PTMs from clinical Although, for example, specific changes in phosphorylation
samples in the near future, as discussed elsewhere [12,13]. In and glycosylation are increasingly reported as potential markers
this review, we define ‘clinical samples’ as samples obtained of disease, and many PTM assays have been developed so far
from a human subject. This includes (i) bodily fluids such as (for instance reviewed in [23-25]), only few are really routinely
serum, plasma, urine, nasal fluid, cerebrospinal fluid, etc., and used for diagnostic purposes to date, due to a variety of
stool; (ii) primary cells, including reproductive cells, blood cells reasons [26]. We are still in the early days of PTM-specific
such as erythrocytes, leukocytes, platelets, etc.; (iii) tissue sam- research and detection and many potential PTM biomarkers
ples/biopsies; whereas studies including (primary) cell culture, and the corresponding assays still need to be validated exten-
patient derived xenografts or cell lines, and animal models are sively. Consequently, most clinical assays are genomics-based
not considered as ‘clinical’ in this review. or involve the use of antibodies, which are mainly available for
Numerous diseases are driven by the dysregulation of the proteins rather than for specific PTMs. For instance, the expert
proteome, either due to changes in expression levels, changes curated Cancer Biomarkers database (https://www.cancergen
in the primary sequence, and/or changes in PTMs. Non-synon- omeinterpreter.org/biomarkers) reports around 200 different
ymous single nucleotide polymorphisms (SNPs) can alter the genes as potential cancer biomarkers, ~50 of those as
activity or specificity of enzymes that attach/remove a certain approved. Still, whether pre-clinical, from case reports and
PTM to proteins and thereby can change for example phos- early trials, or approved, virtually all of them are either point
phorylation, glycosylation, or acetylation patterns in a path- mutations, gene amplifications, or expression changes, whereas
way-specific or even proteome-wide manner. SNPs can also no specific PTM markers are listed. Nevertheless, some PTM-
affect the range of substrates of these enzymes by altering specific assays are currently used. Glycated hemoglobin (HbA1c)
consensus/targeting/recognition motifs on common sub- is formed non-enzymatically upon contact with plasma glucose
strates or by incorporating such motifs into novel (non-phy- and is increased in diabetes [27]. Whereas diabetes can be
siological) substrates. Both cases, mutations of enzymes and diagnosed by measuring fasting glucose, HbA1c has several
substrates, can have severe consequences for cellular home- advantages such as higher repeatability [28]. HbA1c can also
ostasis [14,15]. For many types of cancer, a dysregulation of be measured in the non-fasting state, and elevated levels may
kinases has been reported, which can, for example, lead to be a risk factor for macrovascular disease [28]. The phosphor-
constitutive kinase activity and hyperactive signaling path- ylation levels of vasodilator stimulated phosphoprotein (VASP)
ways [16-18]. SNPs can also induce mistargeting of proteins are measured as indicator of P2Y12 receptor inhibition during
such as has been reported for the renal Fanconi syndrome, antiplatelet treatment [29]. Different glycan structures on the
where the mutation E3K in EHHADH produces a de novo surface of red blood cells define the blood type. Besides that,
mitochondrial import sequence [19,20]. Upon mitochondrial there is huge list of potential biomarkers reported in the litera-
import the mitochondrial targeting sequence is proteolytically ture, though their diagnostic or predictive value remains to be
removed and the truncated and mislocalized protein impairs proven in many cases (see supplemental table 1).
energy production [19,20]. Differential antibody glycosylation Considering the complexity of human health and disease,
has been associated with autoimmunity [21], and murine in many cases a single molecule (e.g. a single gene, protein,
cytomegalovirus has been reported to activate intracellular glycan or phosphorylation site) may be insufficient as specific
PTM-based signaling pathways through expression of the biomarker for diagnosis and for predicting effective treatment.
viral G protein-coupled receptor M33 [22]. Owing to these Recent studies indicate, that combining multiple ‘omics’ levels
fundamental roles of PTMs in health and disease, their analysis such as (i) the analysis of protein expression and PTMs, (ii)
is important from two different aspects: First, to understand proteomics, metabolomics and lipidomics, or (iii) proteoge-
disease mechanisms on the molecular level. Second, in the nomics integrating genomics, transcriptomics and proteomics
context of precision medicine: To identify PTM signatures that data is beneficial for identifying specific disease patterns and
can be correlated with disease (progression), and that may signatures. Particularly in cancer, it has emerged that informa-
help to diagnose and stratify patients and to predict the tion from next-generation sequencing or RNAseq alone is not
effectiveness of a treatment and guide therapy. sufficient to understand the mechanisms underlying the
518 R. MNATSAKANYAN ET AL.

development of resistance in certain tumors [30], such that clinical environments, but also due to underestimating how small
additional data on protein expression and PTM levels is variations in SOPs, such as changing incubation times by only a
needed. Thus, mutations in the receptor tyrosine kinase epi- few minutes or buffers by a few µL, different lengths of time
dermal growth factor receptor (EGFR) are apparent in many between sampling and freezing, etc., can severely affect the pro-
cancer types and lead to aberrant phosphorylation signaling. teome and—particularly—PTM signaling pathways. Therefore, a
EGFR mutations are often used to select treatment with spe- close collaboration between clinicians and analytical scientists is
cific EGFR inhibitors for non-small cell lung cancer. Response mandatory to jointly (i) design realistic and robust SOPs [39-44]
rates, however, are still too low (40-80%) while concurrently and (ii) (ideally) to monitor their correct implementation, particu-
10-15% of patients without mutation do respond [31]. larly in the beginning of a project but also throughout its progress.
Obtaining data on protein phosphorylation levels for down- Because minor deviations from the agreed-upon sampling SOPs
stream targets of the EGFR signaling pathway might comple- cannot be completely excluded, it is also important to track and
ment the non-conclusive genomics data. Indeed, in a recent document these thoroughly [45] so that later inconsistent results
study by the Clinical Proteomic Tumor Analysis Consortium may be correlated with such changes. An important issue, in the
(CPTAC), only integration and clustering of DNA, RNA, protein, case of tissue, is how the samples are provided. Fresh-frozen tissue
and protein phosphorylation profiles allowed distinguishing might be preferable, but often clinical specimen are provided as
subtypes in 77 breast cancer tumors [32]. FFPE samples. Although sample preparation protocols for FFPE
tissues have been published [46-48], this type of sample prepara-
tion considerably complicates the entire analysis, and may reduce
3. General considerations for sampling, sample
both sensitivity and reproducibility. It should be kept in mind that
preparation and data analysis
fixation processes can potentially alter labile PTM signatures, such
When conducting PTM (and also global) proteome analyses on as phosphorylation, if not proteome-wide at least for certain path-
clinical samples, several important aspects have to be consid- ways that quickly respond to cell stress.
ered. While bodily fluids (such as blood, urine, or tear fluid) are Finally, variation can be introduced during the actual proteo-
usually available in high amounts and (almost) non-invasively, mic sample preparation and LC-MS analysis. Therefore, here also it
tissue samples are often limited in amount and/or are often is mandatory to strictly follow well-designed SOPs detailing all
formalin-fixed and paraffin-embedded (FFPE). For biopsies, individual steps, from lysis to sample analysis [49-51], including
most of the sample is often needed for histological purposes, quality control of proteolytic digestion prior to LC-MS analysis [52],
or is stored in dedicated biobanks. Consequently, the amounts as well as controls for labeling efficiency in case chemical stable
that remain for proteomic analyses are often strictly limited isotope labeling (SIL) is used [53,54]. Lysis and homogenization
(low to medium µg-range of protein) and downstream com- should be harsh and fast to preserve the biological state of the
patibility with sensitive proteomics workflows can be poor. sample and avoid artificial changes, particularly of fast and rever-
Another key challenge when analyzing PTMs from clinical sible PTMs. Tissues should be kept on ice and ideally, inhibitors
samples is heterogeneity, which may have a multitude of should be used where applicable (e.g. phosphatase inhibitor cock-
preanalytical sources that are usually not relevant or less tails for phosphoproteomics), including the use of protease inhi-
relevant for cell culture experiments and are only partially bitors (particularly when analyzing proteolytic cleavage). Protein
relevant for animal models [33]. First, even for healthy control digestion should be controlled prior to extensive analytical work-
groups, biological variation between individuals can be sub- flows, as unreproducible and inefficient digestion can substantially
stantial. This is due not only to generally occurring genetic affect and ruin sample analysis, including strong variation
variations, but, more importantly can be due to considerable between samples and artificial fold-changes. Digestion can be
differences in life style, diet, environment, and age, as well as controlled in multiple ways that differ in precision, effort, and
between genders. While it may already be challenging to sensitivity [52]. Gel electrophoresis of samples pre- and post-
clearly state what defines a ‘healthy’ control, the variation digestion does not require special equipment but is rather slow,
across patient cohorts can be substantially higher, and often cannot be automated and poor resolution in the low MW range
patients present with multiple diseases. Particularly for cancer, and comparably poor sensitivity do not allow to reliably evaluate
during the past decade we have learned that even for specific the reproducibility of individual digests. MALDI MS spectra can be
subtypes of cancer such as breast cancer, tumors are indivi- used for a quick and very sensitive overall pattern comparison
dual and may require specific, personalized, treatment [34]. using only small aliquots of the digested samples, but due to the
Therefore, a thorough and clear subtyping of patient cohorts huge sample complexity may lack the sensitivity to detect differ-
is as important as maintaining a sufficiently high number of ences apart from major components that are easy to digest.
individuals for both patient and matching (age, gender, etc.) Monolithic HPLC columns require a dedicated HPLC system, ide-
control groups [35]. ally with nL/µL UV detector, but are fast, sensitive, automated, and
Another major source of variation lies in the sampling proce- robust. Complete UV traces allow a reliable assessment of digest
dure [36-38]. Clinical samples have to be collected and stored over efficiency and reproducibility [52]. Finally, digested (but not pre-
time. While here the need for well-defined standard operating digest) samples can be analyzed by short LC-MS gradients, pre-
procedures (SOPs) that define how, when, and under exactly ferably using not the newest instrument generations in the labora-
which conditions samples should be taken, is well-accepted, tory, to assess the overall reproducibility. However, samples with
their implementation into daily clinical practice is often not poor digest efficiency may ruin the column or contaminate the
straightforward. This could be due not only to different personnel HPLC. LC-MS instruments have to be well-maintained, including
being involved in the sampling and the inevitable daily stress in regular cleaning, calibration, and (ideally) the analysis of a daily
EXPERT REVIEW OF PROTEOMICS 519

quality-control analysis of well-designed samples [55]. For this signaling PTM, phosphorylation, there might be only minor
purpose, specifically designed peptide mixtures [44,50,56] and qualitative changes (i.e. presence of phosphorylation sites)
software tools [57-60] allow monitoring the reproducibility of between control and stimulated or diseased conditions,
retention times, intensities, sensitivity, and mass deviation with whereas quantitative differences (i.e. degree of phosphoryla-
and without background. tion) can be vast. It is therefore unlikely that a specific phos-
For clinical samples, data analysis is substantially more phorylation site can be solely detected under a specific
challenging than for common proteome analyses, as it disease state and therefore just used as a biomarker. It is,
requires the analysis of larger sample cohorts as well as however, more likely that this phosphorylation is significantly
more sophisticated data analysis and statistical approaches. more or less prominent under disease conditions, for instance
Analyzing a large number of samples, however, leads to an representative of an increased or reduced activity of the cor-
additional challenge: missing values [61-63], in both label-free responding protein, while protein expression may be
and label-based quantitation approaches. Thus, even for tech- unchanged. Importantly, in some cases a phosphorylation
nical replicates analyzed consecutively under highly reprodu- might be only detectable due to a disease-relevant SNP: For
cible conditions, the overlap of confidently identified and even instance when single amino acid substitutions lead to the
more so quantified peptides and proteins is usually far below presence/exposure of either novel Ser/Thr/Tyr residues or
100% in discovery experiments. Generally, when analyzing novel kinase consensus motifs inducing a non-standard phos-
large cohorts, one should (i) only place samples on the auto- phorylation site. In both cases, the novel phosphorylated pep-
sampler immediately prior to injection, (ii) run daily quality tide might only be detectable using protein databases that
control samples, (iii) analyze samples in randomized order, to consider the respective mutation. Therefore, as long as a
avoid systematic biases derived from changing instrument specific PTM is not present only due to SNPs, a mere qualita-
performance, and (iv) use SIL peptides for precise quantitation tive analysis of clinical samples is not sufficient, and well-
for all steps following discovery. defined and carefully-conducted quantitative experiments
are mandatory to subtract relevant signaling from back-
ground. This is particularly relevant as clinical tissue samples
represent cell populations that may differ in their respective
4. From discovery to validation: sample analysis by
PTM (e.g. phosphorylation) status, such that disease markers
quantitative MS
may be suppressed in the final analysis. For instance, hetero-
For clinical samples, three different kinds of analysis plat- geneous tumors may contain certain cells with a significantly
forms have to be considered. First, discovery would rely on regulated PTM, while surrounding cells may mask the extent
non-targeted methods that provide high sensitivity and cov- of this regulation. The often transient and sub-stoichiometric
erage (in-depth analysis), but which do not necessarily pro- nature of PTMs renders an enrichment step prior to analysis
vide high throughput. Here the goal is to identify potential mandatory to make them accessible for MS analysis, with the
biomarker panels or targets from a manageable number of most efficient methods typically applied on the peptide level.
representative samples. This is mainly achieved through rela- This substantially improves the identification and quantitation
tive quantitation, where fold-changes between samples are of modified peptides, however, at the expense of more elabo-
detected, however without knowing analyte concentrations rate workflows and—consequently—further increases in tech-
or amounts. Second, there is validation with larger cohorts nical variation (as well as additional shortcomings of bottom-
focusing on a limited number of candidate proteins/pep- up PTM analysis discussed elsewhere [13]). Some examples of
tides, usually applying targeted MS techniques [64,65] such enrichment strategies that have been successfully applied to
as multiple reaction monitoring (MRM) [66,67] or parallel clinical samples will be given in the following chapters. There,
reaction monitoring (PRM) [68], and usually employing abso- we will focus on four classes of PTMs of high abundance and
lute quantitation where the concentration or amount of an relevance (phosphorylation, glycosylation, proteolytic clea-
analyte in a sample is precisely determined using a specific vage, and redox modifications) that can be enriched without
internal standard (discussed more in detail in the following antibody IP, as such methods often require relatively large
chapter). This not only allows obtaining more reliable fold- amounts of sample and are usually not applied to clinical
changes, but in case of PTMs enables determining site occu- samples.
pancy, for example which share of EGFR molecules in a cell
is phosphorylated at a specific position. This information
5. Methods for quantifying PTMs in clinical samples
may be more biologically relevant then mere fold-changes
usually obtained in MS-based proteomics. Third, there is MS-based discovery is still mostly done using data-dependent
translation to the clinic. Here, sensitivity is still an issue, acquisition (DDA) methods, where the MS continuously applies
but robustness, throughput, and precise quantitation of a analysis cycles in which usually the top 5-20 most intense peptides
limited number of candidates are even more important, so are subjected to fragmentation, slight variations in (i) retention
that absolute quantification is essential. times, (ii) signal intensities, as well as (iii) co-isolation of peptides,
As aforementioned, there is usually an equilibrium—in the and (iv) poor MS/MS spectrum quality, can cause so-called ‘under-
case of reversible PTMs often very dynamic—between mod- sampling’—in other words, although a peptide is clearly present in
ified and non-modified states of amino acid residues and a sample, it will not be identified and therefore not quantified. This
proteins, which can be shifted in order to induce certain affects both label-free and label-based approaches, although the
biochemical effects. For instance, for the best-studied latter usually include numerous multiplexed experiments in order
520 R. MNATSAKANYAN ET AL.

to analyze sufficiently high numbers of samples (e.g. ten 10-plex cohorts [75-77]. Here, in contrast to DDA, individual ions are not
experiments). In label-free quantitation (discovery), this has been targeted for isolation and fragmentation, but instead a mass
compensated for by various software tools that use so-called range of interest (e.g. 300-1500 m/z) is divided in multiple
matching or alignment of LC-MS runs [69-71]. Here, different smaller m/z windows that are consecutively selected for frag-
LC-MS runs can be aligned based on unique characteristics of mentation, leading to hybrid MS/MS spectra often including
‘features’ (i.e. ions represented by e.g. m/z, retention time window, several peptides. Individual fragment ion spectra are then
elution profile, isotope pattern). Thus, it is sufficient that a specific later assembled based on elution profiles. In DIA peptides are
peptide (feature) is only reliably identified in a single LC-MS run to continuously fragmented throughout their elution profile, and
trace it in all other analyses, as long as these are sufficiently quantitation can be conducted using fragment ions rather than
reproducible. Thus, quantitation becomes almost independent precursor ions, reducing interference and allowing a more
of identification, allowing a considerably improved coverage robust quantitation than in DDA. A potential shortcoming of
across different samples. Still, inconsistent behavior during DIA methods is that, for optimal performance, they require the
LC-MS analysis limits the alignment capabilities of the different generation of comprehensive spectral libraries, which may not
software tools. Even when identified and clearly present in multi- be optimal, particularly for discovering novel candidates under
ple runs, the same peptide may still be represented by different disease conditions. Thus, spectral libraries would be ideally
features, so that the quantitative information is incomplete with- required for representative samples of all cohorts included in
out manual inspection (Figure 3). In large datasets, missing values the study.
can be predicted by imputation [62], which facilitates statistical Generally, MS methods must be thoroughly validated and
data analysis but which is certainly not ideal. the transfer to a clinical environment for daily use as routine
For modified peptides, quantitation is even more challen- procedure has to be realistic and feasible. This may include a
ging. First, single-peptide quantitation in discovery approaches shift from nano-LC typically used in discovery proteomics to
is less robust than protein quantitation based on multiple pep- larger columns and flow rates that are considerably more
tide measurements. Second, the issue of missing values is more robust at the expense of slight sensitivity losses [78]. There is
pronounced because, in addition to alignment problems, for always a trade-off between proteomic coverage and analysis
PTMs the position of the modified amino acid has to be unam- time on one hand, and precision and costs on the other.
biguously identified within the peptide sequence [72] (Figure 3). Sample fractionation prior to LC-MS will substantially improve
This can be complicated, as multiple modifiable amino acids coverage, not only for global proteomics, but also with regard
can be in close proximity within a sequence (for example, the to PTM-centric workflows where fractionation can boost the
unambiguous localization of the phosphorylation site in the identification of modified peptides. However, fractionation in
sequence SSSLPAYGR in human dermatin is challenging) and, label-free approaches is challenging as—even under ideal and
in addition, MS/MS spectra quality can be severely impaired reproducible conditions—it will add another level of technical
due to the modification(s) [73,74]. Consequently, label-free variation which is extremely challenging to handle with data
PTM-centric discovery studies suffer even more from missing analysis software tools. An alternative strategy, one that is
values and inconsistent quantitation (Figure 3). Recent studies particularly relevant for PTM-centric studies due to more com-
demonstrated that data-independent acquisition (DIA) strate- plex and susceptible workflows, is the use of chemical SIL
gies allow a more consistent, reproducible, and precise quanti- strategies allowing multiplexing of samples. The most popular
tation of proteomes and particularly of larger sample sets and commercial labels (iTRAQ [79,80] and TMT [81,82]) allow

Figure 3. Issues with alignment of LC-MS runs in label-free discovery experiments, exemplified by theoretical extracted ion chromatograms leading to inconsistent
quantitation of peptides. (A) Alignment can be generally impaired by inconsistent retention times due to non-linear shifts of peptide and background peaks. A
substantial retention time shift of 20 s in one of the LC-MS runs might lead to misassignment of the peptide to another feature (feature 2). B) For PTM peptides
inconsistent assignment of PTM modification sites within an identified peptide can also impair alignment; preferentially if various modifiable amino acid residues are
in close proximity and if MS/MS spectra quality is insufficient (‘s’ indicates the phosphorylation site assigned by the database search). C) Indeed, both effects can
superimpose, leading to multiple features that may represent a single peptide. In targeted approaches this can be compensated for by the use of stable isotope
labeled (SIL) reference peptides.
EXPERT REVIEW OF PROTEOMICS 521

multiplexing of eight and ten samples, respectively. Though this glycosylation [85], proteolysis [86], Lys acetylation [87], cysteine
multiplexing capacity is still way too low for virtually any clinical modifications [88], or combinations thereof. However, TMT and
setting, several such experiments could be combined in order iTRAQ suffer from ratio compression (Figure 4), an effect caused
to analyze sufficient numbers of patient samples. Ideally, this by the potential co-isolation of background peptides and con-
would imply the use of a mixture of samples (all or a subset) sistent, low-level background fragmentation [89], as virtually
that is applied as one channel to every single multiplexing every peptide in the sample releases the same reporter ions
experiment for normalization purposes. Because, after labeling, (labeling efficiency >97% can be readily achieved) [90,91].
the same peptide derived from different samples is always Notably, this effect renders reporter ion-based methods incom-
virtually isobaric, even multiplexing 10 samples does not con- patible with DIA methods as it cannot be deduced how much
siderably increase sample complexity. Consequently, the differ- of a specific reporter ion signal in a hybrid spectrum derives
ently labeled versions of a single peptide will be isolated and from a specific peptide (Figure 4). However, selective PTM
fragmented just like a single precursor in DDA experiments, enrichment and the usage of dedicated DDA analysis strategies,
releasing reporter ions that reflect the relative amounts of the particularly involving narrow m/z isolation windows with mod-
peptide in the different samples in the low m/z region of the ern instruments of 0.4 to 0.8 m/z can largely compensate for
MS/MS spectrum, which furthermore contains the usual peptide ratio compression. Importantly, iTRAQ and TMT can alter the
backbone information (Figure 4). Once samples are multiplexed, physicochemical properties of peptides and thus affect the
they can undergo various enrichment and fractionation strate- efficiency of PTM enrichment and MS detection, but also chro-
gies without largely compromising precision. Thus, iTRAQ and matographic behavior. Consequently, the validation of iTRAQ/
TMT are well-suited for highly sensitive PTM-centric analysis, as TMT derived target peptides for example by targeted methods
multiplexing allows quantitative PTM analysis even from as little such as parallel reaction monitoring (PRM) [92] with unlabeled
as 10 µg of protein start material per sample or condition [83] samples may not always be successful, even for sequences
and has been applied to quantify e.g. phosphorylation [84], which should be well detectable. Recent improvements

Figure 4. Problems with reporter ion-based quantitation, demonstrated for iTRAQ 8plex. A) In DDA experiments, all differentially labeled versions of an iTRAQ/TMT
labeled peptide will contribute to (basically) a single isotope pattern in MS survey scans. In this case, an isolation window of 1.6 m/z (dashed lines) is sufficient to
prevent co-isolation. Upon fragmentation, the reporter ions reflecting the relative amounts of the peptide in the eight samples will be released. B) iTRAQ 8plex
reporter ion area of an MS/MS spectrum, exemplified by 4 diseased samples (red) and four healthy controls (blue). Note that the higher m/z range of the MS/MS (not
shown here) contains the fragment ions representing the peptide backbone. From, the reporter ions it can be deduced that the peptide is significantly
downregulated in the diseased samples. C) If another peptide is in close m/z proximity, an isolation window of 1.6 m/z (grey) may be too wide, leading to co-
isolation and ratio compression (representative MS/MS spectrum shown in D), whereas a narrower isolation window of 0.8 m/z (black) may prevent this;
representative MS/MS spectrum shown in B). D) Ratio compression due to co-isolation distorts quantitation: The peptide no longer appears to be significantly
downregulated. E) In DIA experiments, wider windows are selected for continuous fragmentation of the entire mass range, as represented here by 4.0 m/z. F) In DIA,
the elution profiles of the fragment ions are assembled from individual MS/MS scans, and matching elution profiles point to fragment ions from the same precursor
(as demonstrated by the black traces). In contrast, reporter ions will be constantly present in all MS/MS spectra with varying intensities, rendering it impossible to
infer reporter ion intensities for individual peptides.
522 R. MNATSAKANYAN ET AL.

exploiting mass defects and ultra-high resolution MS, such as validation approaches. They have the advantage of always com-
the NeuCode approach developed by the Coon lab (for a bining peptide (or protein) quantitation with identification,
detailed explanation of the approach we refer to [93,94]) instead of just detecting an increase in the signal as in anti-
show great promise that one of the drawbacks of current body-based procedures. In MRM, the MS is set up to specifically
reporter ion methods, that is the limited multiplexing capacity, quantify a pre-defined set of candidates in a very specific man-
will be overcome in the future, allowing for more comprehen- ner, allowing higher precision and throughput, as sample analy-
sive studies. Importantly, this would massively reduce the pro- sis can often be performed in one hour or less. However, the non-
blem of missing values in large scale studies, because a targeted bulk of the proteome remains a black box. Ideally,
quantitative value will be obtained for every peptide as long MRM/PRM assays would involve the usage of SIL reference pep-
as the peptide is really present in the sample above the detec- tides. Here, the peptides-of-interest are synthesized with incor-
tion limit. porated stable isotopes and their sequences, such as 13C, 15N or
18
While these approaches allow the discovery-based quantifi- O. The label is usually introduced in the C-terminal amino acid
cation of thousands of PTMs across multiple samples, validation and therefore the y-ion series shows a shifted m/z. A SIL refer-
of interesting targets will be usually conducted with MS-inde- ence and the corresponding endogenous peptide share the
pendent technologies, for instance immunoassays such as same sequence and physicochemical properties, but differ by
ELISA. These are straightforward to implement and provide mass—ideally by at least 4 Da. Thus, they have the same dose/
high throughput, but suffer from low to medium multiplexing response rates in MS. Consequently, a well-defined amount (e.g.
capacities and are relatively expensive [95], although substantial 50 fmol) of SIL reference peptide can be spiked into a sample,
improvements have recently been achieved for antibody-based allowing precise absolute quantitation of endogenous peptide
assays. For instance Treindl et al. have developed a complex (and consequently protein) amounts/concentrations by compar-
workflow that allows antibody-based quantitation of hundreds ing their signals (i.e. intensity or area under the curve) over
of proteins from a single sample [96]. Proteins are first sepa- several orders of magnitude, down to amol amounts on column.
rated by gel electrophoresis, blotted on a membrane and bio- The usage of SIL peptides further increases the confidence in
tinylated. Next, the membrane lane is cut into 96 bands and peptide identification, as the SIL and endogenous peptides co-
proteins are eluted in a 96-well plate. Next, Neutravidin-coated elute and have identical fragmentation patterns, after the inten-
and differentially color-coded Luminex beads are added to each tionally-introduced mass shift has been taken into account. The
well, so that different colors represent the different molecular key to precise protein quantitation using SIL reference peptides
weight areas of the original gel. The beads can be pooled and is the selection of appropriate peptides, that are proteotypic and
small aliquots can be incubated with hundreds of antibodies not prone to degradation or modifications artificially induced
when using 5-20 μg of protein, comparable to a standard during sample preparation.
Western blot analysis. A Phycoerythrin-labeled secondary anti- Once set up and optimized, MRM and PRM enable a highly
body is used for signal generation, and the samples are ana- sensitive quantitation of 100s of peptides per LC-MS run in
lyzed on a flow cytometer, generating discrete signals for 96 ‘scheduled mode’, which relies on reproducible chromatogra-
bead populations which can be used for visualization. phy with stable retention times. For targeted MS, peptides-of-
Still, completely relying on antibody specificity can be an interest are typically evaluated for their suitability, namely
issue, even for validated antibodies, and even more so for PTM- detectability, stability, and uniqueness [100]. Well-generated,
specific antibodies (Figure 5). Notably, antibodies are not even quality controlled, and validated targeted assays [64] enable
available for many candidates of interest that appear in system- the analysis of larger sample cohorts in accordance with the
wide quantitative PTM-centric discovery studies. Therefore, demands of clinical research, including good inter-laboratory
recently, MRM and PRM (discussed in detail elsewhere [97-99]), reproducibility and precision [101]. To this end, more than 1500
have gained increasing attention as powerful alternative characterized targeted assays are available through the CPTAC
Assay Portal [102] (https://proteomics.cancer.gov/assay-portal),
including >200 for phosphopeptides.
If the sensitivity of MRM/PRM is not sufficient to quantify
the peptide of interest from whole cell or tissue lysate, as
often the case for modified peptides, these methods can be
combined with fast enrichment procedures to reduce the
huge background of non-modified peptides [103,104]. If 90%
of the non-modified background can be removed, a larger
proportion of the PTM-proteome of interest can be loaded
on-column to improve sensitivity. If this is still not sufficient,
combining anti-peptide antibody immunoprecipitation (IP)
with MS, such as ‘Stable Isotope Standards and Capture by
Anti-Peptide Antibodies’ (SISCAPA) [105-107] and immuno
MALDI (iMALDI) [108-111], or TXP antibodies [112] that recog-
nize peptides with a common epitope of 3-4 amino acids at
Figure 5. Antibody specificity as an issue. Depicted are a full length anti-Zyxin the N- or C-terminus, prior to (LC-)MS analysis can consider-
antibody and two antibodies against phosphorylated Zyxin (p-Zyxin) from
different vendors showing poor specificity. Importantly, there can be substantial ably improve the accessibility of target peptides. The unique
differences between antibodies from different vendors. capabilities of MS can even compensate for low antibody
EXPERT REVIEW OF PROTEOMICS 523

specificity, while the use of SIL reference peptides allows 10 individuals with and without gastric disease [143]. Mucins were
absolute quantitation from bodily fluids, cells, and tissues isolated from frozen gastric specimen using isopycnic density
[42]. In a recent project which shows great promise for clinical centrifugation and pooling of mucin-containing fractions.
settings, Popp et al. used iMALDI (a technique that combines O-glycans were released and analyzed by LC-MS. The authors
anti-peptide antibody immuno-enrichment with on target elu- found a large diversity in gastric O-glycosylation patterns,
tion and MALDI MS) for the accurate quantitation of AKT1 and and concluded that the high individual variation may render it
AKT2 out of cell lysate, flash frozen and FFPE tissues [111]. difficult to identify cancer-specific patterns, though they did
Translation to clinical settings requires robust methods that observe higher sialylation and sulfation patterns on O-glycans
are precise, cheap and provide high throughput, among those identified from cancerous tissue. Hinneburg et al. reported a
immunoassays (given the previously mentioned limitations), novel approach to study N- and O-glycomes simultaneously
targeted MS with robust LC conditions, as well as SISCAPA from FFPE tissue [144]. Cells were isolated from FFPE tissue
[105-107] and iMALDI [108-111]. through laser capture microdissection, proteins were extracted,
blotted on PVDF membranes, and the N- and O-glycans were
sequentially released, followed by nano-LC-MS using porous gra-
6. Analyzing protein glycosylation and
phitized carbon columns. Using this method, the authors were
phosphorylation in clinical samples
able to characterize the N- and O-glycomes of tissue specimens,
Protein glycosylation and phosphorylation are the two most abun- and even to differentiate and relatively quantify structural isomers.
dant PTMs. While phosphorylation is generally reversible and Recently, Everest-Dass et al. used MALDI imaging to study
allows rapid signaling, glycosylation represents a wide range of N-glycans in ovarian cancer FFPE tissues [141]. N-linked glycans
heterogeneous sugar structures of different complexity, which can were first released and analyzed by porous graphitized carbon
make up more than 90% of a protein’s molecular weight and may LC-MS and the identified glycan profiles were then used to deter-
be reversible or irreversible. Phosphorylation and glycosylation mine the spatial distribution of released N-glycans on FFPE ovar-
play key roles in many clinically relevant processes such as immu- ian-cancer sections by MALDI imaging. Interestingly, the authors
nity [113,114], inflammation [115,116], or metastasis [117,118]. were able to detect differences between the tumor region which
Aberrant modification patterns can be linked to multiple diseases, had high levels of high mannose glycans, and the intervening
including Alzheimer’s disease [20,119-121], diabetes [122-125], stroma which had high levels of complex/hybrid N-glycans.
cancer [126-129], and cardiovascular diseases [130-132]. Both Compared to glycosylation, phosphorylation-based signal-
PTMs have been extensively studied by MS. However, while ing can be very rapid, dynamic, and easily altered during
numerous strategies for the direct analysis of phosphopeptides sampling and sample processing. Thus, strategies applicable
from complex samples after specific enrichment have been devel- to clinical samples were developed only after the develop-
oped, because of the enormous complexity of glycan structures ment of robust and highly sensitive strategies that allowed
and glycopeptide fragmentation patterns, in-depth analysis stra- the system-wide precise quantitation of phosphorylation
tegies for protein glycosylation usually employ the release of changes (extensively reviewed elsewhere [145-148]), and
glycans, so that either previously-glycosylated peptides (site map- therefore phosphoproteomics of clinical samples has recently
ping) or the glycan structures by themselves (without the peptide gained more attention. The first quantitative phosphoproteo-
backbone) can be analyzed in detail. To date, most studies on mics studies of patient samples included the temporal profil-
PTMs in clinical samples have focused on either protein phosphor- ing of phospho-signaling in human platelets obtained from
ylation or glycosylation, mostly with the goal of discovering bio- fresh blood donations, after specific treatment with inhibiting
markers (panels of proteins or affected pathways rather than and activating compounds. This analysis revealed proteins in
single proteins) and stratifying patients to improve risk assess- platelets that might be relevant for hemostasis and thrombo-
ment, precision medicine, and therapy guidance (reviewed sis [104]. Here, chemical stable-isotope labeling, multiplexing,
by [133,134]). and TiO2/HILIC-based phosphopeptide enrichment/fractiona-
For glycan profiling, different approaches have been applied to tion [149] were combined with nano-LC-MS to quantify thou-
the study of clinical samples, e.g. focusing on either on glycomes sands of phosphorylation sites from approximately 100 µg of
by LC-MS after glycan release and capture, or glycan spatial dis- protein per sample. Schweppe et al. used strong cation
tributions by MALDI imaging. MS-based profiling of glycans in exchange (SCX) chromatographic fractionation followed by
bodily fluids had already started in the 2000s [135-140], as those TiO2 phosphopeptide enrichment to quantify non-small lung
samples can be easily obtained in rather large amounts. In 2012, cancer (NSLC) tissue from human individuals using a super-
Alley et al. studied N-glycans in blood sera obtained from ovarian SILAC approach [150]. For this study, NSLC cancer cell lines
cancer patients and controls [141]. They were able to observe were grown in SILAC medium with heavy arginine and lysine
differences that allowed distinguishing patient samples from con- and were spiked in as reference standard, allowing the quan-
trols. Sonneveld et al. used nano-LC-MS to study aberrant glyco- titation of two individual tumor samples against the super-
sylation in autoimmune hemolytic anemia (AIHA) using a total of SILAC mix and thus against each other [151]. Dazert et al.
103 plasma samples [142]. In AIHA, red blood cells (RBC) are analyzed primary tumor and control non-tumor biopsies
destroyed by IgG anti-RBC autoantibodies, which can lead to from a sorafenib-treated hepatocellular carcinoma (HCC)
hemolysis. They were able to demonstrate aberrant galactosyla- patient, 7 weeks before treatment and 7.5 weeks after a non-
tion patterns in IgG1-anti-RBC autoantibodies. More recently, gly- effective treatment [152]. They used a super-SILAC standard
coproteomics has also been increasingly applied to the study of consisting of five different cell lines for normalization,
tissue samples. Thus, Jin et al. studied mucin o-glycosylation from SCX-based fractionation, and TiO2-based enrichment followed
524 R. MNATSAKANYAN ET AL.

by nano-LC-MS. A small portion of each SCX fraction was used approximately one quarter of the phosphoproteome showed
for global proteome analysis. The authors were able to rapid changes, many in critical cancer pathways related to
observe reduced phosphorylation of the Raf-Erk-Rsk pathway, stress response, transcriptional regulation, and cell death.
indicating a successful inhibition of the target kinase Raf, but This underscores the need for well-standardized and docu-
downstream targets did not show reduced phosphorylation. A mented sampling procedures for clinical proteomics. This
pathway analysis combining both proteome and phosphopro- was further confirmed by Gajadhar et al. who analyzed high-
teome data revealed the potential role of two processes in grade serous ovarian carcinoma tissue and colon adenocarci-
developing resistance: epithelial-to-mesenchymal transition, noma biopsies from five patients each [162]. After vessel liga-
and cellular adhesion molecule-dependent drug resistance. tion, specimens were surgically removed and the first biopsy
The validation of phosphoproteomic changes obtained from was taken immediately and snap frozen (0 min). Further biop-
discovery experiments in sample cohorts is still mainly done using sies were taken after 10, 30, and 60 min of cold ischemia.
antibodies. However, because of the aforementioned issues of Samples were labeled with the iTRAQ-4plex reagents, and
antibody availability and specificity (which is particularly true for phosphotyrosine (pTyr) peptides were enriched using a pTyr
phosphospecific antibodies, see (Figure 5)), a combination of phos- antibody mixture, followed by IMAC enrichment and LC-MS.
phopeptide enrichment and targeted MS is increasingly used Ten percent of each sample was further fractionated and
instead of antibody-based approaches. Targeted MS is flexible, analyzed by LC-MS for global proteome quantitation. The
can be multiplexed, and allows the highly specific and sensitive authors observed a rapid, unpredictable, and widespread
quantitation of phosphopeptides by single-shot LC-MS analyses, impact on phosphotyrosine patterns in which up to 50% of
e.g. after TiO2 [103,104] or IMAC-based enrichment [153,154]. In pTyr sites were changed more than 2-fold, in addition to
contrast to Western blot or ELISA, MS-based methods allow the observing a considerable spatial heterogeneity within the tis-
unambiguous connection between qualitative (peptide sequence sue. Notably, both spatial heterogeneity and the dynamic
and phosphorylation site) and quantitative information with high response to ischemia varied substantially between the differ-
precision, and, if SIL peptides are used, even absolute quantitation, ent patients. Huang et al. applied quantitative (phospho)pro-
including the assessment of phosphorylation levels (stoichiome- teomics to study 24 breast cancer-derived xenografts (PDX)
try). Thus, in order to quantify a specific phosphorylation-site of models [30]. They were able not only to confirm predicted
interest, PHD Ser125, in fresh frozen colorectal cancer biopsies, genomic targets, but also to find protein expression and
Di Conza et al. applied HPLC-based electrostatic repulsion-hydro- phosphorylation changes that could not be explained based
philic interaction liquid chromatography (ERLIC) [155,156] for com- on genomic data, showing the potential of MS-based proteo-
bined phosphopeptide enrichment and fractionation [157]. In mics for identifying novel drug targets.
ERLIC, the majority of the unphosphorylated peptides are found
in the flow-through and the early fractions [158], while phospho-
7. Analyzing endogenous proteolytic cleavage
peptides are efficiently retained [159] and separated according to
(proteolysis) in clinical samples
the number of phosphoamino acids [160]. Using a SIL reference for
the phosphopeptide AKPPADPAAAAsPCR, the authors first deter- Proteolysis is the irreversible cleavage of proteins into smaller
mined a retention-time window, which then allowed them to polypeptides, and is usually catalyzed by proteases. In addition
specifically collect the enriched phosphopeptide from only 32 µg to its role in protein degradation and apoptosis, proteolysis also
of total protein from the tissue samples. Absolute quantitation of affects protein localization, function, activity, and protein-
the phosphopeptide, and the non-phosphorylated counterpart in protein interactions. Aberrant proteolytic cleavage is connected
complete digest, using PRM, allowed them to determine the phos- to a variety of diseases, including cardiovascular and neurode-
phorylation stoichiometry in cancerous and healthy tissue samples generative diseases [163,164], inflammation and impaired
from ten individuals. Their results showed significantly reduced wound healing [165], as well as tumor metastasis [166]. This
phosphorylation levels in the cancer tissue, in line with the hypoth- can be caused by, for example, dysregulation of proteases, or
esis based on from cell culture experiments [157]. SNPs that suddenly render a protein a non-physiological sub-
CPTAC has conducted several relevant phosphoproteomics strate of a protease. To better understand the role of proteolysis
studies in cancer tissues. Mertins et al. addressed the afore- in disease, it is important to not only identify the substrates that
mentioned issue that tissue collection cannot always be stan- are cleaved under pathophysiological conditions, but also to
dardized, and consequently ischemic time can vary between determine where they are cleaved and how they are differen-
tissue samples collected for the same study [161]. This, how- tially regulated between healthy and diseased states. While the
ever, can alter phosphorylation levels of biopsy samples as the protease responsible for a specific cleavage might be a promis-
authors demonstrated. They collected human ovarian tumor ing drug target, its reliable identification is rather challenging
and breast cancer xenograft tissue from different patients and not straightforward. This may usually require cell culture-
without vascular interruption (0 min) and then allowed based differential control experiments either with specific pro-
defined ischemic time intervals of 5, 30, and 60 min. Samples tease inhibitors or the usage of specific protease knock outs.
were labeled with 4-plex iTRAQ, fractionated, and enriched for Analysis may be conducted by MS or by Western blot-based
phosphorylated peptides using IMAC. A total of ~3 mg protein migration shifts.
was used per experiment, 10% of which was used for global As is the case for phosphorylation and glycosylation, it can be
proteome quantitation. Interestingly, the global proteome and anticipated that specific proteolytic signatures indicate aberrant
most of the more than 25,000 phosphorylation sites did not cleavage, perhaps even by a specific protease, and—importantly
show significant changes after 60 min. However, —may serve as markers for diagnosis and treatment.
EXPERT REVIEW OF PROTEOMICS 525

Proteolysis leads to the formation of novel (‘neo’) protein Subtiligase labeling and N-terminal enrichment [170] was
N-termini and MS-based proteomics has become an indispen- used for enriching protein N-termini in plasma and serum
sable tool to identify these neo N-termini. This is preferentially obtained from healthy donors, in order to elucidate the roles
done through the system-wide specific enrichment of of proteases in blood. The authors identified 772 unique
N-terminal peptides, which include not only the original but N-terminal peptides from 222 proteins, ranging over six orders
also neo protein N-termini, followed by LC-MS. This approach of magnitude in abundance [177]. They were able to validate
has been referred to as N-terminomics. A variety of methods known proteolytic processes, including peptide cleavage in
have been developed for this purpose, among these are com- coagulation and complement activation, and also found a
bined fractional diagonal chromatography (COFRADIC) [167], large number of cleavages that had not previously been
terminal-amine isotopic labeling of substrates (TAILS) [168], reported. Recently, using the same method and combining
charge-based fractional diagonal chromatography discovery with subsequent quantitation by MRM, Witta et al.
(ChaFRADIC) [169], and subtiligase N-terminal enrichment analyzed plasma from patients after chemotherapy to detect
[170]. All of these methods include the specific labeling of proteolytic signatures released from apoptotic cells which may
protein N-termini prior to in vitro digestion in order to distin- serve as indicators of chemotherapy-induced cell death [178].
guish the specifically labeled N-terminal from internal pep- The experiment was performed in 1.5-mL cell-free plasma
tides. These methods allow the identification of protease samples from 5 different patients, which included two acute
substrates through the differential formation of neo myeloid leukemia (AML) patients and one patient with non-
N-terminal peptides between different conditions—usually, Hodgkin lymphoma (NHL) of diffuse large B-cell lymphoma
protease knockout vs. wild-type, but increasingly also patients subtype. The authors reported 153 proteolytic peptides which
vs. controls. Notably, the identification of neo N-terminal pep- may serve as an initial library of proteolytic biomarkers.
tides appearing under specific conditions directly reveals the Solari et al. applied quantitative iTRAQ-8plex-based ChaFRADIC
respective cleavage sites, and also allows the researcher to and (phospho)proteomics to platelets isolated from the only Scott
deduce consensus motifs. In addition to these direct strate- syndrome patient available worldwide. The Scott syndrome is a
gies, there are also more indirect methods such PROTOMAP, rare but likely underdiagnosed bleeding disorder associated with
which combines SDS-PAGE with LC-MS to globally identify mutations in ANO6 that lead to an impaired procoagulant
shifts in protein migration that are indicative of proteolytic response [179]. From only 20 µg of protein per sample, they
processing [171]. This method, however, cannot readily reveal quantified 1596 N-terminal peptides between activated patient
cleavage sites or motifs. In general protease inhibitors should and control platelets, 180 of which were confirmed as calpain-
be used during sample homogenization/lysis, especially in regulated (corresponding to 106 proteins) and 23 (corresponding
case of tissues, to ensure that ongoing endogenous protease to 23 proteins) were confirmed as caspase-regulated. The authors
activity does not alter the cleavage patterns. detected reduced calpain-dependent cleavage of cytoskeleton-
To date, there are still only few studies that have applied linked and signaling proteins in the Scott patient, in agreement
quantitative N-terminomics to patient-derived samples; in the with increased phosphorylation states. While in this study, the
large majority of studies, these techniques have been used to high sensitivity was achieved using a rather elaborate HPLC-
study fundamental and clinically relevant processes in cell cul- based ChaFRADIC workflow, the authors have recently developed
ture or animal models. Lange et al. used TAILS and LC-MS to a tip-based improvement of the procedure that allows quantita-
study erythrocytes obtained from 6 mL of whole blood per tive N-terminomics with high sensitivity [180].
healthy donor. The extremely high abundance of hemoglobin
in erythrocytes makes their proteomic analysis quite challenging.
8. Analyzing oxidative Cys modifications in clinical
Using TAILS and thus specifically focusing on protein N-termini
samples
allowed the researchers to substantially reduce sample complex-
ity in order to improve the detection of low-abundance proteins. A high diversity of protein PTMs are formed as a result of
In this way, the authors were able to identify 1369 N-terminal direct or indirect interaction with reactive oxygen/nitrogen
peptides from 1234 proteins, 281 of which were novel erythro- species (ROS/RNS) [181,182]. Small reactive radical or non-
cyte proteins while six previously-missing proteins [172,173] radical ROS/RNS, such as superoxide (O2·−2) produced by the
were identified by MS for the first time [174]. The same group NAD(P)H oxidase family, and nitrogen monoxide (NO) pro-
also analyzed human dental pulp as a unique tissue that could duced by nitric oxide synthases (NOS), are byproducts of
lead to the identification of additional missing proteins, and cellular respiration [183] and primarily form redox modifica-
found that most of the identified N-termini represented proteo- tions on cysteine (Cys), due to the high reactivity of its thiol
lytic cleavage sites [175]. In another study, TAILS was used to side chain [182,184]. Reversible cysteine redox PTMs include
compare B cells from the only known living patient worldwide S-nitrosylation (SNO), S-sulfenylation (SOH), disulfide forma-
with a genetic MALT1 deficiency, which leads to immunodefi- tion (S–S), S-glutathionylation (SSG), S-cysteinylation (S-Cys),
ciency and immune dysregulation, with heterozygous family and S-sulfhydration (SSH), while sulfinylation (SO2H) and
members presenting no immunological disorders. MALT1 is a sulfonylation (SO3H) are generally considered to be irrever-
paracaspase that plays an important role in regulating lympho- sible [181,184]. Cys modifications are key regulators of cel-
cyte responses in NF-κB activation. The authors were able to lular redox homeostasis [185]. At low ROS levels, reversible/
identify HOIL1, a member of the linear ubiquitin chain assembly protective PTMs are formed that are associated with cell
complex, as a novel MALT1 substrate, inhibiting reactivation of proliferation, differentiation, and redox signaling, while
the NF-κB pathway [176]. increasing ROS levels result in irreversible modifications
526 R. MNATSAKANYAN ET AL.

and damage to proteins [186]. Importantly, specific redox- tags (ICAT) [200]. These assays include blocking of the free
reactive Cys residues can be present in their reduced forms thiols, followed by selective reduction of modified cysteines
or may compete for different types of redox modifications, and introduction of functional groups enabling stable-adduct
making them into ‘redox-switches’ which sense the redox formation, enrichment, and (preferably) also quantitation of
microenvironment and allow reversible and—if required— the switched PTMs. Using a IodoTMT switch assay, including
irreversible fine tuning of biochemical processes [181,185]. specific reduction of SNO by sodium ascorbate, Pan et al.
ROS-mediated signaling is involved in many key metabolic identified 12 significantly regulated SNO proteins in hypoxic
processes, and affects a wide variety of upstream and cardiomyocytes [201]. Another modified BST, disulfide
downstream protein targets, thereby influencing the regula- exchange based RAC, showed high specificity (~95 %) and
tion of other PTMs such as phosphorylation, acetylation, and superior sensitivity for proteins higher than 100 kDa compared
ubiquitination [88,187,188]. to biotin-based enrichment [199,202]. Using another RAC
Although the functional relevance of Cys oxidative PTMs in approach in combination with iTRAQ labeling, Forrester et al.
human diseases, such as cancer [189], cardiovascular [188,190], demonstrated the progressive decay of SNO signal over time
inflammatory [191], and neurodegenerative diseases [191,192] as a result of CysNO treatment of HEK293 cells. Roughly 300
is clear, the molecular mechanisms of redox signaling still negatively-regulated SNO sites were quantified [202], possibly
remain unclear. A number of proteomics techniques have indicating the presence of regulatory mechanisms for removal
been developed to study the targets and modulations of of SNO or exchange of the transient SNO to more stable
redox signaling [193]. Because redox proteomics is still an disulfide [203]. A modified form of ICAT: SNOxICAT, allowed
emerging field with continuing method development, for quantitative analysis of SNO site occupancy in vivo and
research has mainly focused on basic method development demonstrated that the exposure to NO2 leads to widespread
and the application of the newly-developed methods to cell SNO formation only in ischemic mouse hearts, but not in
culture and animal models [193,194]; only a few redox pro- ischemia or NO2 exposure solely [204]. Ibáñez-Vea et al.
teomics studies have involved human subjects [195,196]. A applied titanium dioxide (TiO2) chromatography enrichment
major challenge in transferring redox proteomics methods to after SNO reduction by ascorbate followed by labeling with
clinical samples is the necessity for blocking free Cys residues cysteine specific phosphonate adaptable tag (CysPAT) and
and preserving endogenous levels as much and as soon as identified high number of SNO proteins (569 endogeneous
possible. In contrast to purely enzymatically driven PTMs, Cys and 795 chemically induced) in macrophages [205].
redox states can even change in denatured samples and dur- SOH is a highly reactive and unstable modification [206].
ing sample preparation, which means that protocols have to Depending on the local redox-environment, it can be
be specifically optimized to reduce the occurrence of new further converted to other reversible and irreversible
(and exchange of existing) redox modifications. This involves cysteine PTMs. As an intermediate form of cysteine oxida-
avoiding exposure to UV light and atmospheric oxygen, avoid- tion, it is a key player in redox signaling [207]. Notably, the
ing lysis of the cells before the addition of blocking reagent, unique reactivity of SOH to dimedone compounds allows its
avoiding common pathogen inactivation procedures [197] etc. use for direct labeling in living cells. Thus, stable isotope
While this is already complicated for cell culture and requires coded alkyne-functionalized dimedone (DYn-2 and d6-DYn-
careful use of controls, it represents a major challenge for 2) probes enable the enrichment and relative quantitation
clinical samples, particularly tissues. In the following section, of SOH peptides. In a recent study of human RKO cells,
we will briefly discuss the most relevant Cys redox modifica- about 1000 SOH-modified Cys sites from more than 700
tions, their roles in health and disease, and the current state of proteins were identified in response to epidermal growth
proteomic investigations. factor (EGF) stimulation and H2O2 treatment, from which
The important role of NO in cardiac function and neurotrans- 215 and 360 SOH sites could be quantified, respectively
mission has been suggested by multiple studies [186,188,192], [208]. Indirect analysis of SOH by modified BST and specific
and the cardioprotective potential of S-nitrosylation has been reduction of SOH by arsenite was used for the study of a
attributed to its ability to shield thiol groups from irreversible SILAC-labeled THP-1 human monocytic cell line treated with
oxidation [186-188]. The neuronal isoform of NOS produces NO H2O2 or activated human platelet releasate. More than 200
in the brain, which was first thought to be a neurotoxin associated SOH peptides were quantified [209].
with neurodegenerative diseases. Recent studies, however, also One of the important functions of cellular disulfide bonds is
indicate the protective function of SNO in neurotransmission and the stabilization of protein structure via intramolecular and inter-
synaptic plasticity [192]. molecular covalent linkages [210]. Nevertheless, specific Cys resi-
Direct analysis of SNO is highly challenging as it is not only dues form redox-active disulfides as a result of reduction by
present at low levels, but—more importantly—also labile. oxidoreductases, interaction with small-molecule thiols such as
Currently, phenylmercury-resin-based enrichment approaches glutathione (GSH) and free cysteine, or by replacement of less-
allow the direct capture of SNO via formation of stable thiol- stable SNO/SOH. Allosteric redox-active disulfides induce a
Hg bonds. Using this technique Raju et al. identified 269 functional change in the protein and are involved in redox
endogenous SNO sites in wild-type mouse brain [192]. A signaling [210]. A recent study has demonstrated the role of
broad spectrum of indirect methods to enrich and analyze redox-active S-S in cancer: In vascular endothelial growth factor
SNO and other cysteine PTMs utilize different types of mod- receptor (VEGFR2), Cys1199 and Cys1206 form an intra-molecular
ified biotin switch techniques (BST) such as iodoTMT [198], disulfide bond at elevated ROS levels, suppressing VEGFR2 activ-
resin-assisted capture (RAC) [199], and isotope-coded affinity ity [189]. Importantly, knockdown of the key antioxidant enzyme
EXPERT REVIEW OF PROTEOMICS 527

peroxiredoxin III resulted in inactivation of VEGFR2 and repres- second alternative cohort of the same size for the validation
sion of tumor angiogenesis in vivo [211]. phase, both in a non-targeted rather than targeted mode [218].
Proteome-wide analysis of intact disulfides represents a sig- Obtaining well-classified samples for a sufficient number of
nificant challenge. For analysis of native disulfides, Lu et al. used patients and matched controls that have been collected under
a combination of proteases, including Lys-C, trypsin, subtilisin, well-defined conditions that are compatible with downstream
Glu-C, elastase, proteinase K, and/or Asp-N for protein diges- proteomic analysis, is often a bottleneck. However, beside
tion, extensively fractionated the resulting digests, performed stratification sample size is a major determinant of the relia-
LC-MS, and used their pLink-SS data analysis software for the bility of the biomarkers and targets which have been identi-
identification of cross-linked peptides. In this way, more than fied in clinical samples, as sample size directly affects the
550 disulfide bonds were mapped in the secretome of human statistical power [219,220]. Skates et al. calculated the prob-
umbilical vein endothelial cells (HUVECs) [212]. abilities for a biomarker passing through the biomarker pipe-
SSG has been suggested to provide more persistent protec- line from discovery to validation, and to then clinical
tion against oxidative damage than transient SNO or SOH. validation as a function of the number of biospecimens and
Murdoch et al. showed that SSG is increased in ischemic for different scenarios [221]. In this study, they hypothesized
muscle even several days after hindlimb ischemia, and that it that approximately 8000 proteins would be quantified during
facilitates revascularization [213]. SSG of specific cysteines in discovery, leading to 20/50/100 candidates for measurement
the upstream and downstream targets of VEGFR2 were shown in the validation state, and then 2/5/10 candidates for mea-
to considerably increase cardiac angiogenesis [187]. As there surement in the clinical validation state. Thus, if 10 controls are
are no direct methods available for the analysis of endogen- compared against 10 patients during discovery, the likelihood
ous SSG, most studies are based on modified BST and the that an authentic biomarker present in 50% of the cases with a
specific reduction of SSG by glutaredoxin [214,215]. median distance of 3 standard deviations is passed to the
The free intracellular cysteine concentration of ~100 µM is validation step is only 15% when 20 targeted assays are
the source of S-Cys [210]. An analysis of the cysteinylation of performed. In the case where 50 (100) targeted assays are
human serum albumin (HSA) at Cys34 in human plasma sam- performed, this likelihood increases to 24% (35%). However,
ples obtained from 229 patients (139 of whom had liver disease, increasing the number of samples has a much stronger effect.
38 of whom had kidney disease, and 52 of whom had diabetes Under the aforementioned conditions, with 20 targeted assays
mellitus), showed a strong correlation of increased S-Cys34 with and 25 (50) patients and controls, the likelihood increases to
these three oxidative stress-related diseases [195]. 60% (93%).
All these examples clearly demonstrate the high relevance These theoretical considerations underline the importance
of Cys modifications for human health, and the need to further of cohort size for biomarker discovery, but sample normal-
improve the sensitivity and robustness of current methods so ization and statistical tests to actually identify potential bio-
that they can be applied to clinical samples in the future. As markers in the discovery phase are also relevant. The
demonstrated for phosphorylation, it can be anticipated that conventional two-sided t-test to estimate the probability that
additional information on Cys redox status will be another a value is significantly different is not sufficient—if many
important layer of information to stratify patient samples, individual tests are conducted (i.e. for thousands of proteins),
and may lead to important markers of disease. a substantial share will yield significant p-values just by chance
(false positives) [222]. Therefore, corrections such as Benjamini
Hochberg are typically applied in order to take false discovery
rates into account for multiple comparisons. In addition to the
9. Study design for biomarker discovery
statistical significance, the level of regulation must also be
With the continuous advancement of proteomic instrumenta- considered. A statistically highly significant 1.1-fold regulation
tion, methods, algorithms, and data analysis strategies, there may not be relevant—the median standard deviation of the
has been also a paradigm shift with regard to study design. In entire dataset needs to be considered, too, in order to apply
the early days of clinical proteomics, two-dimensional PAGE was realistic cutoffs that take into account the quality of the indi-
a widely-used but tedious and limited technology (with regard vidual dataset (e.g. 3 times the standard deviation as in the
to quantitation, precision and coverage), and studies usually aforementioned example from Stakes et al.).
included only a few patients; nowadays, there is a general Sometimes sample size is inherently limited, for instance in
trend for the application of sophisticated gel-free workflows case of rare disorders with low prevalence, rendering the
to larger cohorts. Cross-sectional comparisons between differ- estimation of the variance uncertain. Nevertheless, such rare
ent cohorts are complemented more and more by longitudinal disorders may still be analyzed by proteomics [223]. For small
studies that follow patients over time [216]. Particularly for sample sizes, the t-test may consider that a potential marker
biofluids which tend to show a large variation between showing strong regulation but (concurrently) high intra-group
patients, this is a promising strategy for the identification bio- variance is not significant [224] (Figure 6). Large intra-group
marker panels. Still today, most studies follow the ‘biomarker variations can particularly appear between patients. Thus, one
pipeline’ and include smaller initial cohorts for identifying strategy to compensate for the inaccuracy of the estimation of
potential markers and then expand this to larger cohorts for variance in small sample sizes is ‘empirical Bayes method
validation [217]. If sufficient instrumentation and computational shrinking’ [224] which uses a moderated t-test where the
power are available, Geyer et al. propose an alternative strategy, p-value is assessed by the fold-change rather than the initially
which utilizes a large cohort in the discovery phase, and a estimated variance. Notably, empirical Bayes methods have
528 R. MNATSAKANYAN ET AL.

Figure 6. Limits of t-test based statistics for small sample sizes. (A) Two simulated potential markers 1 and 2. Although the first marker shows a considerable difference
between patients and controls, the strong intra-group variance leads to a poor p-value. In contrast, the second marker yields a low p-value and might be considered as
statistically significant, but the difference between patients and groups is negligible. (B) Volcano plot of the two markers in the background of an iTRAQ-based
phosphoproteomics experiment based on a t-test. (C) Volcano plot of the same data based on the moderated t-test provided in the Limma package [226-228]. Only marker
1 is actually significant. A detailed description for the use of this package was recently published by Kammers et al. [224]. (Figure reprinted with permission, Pagel et al., Expert
Rev Proteomics 2015).

been successfully applied in the microarray field for approxi- search for specific PTM signatures of disease is a promising com-
mately 10 years [225] and can be adapted for use in proteo- plementary strategy, as demonstrated for potential proteolytic
mics studies [224,226,227]. signatures in postchemotherapeutic blood [178], for example.
Despite these promising alternative data analysis strategies, Compared to genomics and transcriptomics, proteomic work-
the major important determinant of successful biomarker dis- flows are still considerably more challenging and less standar-
covery still remains: The most sophisticated data analysis stra- dized. Thus, a general transfer of discovery proteomics workflows
tegies cannot compensate for poor sample or data quality. into routine clinics might be unrealistic in the near future.
There is a continuously increasing number of clinical proteo- Nevertheless, interdisciplinary collaborations between clinicians,
mics studies facing these issues, but there are also continuous proteomics researchers, biochemists, bioinformaticians, and sta-
improvements in available methods and instrumentation. tisticians can provide important and novel insights into disease
Thus, it can be anticipated that improved statistical methods mechanisms, biomarkers, and therapeutic targets. Pilot projects
and tools will be developed to improve the detection and such as CPTAC have made great progress in standardization and
verification of biomarker candidates in proteomics studies. identifying potential pitfalls for in-depth (phospho)proteomics
analyses of cancer tissues [102,161], and different cancer tissues
have been analyzed using proteogenomic workflows, leading to
10. Expert commentary
the relative quantitation of thousands of proteins and phospho-
In recent years (i) technologies for proteomic sample preparation, peptides [30,32].
PTM enrichment, chromatographic separation, and (ii) LC and MS In addition to discovery, the transfer of MS-based assays into
instrumentation, as well as (iii) data analysis strategies, have been the clinic is an exciting opportunity. In cases where, for example
considerably improved. Concurrently, there has been an increas- current immunohistochemistry assays have limited predictive
ing awareness of the importance of quality control and standardi- value, technologies such as MALDI imaging or iMALDI may be
zation in MS-based proteomics in order to produce more reliable promising alternatives for the future [111,231]. Standardized
and robust quantitative data. Particularly for phosphorylation, MRM and PRM assays can be used to quantify defined sets of
glycosylation, proteolytic cleavage, and Cys modifications, highly proteins and modified peptides. For phosphopeptides, this has
sensitive methods have been developed that can even be applied already been demonstrated, but existing workflows are still too
to limited sample amounts, as often the case for biopsies. complex and time-consuming. Thus, Di Conza et al. were able
Consequently, an increasing number of clinical proteomics studies to use a tailored PRM workflow for absolute quantitation of
that focus on PTMs are emerging. Combining different and com- phosphorylation levels for PHD2 Ser125 in colorectal cancer
plementary layers of information, e.g. changes in (i) protein abun- biopsies [157]. Although this is an exciting example which
dance and PTM levels, (ii) differences in PTMs, or (iii) changes in the demonstrates the unique capabilities of MS for the absolute
genome, transcriptome, or proteome (proteogenomics [229,230]) quantitation of phosphorylation levels from small amounts of
holds great potential for discovering important drivers and mar- tissue, the analysis required a specific ERLIC enrichment step;
kers of disease. Mere knowledge of protein abundance without although this could probably be automated, it still might be too
spatial information and without full isoform resolution (both of laborious to transfer into clinics. Also, it will be very important
which are missing in conventional proteomics studies) might, in to ensure that such workflows work for FFPE samples. An
some cases, be insufficient or even misleading. Therefore, the alternative indirect workflow for determining phosphorylation
EXPERT REVIEW OF PROTEOMICS 529

stoichiometry is by spiking in the non-phosphorylated SIL refer- well as for rare diseases where the increase in the number
ence peptide and splitting the sample in two aliquots, one of of available patients is even more critical.
which treated with phosphatase [232]. Individual measurement In summary, MS has just recently began to reveal its real
then allows assessment of phosphorylation stoichiometry. potential for clinical research, and the near future will be an
However, phospo-isomers cannot be distinguished after phos- exciting era where researchers will have the opportunity to
phatase treatment. demonstrate that the unique capabilities of MS-based pro-
teomics can be translated into clinical environments in
order to improve human health and healthy aging.
11. Five-year perspective
The increasing awareness of quality measures in the field of Key issues
proteomics, initially with regard to reliable peptide and pro-
tein identification through the implementation of false discov- ● To discover PTM signatures in clinical samples that may
ery rate assessments [233] and site localization algorithms [72], serve as biomarkers or targets, it is vital to have an optimal
but (importantly) also for system performance [50] and more and elaborate study design, including the use of matching
precise quantitation, have been critical milestones in the move controls and thorough stratification of patient and control
from cell biology-driven proteomics to clinical proteomics. It samples. Including inappropriate samples may lead to
can be anticipated that instrumentation will further improve in increased intra-group variation and therefore could mask
the next 5 years, leading to more robust, more rapid, and potential markers. On the contrary, having well-character-
more sensitive LC-MS analyses with higher resolution. (Semi-) ized samples may allow reducing the number of samples.
automated and parallelized sample preparation workflows are ● Also, to keep preanalytical variance as low as possible it is
increasingly being developed and applied to reduce technical important to strictly follow elaborate SOPs during sampling
variation and increase analysis throughput. These develop- and sample processing. Ideally the sample collection should
ments will have an impact on DDA, DIA, and targeted meth- be commonly planned by clinicians and proteomics experts
ods, and will further improve the coverage, precision, and together and all steps should be followed strictly. In case
reproducibility of MS-based identification and quantitation. small deviations from agreed-upon SOPs cannot be
In this way, precise and deep proteomic profiling will become excluded, these should be well-documented so that later
less time-consuming and more cost-effective, facilitating the inconsistent results may be correlated with such changes.
implementation of studies on large sample cohorts as well as ● PTM enrichments procedures should first be tested and
complex longitudinal studies. It will be important that data optimized for sensitivity and robustness using reference
analysis pipelines are able to match the speed of the experi- samples such as HeLa or other cell culture models. As
mental progress, particularly regarding data amounts, compre- these procedures are often complex, the use of chemical
hensiveness, and complexity. In addition to label-free analysis labeling procedures that allow multiplexing prior to enrich-
of samples, the increased multiplexing capacities of NeuCode ment should be considered in order to reduce technical
will allow multi-level analysis of PTMs even from clinical variation. Additionally, multiplexing may be used to
samples [234]. As instrumentation for targeted MS is compara- improve sensitivity as in total more sample material can
tively robust, inexpensive, and widespread, it can be antici- be used for the analysis.
pated that clinics will increasingly consider establishing MS- ● For data analysis, it is important to consider how to deal
based assays for the quantitation of proteins and PTM levels with missing values. The selection of interesting targets
[217]. MALDI imaging also holds a great potential for improv- should not be solely based on p-values and should addi-
ing or complementing current histopathology, with the ulti- tionally require reasonable fold-changes that take into
mate goal of having powerful algorithms that can help with account the standard deviation and distribution of the
diagnosis [235]. As a rapid and straightforward approach, whole dataset at hand.
iMALDI may allow the MS-based precise quantitation of pro- ● The validation of targets identified in the discovery phase
tein panels that may help to stratify patients [110,111]. should include a larger independent sample cohort, ideally
Large multinational consortium-driven proteogenomic collected using the same SOPs. The use of solely antibody-
studies will increasingly exploit the unique possibilities of based assays has to be carefully considered, as particularly
synergizing genomic, transcriptomic, proteomic, metabolo- modification-specific protein antibodies often show poor
mic, and lipidomic datasets [236]. Thus, the ‘cancer moon- specificity. More promising alternatives may be (i) targeted
shot’ program initiated by former US vice-president Joe LC-MS in case of moderate to high-abundance targets using
Biden [237,238] led to the formation of the International whole lysates and (ii) after enrichment e.g. of phosphopep-
Cancer Proteogenomics Consortium (ICPC), bringing tides, or (iii) using a combination of anti-peptide antibodies
together ‘some of the world’s leading cancer and proteo- and (targeted) MS such as SISCAPA and iMALDI.
genomic research centers’ in order to improve our under- Compatibility with FFPE tissue samples should be evaluated
standing of cancer. Such large worldwide consortia hold a thoroughly.
great potential, as they enable access to larger patient ● For transfer into the clinic promising assays should be
cohorts, complementary technologies, and improved data straightforward, robust, ideally fast and precise and there-
sharing. It may be expected that similar large coordinated fore should have potential for (partial) automation and
efforts will be undertaken for other widespread diseases, as provide a simple and clear read-out.
530 R. MNATSAKANYAN ET AL.

Funding 10. Benayoun BA, Veitia RA. A post-translational modification code for
transcription factors: sorting through a sea of signals. Trends Cell
The authors R. Mnatsakanyan, G.Shema, A. Sickmann were supported by the Biol. 2009;19(5):189–197.
Ministerium für Innovation, Wissenschaft und Forschung des Landes 11. Gu B, Zhu WG. Surf the post-translational modification network of
Nordrhein-Westfalen, the Senatsverwaltung für Wirtschaft, Technologie und p53 regulation. Int J Biol Sci. 2012;8(5):672–684.
Forschung des Landes Berlin, and the Bundesministerium für Bildung und 12. Venne AS, Kollipara L, Zahedi RP. The next level of complexity: crosstalk
Forschung. C.H. Borchers and R.P.Zahedi are grateful to Genome Canada and of posttranslational modifications. Proteomics. 2014;14(4–5):513–524.
Genome British Columbia for financial support (project codes 204PRO for 13. Solari FA, Dell’Aica M, Sickmann A, et al. Why phosphoproteomics
operations and 214PRO for technology development). CHB is also grateful for is still a challenge. Mol Biosyst. 2015;11(6):1487–1493.
support from the Leading Edge Endowment Fund (University of Victoria) and 14. Ren J, Jiang C, Gao X, et al. PhosSNP for systematic analysis of
for support from the Segal McGill Chair in Molecular Oncology at McGill genetic polymorphisms that influence protein phosphorylation.
University (Montreal, Quebec, Canada). C.H.Borchers, A.Spatz, M.Basik and Molecular & cellular proteomics: MCP. 2010;9(4):623–634.
G.Batist are grateful for support from the Warren Y. Soper Charitable Trust 15. Ruggles KV, Krug K, Wang X, et al. Methods, Tools and Current
and the Alvin Segal Family Foundation to the Jewish General Hospital Perspectives in Proteogenomics. Molecular & cellular proteomics:
(Montreal, Quebec, Canada). A.Spatz, M.Basik and G.Batist are also grateful MCP. 2017;16(6):959–981.
for support from Genome Canada's Business-Led Networks of Centres of • Excellent overview of the state-of-the art in proteogenomics.
Excellence progran, Exactis Innovations, and the Fonds de recherche du 16. Slamon DJ, Clark GM, Wong SG, et al. Human breast cancer: corre-
Québec - Santé, Axe cancer du sein et de l'ovaire (FRSQ-ACSO). lation of relapse and survival with amplification of the HER-2/neu
oncogene. Science. 1987;235(4785):177–182.
17. Yuste L, Montero JC, Esparis-Ogando A, et al. Activation of ErbB2 by
overexpression or by transmembrane neuregulin results in differ-
Declaration of interest ential signaling and sensitivity to herceptin. Cancer Res. 2005;65
C.H.Borchers is the Chief Scientific Officer of MRM Proteomics, Inc. The (15):6801–6810.
other authors have no other relevant affiliations or financial involvement 18. Paez JG, Janne PA, Lee JC, et al. EGFR mutations in lung cancer:
with any organization or entity with a financial interest in or financial correlation with clinical response to gefitinib therapy. Science.
conflict with the subject matter or materials discussed in the manuscript 2004;304(5676):1497–1500.
apart from those disclosed. 19. Klootwijk ED, Reichold M, Helip-Wooley A, et al. Mistargeting of
peroxisomal EHHADH and inherited renal Fanconi’s syndrome. N
Engl J Med. 2014;370(2):129–138.
20. Ohno M. Roles of eIF2alpha kinases in the pathogenesis of
Reviewer disclosures Alzheimer’s disease. Front Mol Neurosci. 2014;7:22.
21. Seeling M, Bruckner C, Nimmerjahn F. Differential antibody glyco-
Peer reviewers on this manuscript have no relevant financial or other sylation in autoimmunity: sweet biomarker or modulator of disease
relationships to disclose. activity? Nat Rev Rheumatol. 2017;13(10):621–630.
22. Sherrill JD, Stropes MP, Schneider OD, et al. Activation of intracel-
lular signaling pathways by the murine cytomegalovirus G protein-
ORCID coupled receptor M33 occurs via PLC-{beta}/PKC-dependent and
-independent mechanisms. J Virol. 2009;83(16):8141–8152.
Christoph H. Borchers http://orcid.org/0000-0003-2394-6512
23. Powers AD, Palecek SP. Protein analytical assays for diagnosing,
Albert Sickmann http://orcid.org/0000-0002-2388-5265
monitoring, and choosing treatment for cancer patients. J Healthc
René P. Zahedi http://orcid.org/0000-0002-4960-5460
Eng. 2012;3(4):503–534.
24. Chandler K, Goldman R. Glycoprotein disease markers and single pro-
tein-omics. Molecular & cellular proteomics: MCP. 2013;12(4):836–845.
25. Van Scherpenzeel M, Willems E, Lefeber DJ. Clinical diagnostics and
References
therapy monitoring in the congenital disorders of glycosylation.
Papers of special note have been highlighted as either of interest (•) or of Glycoconj J. 2016;33(3):345–358.
considerable interest (••) to readers. 26. Abdel-Kahaar E, Kabakchiev M, Hartmann B, et al. Performance of a
1. UniProt C. UniProt: a hub for protein information. Nucleic Acids Res. phosphoflow assay to determine phosphorylation of S6 ribosomal
2015;43(Database issue):D204–212. protein as a pharmacodynamic read out for mTOR inhibition. Clin
2. Hornbeck PV, Zhang B, Murray B, et al. PhosphoSitePlus2014: Biochem. 2016;49(15):1181–1187.
mutations, PTMs and recalibrations. Nucleic Acids Res. 2015;43 27. Rahbar S, Blumenfeld O, Ranney HM. Studies of an unusual hemo-
(Database issue):D512–520. globin in patients with diabetes mellitus. Biochem Biophys Res
3. Vaudel M, Burkhart JM, Zahedi RP, et al. PeptideShaker enables Commun. 1969;36(5):838–843.
reanalysis of MS-derived proteomics data sets. Nat Biotechnol. 28. Selvin E, Steffes MW, Zhu H, et al. Glycated hemoglobin, diabetes,
2015;33(1):22–24. and cardiovascular risk in nondiabetic adults. N Engl J Med.
4. Lemeer S, Heck AJ. The phosphoproteomics data explosion. Curr 2010;362(9):800–811.
Opin Chem Biol. 2009;13(4):414–420. 29. Schwarz UR, Geiger J, Walter U, et al. Flow cytometry analysis of
5. Zhao Y, Brickner JR, Majid MC, et al. Crosstalk between ubiquitin intracellular VASP phosphorylation for the assessment of activating
and other post-translational modifications on chromatin during and inhibitory signal transduction pathways in human platelets–
double-strand break repair. Trends Cell Biol. 2014;24(7):426–434. definition and detection of ticlopidine/clopidogrel effects. Thromb
6. Hentschel A, Zahedi RP, Ahrends R. Protein lipid modifications– Haemost. 1999;82(3):1145–1152.
More than just a greasy ballast. Proteomics. 2016;16(5):759–782. 30. Huang KL, Li S, Mertins P, et al. Proteogenomic integration reveals
7. Ruan HB, Nie Y, Yang X. Regulation of protein degradation by therapeutic targets in breast cancer xenografts. Nat Commun.
O-GlcNAcylation: crosstalk with ubiquitination. Molecular & cellular 2017;8:14864.
proteomics: MCP. 2013;12(12):3489–3497. 31. Heist RS, Christiani D. EGFR-targeted therapies in lung cancer:
8. Hart GW, Greis KD, Dong LY, et al. O-linked N-acetylglucosamine: predictors of response and toxicity. Pharmacogenomics. 2009;10
the “yin-yang” of Ser/Thr phosphorylation? Nuclear and cytoplas- (1):59–68.
mic glycosylation. Adv Exp Med Biol. 1995;376:115–123. 32. Mertins P, Mani DR, Ruggles KV, et al. Proteogenomics connects
9. Creixell P, Linding R. Cells shared memory and breaking the PTM somatic mutations to signalling in breast cancer. Nature. 2016;534
code. Molecul sys bio. 2012;8:598. (7605):55–62.
EXPERT REVIEW OF PROTEOMICS 531

•• Impressive proof-of-concept proteogenomics study quantita- 53. Burkhart JM, Vaudel M, Zahedi RP, et al. iTRAQ protein quantification:
tively analyzing the proteomes and phosphoproteomes of 105 a quality-controlled workflow. Proteomics. 2011;11(6):1125–1134.
genomically annotated breast cancers. 54. Bittremieux W, Tabb DL, Impens F, et al. Quality control in mass
33. Salvagno GL, Danese E, Lippi G. Preanalytical variables for liquid spectrometry-based proteomics. Mass Spectrom Rev. 2017. Epub
chromatography-mass spectrometry (LC-MS) analysis of human ahead of print.
blood specimens. Clin Biochem. 2017;50(10–11):582–586. 55. Percy AJ, Mohammed Y, Yang J, et al. A standardized kit for auto-
34. Longo DL. Tumor heterogeneity and personalized medicine. N Engl mated quantitative assessment of candidate protein biomarkers in
J Med. 2012;366(10):956–957. human plasma. Bioanalysis. 2015;7(23):2991–3004.
35. Maes E, Mertens I, Valkenborg D, et al. Proteomics in cancer 56. Zolg DP, Wilhelm M, Yu P, et al. PROCAL: A set of 40 peptide
research: are we ready for clinical practice? Crit Rev Oncol standards for retention time indexing, column performance mon-
Hematol. 2015;96(3):437–448. itoring and collision energy calibration. Proteomics. 2017;17(21).
36. Burkhart JM, Vaudel M, Gambaryan S, et al. The first comprehensive 57. Pichler P, Mazanek M, Dusberger F, et al. SIMPATIQCO: a server-
and quantitative analysis of human platelet protein composition based software suite which facilitates monitoring the time course
allows the comparative analysis of structural and functional path- of LC-MS performance metrics on Orbitrap instruments. J Proteome
ways. Blood. 2012;120(15):e73–82. Res. 2012;11(11):5540–5547.
37. Skold K, Alm H, Scholz B. The impact of biosampling procedures on 58. Bereman MS. Tools for monitoring system suitability in LC MS/MS
molecular data interpretation. Molecular & cellular proteomics: MCP. centric proteomic experiments. Proteomics. 2015;15(5–6):891–902.
2013;12(6):1489–1501. 59. Sandin M, Teleman J, Malmstrom J, et al. Data processing methods
38. Aasebo E, Mjaavatten O, Vaudel M, et al. Freezing effects on the and quality control strategies for label-free LC-MS protein quanti-
acute myeloid leukemia cell proteome and phosphoproteome fication. Biochim Biophys Acta. 2014;1844(1Pt A):29–41.
revealed using optimal quantitative workflows. J Proteomics. 60. Mohammed Y, Percy AJ, Chambers AG, et al. Qualis-SIS: automated
2016;145:214–225. standard curve generation and quality assessment for multiplexed
39. Lalmahomed ZS, Coebergh van den Braak RR, Mh O, et al. targeted quantitative proteomic experiments with labeled stan-
Multicenter fresh frozen tissue sampling in colorectal cancer: does dards. J Proteome Res. 2015;14(2):1137–1146.
the quality meet the standards for state of the art biomarker 61. Aittokallio T. Dealing with missing values in large-scale studies: micro-
research? Cell tissue banking 2017;18(3):425–431. array data imputation and beyond. Brief Bioinform. 2010;11(2):253–264.
40. Ivanov AR, Colangelo CM, Dufresne CP, et al. Interlaboratory studies 62. Lazar C, Gatto L, Ferro M, et al. Accounting for the Multiple Natures of
and initiatives developing standards for proteomics. Proteomics. Missing Values in Label-Free Quantitative Proteomics Data Sets to
2013;13(6):904–909. Compare Imputation Strategies. J Proteome Res. 2016;15(4):1116–1125.
41. Percy AJ, Yang J, Chambers AG, et al. Protocol for Standardizing 63. Tyanova S, Temu T, Sinitcyn P, et al. The Perseus computational
High-to-Moderate Abundance Protein Biomarker Assessments platform for comprehensive analysis of (prote)omics data. Nature
Through an MRM-with-Standard-Peptides Quantitative Approach. methods. 2016;13(9):731–740.
Adv Exp Med Biol. 2016;919:515–530. 64. Carr SA, Abbatiello SE, Ackermann BL, et al. Targeted peptide
42. Percy AJ, Yang J, Hardie DB, et al. Precise quantitation of 136 measurements in biology and medicine: best practices for mass
urinary proteins by LC/MRM-MS using stable isotope labeled pep- spectrometry-based assay development using a fit-for-purpose
tides as internal standards for biomarker discovery and/or verifica- approach. Molecular & cellular proteomics: MCP. 2014;13(3):907–917.
tion studies. Methods. 2015;81:24–33. 65. Ebhardt HA, Root A, Sander C, et al. Applications of targeted
43. Percy AJ, Chambers AG, Smith DS, et al. Standardized protocols for proteomics in systems biology and translational medicine.
quality control of MRM-based plasma proteomic workflows. J Proteomics. 2015;15(18):3193–3208.
Proteome Res. 2013;12(1):222–233. 66. Anderson L, Hunter CL. Quantitative mass spectrometric multiple
44. Percy AJ, Chambers AG, Yang J, et al. Method and platform stan- reaction monitoring assays for major plasma proteins. Molecular &
dardization in MRM-based quantitative plasma proteomics. J cellular proteomics: MCP. 2006;5(4):573–588.
Proteomics. 2013;95:66–76. 67. Ta A, Se A, Schilling B, et al. Multi-site assessment of the precision
45. Malm J, Fehniger TE, Danmyr P, et al. Developments in biobanking and reproducibility of multiple reaction monitoring-based measure-
workflow standardization providing sample integrity and stability. J ments of proteins in plasma. Nat Biotechnol. 2009;27(7):633–641.
Proteomics. 2013;95:38–45. 68. Peterson AC, Russell JD, Bailey DJ, et al. Parallel reaction monitoring
46. Casadonte R, Caprioli RM. Proteomic analysis of formalin-fixed for high resolution and high mass accuracy quantitative, targeted
paraffin-embedded tissue by MALDI imaging mass spectrometry. proteomics. Molecular & cellular proteomics: MCP. 2012;11
Nat Protoc. 2011;6(11):1695–1709. (11):1475–1488.
47. Ostasiewicz P, Wisniewski JRA. Protocol for Large-Scale Proteomic 69. Cox J, Mann M. MaxQuant enables high peptide identification
Analysis of Microdissected Formalin Fixed and Paraffin Embedded rates, individualized p.p.b.-range mass accuracies and proteome-
Tissue. Methods Enzymol. 2017;585:159–176. wide protein quantification. Nat Biotechnol. 2008;26(12):1367–1372.
48. Kennedy JJ, Whiteaker JR, Schoenherr RM, et al. Optimized Protocol 70. Sturm M, Bertsch A, Gropl C, et al. OpenMS - an open-source
for Quantitative Multiple Reaction Monitoring-Based Proteomic software framework for mass spectrometry. BMC bioinformatics.
Analysis of Formalin-Fixed, Paraffin-Embedded Tissues. J Proteome 2008;9:163.
Res. 2016;15(8):2717–2728. 71. Rost HL, Sachsenberg T, Aiche S, et al. OpenMS: a flexible open-
49. Dickhut C, Radau S, Zahedi RP. Fast, efficient, and quality-controlled source software platform for mass spectrometry data analysis.
phosphopeptide enrichment from minute sample amounts using Nature methods. 2016;13(9):741–748.
titanium dioxide. Methods in molecular biology. 2014;1156:417–430. 72. Chalkley RJ, Clauser KR. Modification site localization scoring: stra-
50. Burkhart JM, Premsler T, Sickmann A. Quality control of nano-LC- tegies and performance. Molecular & cellular proteomics: MCP.
MS systems using stable isotope-coded peptides. Proteomics. 2012;11(5):3–14.
2011;11(6):1049–1057. 73. Beck F, Lewandrowski U, Wiltfang M, et al. The good, the bad, the
51. Muller T, Winter D. Systematic Evaluation of Protein Reduction and ugly: validating the mass spectrometric analysis of modified pep-
Alkylation Reveals Massive Unspecific Side Effects by Iodine-containing tides. Proteomics. 2011;11(6):1099–1109.
Reagents. Molecular & cellular proteomics: MCP. 2017;16(7):1173–1187. 74. Mikesh LM, Ueberheide B, Chi A, et al. The utility of ETD mass
52. Burkhart JM, Schumbrutzki C, Wortelkamp S, et al. Systematic and spectrometry in proteomic analysis. Biochim Biophys Acta.
quantitative comparison of digest efficiency and specificity reveals 2006;1764(12):1811–1822.
the impact of trypsin quality on MS-based proteomics. J Proteomics. 75. Gillet LC, Navarro P, Tate S, et al. Targeted data extraction of the
2012;75(4):1454–1462. MS/MS spectra generated by data-independent acquisition: a new
532 R. MNATSAKANYAN ET AL.

concept for consistent and accurate proteome analysis. Molecular & 96. Treindl F, Ruprecht B, Beiter Y, et al. A bead-based western for
cellular proteomics: MCP. 2012;11(6):O111 016717. high-throughput cellular signal transduction analyses. Nat
76. Simburger JM, Dettmer K, Oefner PJ, et al. Optimizing the SWATH-MS- Commun. 2016;7:12852.
workflow for label-free proteomics. J Proteomics. 2016;145:137–140. 97. Shi T, Song E, Nie S, et al. Advances in targeted proteomics and
77. Distler U, Kuharev J, Navarro P, et al. Label-free quantification in ion applications to biomedical research. Proteomics. 2016;16(15-
mobility-enhanced data-independent acquisition proteomics. Nat 16):2160–2182.
Protoc. 2016;11(4):795–812. 98. Bourmaud A, Gallien S, Domon B. Parallel reaction monitoring
78. Percy AJ, Chambers AG, Yang J, et al. Comparison of standard- and using quadrupole-Orbitrap mass spectrometer: principle and appli-
nano-flow liquid chromatography platforms for MRM-based quan- cations. Proteomics. 2016;16(15-16):2146–2159.
titation of putative plasma biomarker proteins. Anal Bioanal Chem. 99. Domon B, Gallien S. Recent advances in targeted proteomics for
2012;404(4):1089–1101. clinical applications. Proteomics Clin Appl. 2015;9(3–4):423–431.
79. Ross PL, Huang YN, Marchese JN, et al. Multiplexed protein quantitation 100. Mohammed Y, Domanski D, Jackson AM, et al. PeptidePicker: a scien-
in Saccharomyces cerevisiae using amine-reactive isobaric tagging tific workflow with web interface for selecting appropriate peptides
reagents. Molecular & cellular proteomics: MCP. 2004;3(12):1154–1169. for targeted proteomics experiments. J Proteomics. 2014;106:151–161.
80. Treitz C, Cassidy L, Hockendorf A, et al. Quantitative proteome 101. Se A, Dr M, Schilling B, et al. Design, implementation and multisite
analysis of Caenorhabditis elegans upon exposure to nematicidal evaluation of a system suitability protocol for the quantitative
Bacillus thuringiensis. J Proteomics. 2015;113:337–350. assessment of instrument performance in liquid chromatography-
81. Thompson A, Schafer J, Kuhn K, et al. Tandem mass tags: a novel multiple reaction monitoring-MS (LC-MRM-MS). Molecular & cellular
quantification strategy for comparative analysis of complex protein proteomics: MCP. 2013;12(9):2623–2639.
mixtures by MS/MS. Anal Chem. 2003;75(8):1895–1904. • A standardization approach for MRM measurements is demon-
82. Erickson BK, Jedrychowski MP, McAlister GC, et al. Evaluating multi- strated to yield precise inter- and intra-lab results at 11 locations.
plexed quantitative phosphopeptide analysis on a hybrid quadru- 102. Whiteaker JR, Halusa GN, Hoofnagle AN, et al. CPTAC Assay Portal: a
pole mass filter/linear ion trap/Orbitrap mass spectrometer. Anal repository of targeted proteomic assays. Nature methods. 2014;11
Chem. 2015;87(2):1241–1249. (7):703–704.
83. Venne AS, Solari, FA, Faden F, et al. An improved workflow for 103. Beck F, Geiger J, Gambaryan S, et al. Temporal quantitative phos-
quantitative N-terminal charge-based fractional diagonal chroma- phoproteomics of ADP stimulation reveals novel central nodes in
tography (ChaFRADIC) to study proteolytic events in Arabidopsis platelet activation and inhibition. Blood. 2017;129(2):e1–e12.
thaliana. Proteomics. 2015;15(14):2458–69. • Combination of TiO2 enrichment and PRM for targeted valida-
84. Hoffert JD, Pisitkun T, Saeed F, et al. Dynamics of the G protein- tion of phosphopeptide candidates obtained from discovery
coupled vasopressin V2 receptor signaling network revealed by phosphoproteomics. In total 23 phosphopeptides were ana-
quantitative phosphoproteomics. Molecular & cellular proteomics: lyzed in 7 different conditions using platelets obtained from
MCP. 2012;11(2):M111 014613. 5 healthy donors, which would correspond to a total of 800
85. Parker BL, Palmisano G, Edwards AV, et al. Quantitative N-linked Western blots.
glycoproteomics of myocardial ischemia and reperfusion injury 104. Beck F, Geiger J, Gambaryan S, et al. Time-resolved characterization
reveals early remodeling in the extracellular environment. of cAMP/PKA-dependent signaling reveals that platelet inhibition is
Molecular & cellular proteomics: MCP. 2011;10(8):M110 006833. a concerted process involving multiple signaling pathways. Blood.
86. Prudova A, Auf Dem Keller U, Butler GS, et al. Multiplex 2014;123(5):e1–e10.
N-terminome analysis of MMP-2 and MMP-9 substrate degradomes 105. Anderson NL, Anderson NG, Haines LR, et al. Mass spectrometric
by iTRAQ-TAILS quantitative proteomics. Molecular & cellular pro- quantitation of peptides and proteins using Stable Isotope
teomics: MCP. 2010;9(5):894–911. Standards and Capture by Anti-Peptide Antibodies (SISCAPA). J
87. Melo-Braga MN, Verano-Braga T, Leon IR, et al. Modulation of protein Proteome Res. 2004;3(2):235–244.
phosphorylation, N-glycosylation and Lys-acetylation in grape (Vitis 106. Razavi M, Frick LE, LaMarr WA, et al. High-throughput SISCAPA
vinifera) mesocarp and exocarp owing to Lobesia botrana infection. quantitation of peptides from human plasma digests by ultrafast,
Molecular & cellular proteomics: MCP. 2012;11(10):945–956. liquid chromatography-free mass spectrometry. J Proteome Res.
88. Huang H, Haar Petersen M, Ibanez-Vea M, et al. Simultaneous 2012;11(12):5642–5649.
Enrichment of Cysteine-containing Peptides and Phosphopeptides 107. Lin D, Alborn WE, Slebos RJ, et al. Comparison of protein immunopre-
Using a Cysteine-specific Phosphonate Adaptable Tag (CysPAT) in cipitation-multiple reaction monitoring with ELISA for assay of biomar-
Combination with titanium dioxide (TiO2) Chromatography. Mol ker candidates in plasma. J Proteome Res. 2013;12(12):5996–6003.
Cell Proteomics. 2016;15(10):3282–3296. 108. Popp R, Basik M, Spatz A, et al. How iMALDI can improve clinical
89. Erickson BK, Jedrychowski MP, McAlister GC, et al. Evaluating multi- diagnostics. Analyst. 2018;143(10):2197–2203.
plexed quantitative phosphopeptide analysis on a hybrid quadru- 109. Li H, Popp R, Frohlich B, et al. Peptide and Protein Quantification
pole mass filter/linear ion trap/orbitrap mass spectrometer. Anal Using Automated Immuno-MALDI (iMALDI). Journal of visualized
Chem. 2015;87(2):1241–1249. experiments: JoVE. 2017 126.
90. Vaudel M, Burkhart JM, Radau S, et al. Integral quantification accu- 110. Jiang J, Parker CE, Hoadley KA, et al. Development of an immuno
racy estimation for reporter ion-based quantitative proteomics tandem mass spectrometry (iMALDI) assay for EGFR diagnosis.
(iQuARI). J Proteome Res. 2012;11(10):5072–5080. Proteomics Clin Appl. 2007;1(12):1651–1659.
91. Karp NA, Huber W, Sadowski PG, et al. Addressing accuracy and 111. Popp R, Li H, LeBlanc A, et al. Immuno-Matrix-Assisted Laser
precision issues in iTRAQ quantitation. Molecular & cellular proteo- Desorption/Ionization Assays for Quantifying AKT1 and AKT2 in
mics: MCP. 2010;9(9):1885–1897. Breast and Colorectal Cancer Cell Lines and Tumors. Anal Chem.
92. Gillette MA, Carr SA. Quantitative analysis of peptides and proteins 2017;89(19):10592–10600.
in biomedicine by targeted mass spectrometry. Nature methods. 112. Hoeppe S, Schreiber TD, Planatscher H, et al. Targeting peptide
2013;10(1):28–34. termini, a novel immunoaffinity approach to reduce complexity in
93. Rose CM, Merrill AE, Bailey DJ, et al. Neutron encoded labeling for mass spectrometric protein identification. Molecular & cellular pro-
peptide identification. Anal Chem. 2013;85(10):5129–5137. teomics: MCP. 2011;10(2):M110 002857.
94. Potts GK, Voigt EA, Bailey DJ, et al. Neucode Labels for Multiplexed, 113. Marth JD, Grewal PK. Mammalian glycosylation in immunity. Nat
Absolute Protein Quantification. Anal Chem. 2016;88(6):3295–3303. Rev Immunol. 2008;8(11):874–887.
95. Boesch AW, Zhao Y, Landman AS, et al. A multiplexed assay to 114. Varinou L, Ramsauer K, Karaghiosoff M, et al. Phosphorylation of the
detect antimicrobial peptides in biological fluids and cell secre- Stat1 transactivation domain is required for full-fledged IFN-gamma-
tions. J Immunol Methods. 2013;397(1–2):71–76. dependent innate immunity. Immunity. 2003;19(6):793–802.
EXPERT REVIEW OF PROTEOMICS 533

115. Kaneko Y, Nimmerjahn F, Ravetch JV. Anti-inflammatory activity of 138. Lewandrowski U, Moebius J, Walter U, et al. Elucidation of
immunoglobulin G resulting from Fc sialylation. Science. 2006;313 N-glycosylation sites on human platelet proteins: a glycoproteomic
(5787):670–673. approach. Molecular & cellular proteomics: MCP. 2006;5(2):226–233.
116. Viatour P, Merville MP, Bours V, et al. Phosphorylation of NF-kappaB 139. Block TM, Comunale MA, Lowman M, et al. Use of targeted glyco-
and IkappaB proteins: implications in cancer and inflammation. proteomics to identify serum glycoproteins that correlate with liver
Trends Biochem Sci. 2005;30(1):43–52. cancer in woodchucks and humans. Proc Natl Acad Sci U S A.
117. Hakomori S. Tumor malignancy defined by aberrant glycosylation and 2005;102(3):779–784.
sphingo(glyco)lipid metabolism. Cancer Res. 1996;56(23):5309–5318. 140. Zhao J, Simeone DM, Heidt D, et al. Comparative serum glycopro-
118. Hwang RF, Yokoi K, Bucana CD, et al. Inhibition of platelet-derived teomics using lectin selected sialic acid glycoproteins with mass
growth factor receptor phosphorylation by STI571 (Gleevec) spectrometric analysis: application to pancreatic cancer serum. J
reduces growth and metastasis of human pancreatic carcinoma in Proteome Res. 2006;5(7):1792–1802.
an orthotopic nude mouse model. Clinical cancer research: an 141. Alley WR Jr., Vasseur JA, Goetz JA, et al. N-linked glycan structures
official journal of the American Association for Cancer Research. and their expressions change in the blood sera of ovarian cancer
2003;9(17):6534–6544. patients. J Proteome Res. 2012;11(4):2282–2300.
119. Tepper K, Biernat J, Kumar S, et al. Oligomer formation of tau 142. Sonneveld ME, de Haas M, Koeleman C, et al. Patients with IgG1-
protein hyperphosphorylated in cells. J Biol Chem. 2014;289 anti-red blood cell autoantibodies show aberrant Fc-glycosylation.
(49):34389–34407. Sci Rep. 2017;7(1):8187.
120. Grundke-Iqbal I, Iqbal K, Tung YC, et al. Abnormal phosphorylation 143. Jin C, Kenny DT, Skoog EC, et al. Structural diversity of human
of the microtubule-associated protein tau (tau) in Alzheimer cytos- gastric mucin glycans. Molecular & cellular proteomics: MCP.
keletal pathology. Proc Natl Acad Sci U S A. 1986;83(13):4913–4917. 2017;16(5):743–758.
121. Wang JZ, Grundke-Iqbal I, Iqbal K. Glycosylation of microtubule- 144. Hinneburg H, Korac P, Schirmeister F, et al. Unlocking Cancer
associated protein tau: an abnormal posttranslational modification Glycomes from Histopathological Formalin-fixed and Paraffin-
in Alzheimer’s disease. Nat Med. 1996;2(8):871–875. embedded (FFPE) Tissue Microdissections. Molecular & cellular pro-
122. Scheuner D, Mierde DV, Song B, et al. Control of mRNA translation teomics: MCP. 2017;16(4):524–536.
preserves endoplasmic reticulum function in beta cells and main- 145. Pagel O, Loroch S, Sickmann A, et al. Current strategies and find-
tains glucose homeostasis. Nat Med. 2005;11(7):757–764. ings in clinically relevant post-translational modification-specific
123. Ortsäter H, Grankvist N, Honkanen RE, et al. Protein phosphatases proteomics. Expert Rev Proteomics. 2015;12(3):235–253.
in pancreatic islets. J Endocrinol. 2014;221(3):R121–R144. 146. von Stechow L, Francavilla C, Olsen JV. Recent findings and tech-
124. Meyerovitch J, Backer JM, Kahn CR. Hepatic phosphotyrosine phos- nological advances in phosphoproteomics for cells and tissues.
phatase activity and its alterations in diabetic rats. J Clin Invest. Expert Rev Proteomics. 2015;12(5):469–487.
1989;84(3):976–983. 147. Engholm-Keller K, Larsen MR. Technologies and challenges in large-
125. Brownlee M. Advanced protein glycosylation in diabetes and aging. scale phosphoproteomics. Proteomics. 2013;13(6):910–931.
Annu Rev Med. 1995;46:223–234. 148. Riley NM, Coon JJ. Phosphoproteomics in the Age of Rapid and
126. Blume-Jensen P, Hunter T. Oncogenic kinase signalling. Nature. Deep Proteome Profiling. Anal Chem. 2016;88(1):74–94.
2001;411(6835):355–365. 149. Engholm-Keller K, Birck P, Storling J, et al. TiSH–a robust and
127. Zanivan S, Meves A, Behrendt K, et al. In vivo SILAC-based proteo- sensitive global phosphoproteomics strategy employing a combi-
mics reveals phosphoproteome changes during mouse skin carci- nation of TiO2, SIMAC, and HILIC. J Proteomics. 2012;75(18):5749–
nogenesis. Cell Rep. 2013;3(2):552–566. 5761.
128. Huelsemann MF, Patz M, Beckmann L, et al. Hypoxia-induced p38 150. Geiger T, Cox J, Ostasiewicz P, et al. Super-SILAC mix for quantitative
MAPK activation reduces Mcl-1 expression and facilitates sensitivity proteomics of human tumor tissue. Nature methods. 2010;7(5):383–385.
towards BH3 mimetics in chronic lymphocytic leukemia. Leukemia. 151. Schweppe DK, Rigas JR, Gerber SA. Quantitative phosphoproteomic
2015;29(4):981–984. profiling of human non-small cell lung cancer tumors. J Proteomics.
129. Hakomori S. Glycosylation defining cancer malignancy: new wine in 2013;91:286–296.
an old bottle. Proc Natl Acad Sci U S A. 2002;99(16):10231–10233. 152. Dazert E, Colombi M, Boldanova T, et al. Quantitative proteomics and
130. Decker RS, Decker ML, Kulikovskaya I, et al. Myosin-binding protein phosphoproteomics on serial tumor biopsies from a sorafenib-treated
C phosphorylation, myofibril structure, and contractile function HCC patient. Proc Natl Acad Sci U S A. 2016;113(5):1381–1386.
during low-flow ischemia. Circulation. 2005;111(7):906–912. 153. Kennedy JJ, Yan P, Zhao L, et al. Immobilized Metal Affinity
131. El-Armouche A, Pohlmann L, Schlossarek S, et al. Decreased phos- Chromatography Coupled to Multiple Reaction Monitoring
phorylation levels of cardiac myosin-binding protein-C in human and Enables Reproducible Quantification of Phospho-signaling.
experimental heart failure. J Mol Cell Cardiol. 2007;43(2):223–229. Molecular & cellular proteomics: MCP. 2016;15(2):726–739.
132. Schoutsen B, Blom JJ, Verdouw PD, et al. Calcium transport and 154. de Graaf EL, Kaplon J, Mohammed S, et al. Signal Transduction
phospholamban in sarcoplasmic reticulum of ischemic myocar- Reaction Monitoring Deciphers Site-Specific PI3K-mTOR/MAPK
dium. J Mol Cell Cardiol. 1989;21(7):719–727. Pathway Dynamics in Oncogene-Induced Senescence. J Proteome
133. Nc T, Ab H, Jr G, et al. Quantitative analysis of cell signaling and Res. 2015;14(7):2906–2914.
drug action via mass spectrometry-based systems level phospho- 155. Loroch S, Zahedi RP, Sickmann A. Highly sensitive phosphoproteo-
proteomics. Proteomics. 2009;9(6):1469–1487. mics by tailoring solid-phase extraction to electrostatic repulsion-
134. Kumar A, Baycin-Hizal D, Shiloach J, et al. Coupling enrichment and hydrophilic interaction chromatography. Anal Chem. 2015;87
proteomics methods for understanding and treating disease. (3):1596–1604.
Proteomics Clin Appl. 2015;9(1–2):33–47. 156. Alpert AJ. Electrostatic repulsion hydrophilic interaction chromato-
135. Liu T, Qian WJ, Gritsenko MA, et al. Human plasma N-glycoproteome graphy for isocratic separation of charged solutes and selective
analysis by immunoaffinity subtraction, hydrazide chemistry, and isolation of phosphopeptides. Anal Chem. 2008;80(1):62–76.
mass spectrometry. J Proteome Res. 2005;4(6):2070–2080. 157. Di Conza G, Trusso Cafarello S, Loroch S, et al. The mTOR and PP2A
136. Jung K, Cho W, Regnier FE. Glycoproteomics of plasma based on Pathways Regulate PHD2 Phosphorylation to Fine-Tune HIF1alpha
narrow selectivity lectin affinity chromatography. J Proteome Res. Levels and Colorectal Cancer Cell Survival under Hypoxia. Cell Rep.
2009;8(2):643–650. 2017;18(7):1699–1712.
137. Wang L, Li F, Sun W, et al. Concanavalin A-captured glycoproteins •• Absolute quantitation of phosphorylation levels in colorectal
in healthy human urine. Molecular & cellular proteomics: MCP. cancer biopsies using a tailored ERLIC-PRM approach and
2006;5(3):560–562. requiring only 32 µg of total tissue protein.
534 R. MNATSAKANYAN ET AL.

158. Chien KY, Liu HC, Goshe MB. Development and application of a 177. Wildes D, Wells JA. Sampling the N-terminal proteome of human
phosphoproteomic method using electrostatic repulsion-hydrophi- blood. Proc Natl Acad Sci U S A. 2010;107(10):4561–4566.
lic interaction chromatography (ERLIC), IMAC, and LC-MS/MS ana- 178. Wiita AP, Hsu GW, Lu CM, et al. Circulating proteolytic signatures of
lysis to study Marek’s Disease Virus infection. J Proteome Res. chemotherapy-induced cell death in humans discovered by
2011;10(9):4041–4053. N-terminal labeling. Proc Natl Acad Sci U S A. 2014;111(21):7594–7599..
159. Hao P, Ren Y, Dutta B, et al. Comparative evaluation of electrostatic • N-terminomics study identifying potential proteolytic markers
repulsion-hydrophilic interaction chromatography (ERLIC) and of chemotherapy-induced cell death, which were also vali-
high-pH reversed phase (Hp-RP) chromatography in profiling of dated by MRM.
rat kidney proteome. J Proteomics. 2013;82:254–262. 179. Solari FA, Mattheij NJ, Burkhart JM, et al. Combined Quantification
160. Chen X, Wu D, Zhao Y, et al. Increasing phosphoproteome cover- of the Global Proteome, Phosphoproteome, and Proteolytic
age and identification of phosphorylation motifs through combina- Cleavage to Characterize Altered Platelet Functions in the Human
tion of different HPLC fractionation methods. J Chromatogr B Analyt Scott Syndrome. Molecular & cellular proteomics: MCP. 2016;15
Technol Biomed Life Sci. 2011;879(1):25–34. (10):3154–3169.
161. Mertins P, Yang F, Liu T, et al. Ischemia in tumors induces early and 180. Shema G, Nguyen MTN, Solari FA, et al. Simple, scalable, and
sustained phosphorylation changes in stress kinase pathways but ultrasensitive tip-based identification of protease substrates. Mol
does not affect global protein levels. Molecular & cellular proteo- Cell Proteomics. 2018;17(4):826–834.
mics: MCP. 2014;13(7):1690–1704. 181. Chung HS, Wang SB, Venkatraman V, et al. Cysteine oxidative
162. Gajadhar AS, Johnson H, Slebos RJ, et al. Phosphotyrosine signaling posttranslational modifications: emerging regulation in the cardio-
analysis in human tumors is confounded by systemic ischemia- vascular system. Circ Res. 2013;112(2):382–392.
driven artifacts and intra-specimen heterogeneity. Cancer Res. 182. Kim HJ, Ha S, Lee HY, et al. ROSics: chemistry and proteomics of
2015;75(7):1495–1503. cysteine modifications in redox biology. Mass Spectrom Rev.
163. Petrera A, Lai ZW, Schilling O. Carboxyterminal Protein Processing 2015;34(2):184–208.
in Health and Disease: key Actors and Emerging Technologies. J 183. Gould N, Doulias PT, Tenopoulou M, et al. Regulation of protein
Proteome Res. 2014;13(11):4497–4504. function and signaling by reversible cysteine S-nitrosylation. J Biol
164. Mossmann D, Vögtle FN, Taskin Asli A, et al. Amyloid-β Peptide Chem. 2013;288(37):26473–26479.
Induces Mitochondrial Dysfunction by Inhibition of Preprotein 184. Waszczak C, Akter S, Jacques S, et al. Oxidative post-translational
Maturation. Cell Metab. 2014;20(4):662–669. modifications of cysteine residues in plant signal transduction. J
165. McCarty SM, Percival SL. Proteases and Delayed Wound Healing. Exp Bot. 2015;66(10):2923–2934.
Advances in wound care. 2013;2(8):438–447. 185. Nietzel T, Mostertz J, Hochgrafe F, et al. Redox regulation of mito-
166. Csiszar A, Kutay B, Wirth S, et al. Interleukin-like epithelial-to- chondrial proteins and proteomes by cysteine thiol switches.
mesenchymal transition inducer activity is controlled by proteolytic Mitochondrion. 2016;33:72–83.
processing and plasminogen-urokinase plasminogen activator 186. Sun J, Protein ME. S-nitrosylation and cardioprotection. Circ Res.
receptor system-regulated secretion during breast cancer progres- 2010;106(2):285–296.
sion. Breast cancer research: BCR. 2014;16(5):433. 187. Watanabe Y, Cohen RA, Matsui R. Redox Regulation of Ischemic
167. Gevaert K, Goethals M, Martens L, et al. Exploring proteomes and Angiogenesis- Another Aspect of Reactive Oxygen Species. Circ J.
analyzing protein processing by mass spectrometric identification 2016;80(6):1278–1284.
of sorted N-terminal peptides. Nat Biotechnol. 2003;21(5):566–569. 188. Wang SB, Foster DB, Rucker J, et al. Redox regulation of mitochon-
168. Kleifeld O, Doucet A, Auf Dem Keller U, et al. Isotopic labeling of drial ATP synthase: implications for cardiac resynchronization ther-
terminal amines in complex samples identifies protein N-termini apy. Circ Res. 2011;109(7):750–757.
and protease cleavage products. Nat Biotech. 2010;28(3):281–288. 189. Yuan K, Liu Y, Chen HN, et al. Thiol-based redox proteomics in
169. Venne AS, Vogtle FN, Meisinger C, et al. Novel highly sensitive, cancer research. Proteomics. 2015;15(2–3):287–299.
specific, and straightforward strategy for comprehensive N-terminal 190. Murphy E, Kohr M, Menazza S, et al. Signaling by S-nitrosylation in
proteomics reveals unknown substrates of the mitochondrial pepti- the heart. J Mol Cell Cardiol. 2014;73:18–25.
dase Icp55. J Proteome Res. 2013;12(9):3823–3830. 191. Butterfield DA, Dalle-Donne I. Redox proteomics: from protein
170. Mahrus S, Trinidad JC, Barkan DT, et al. Global sequencing of modifications to cellular dysfunction and disease. Mass Spectrom
proteolytic cleavage sites in apoptosis by specific labeling of pro- Rev. 2014;33(1):1–6.
tein N termini. Cell. 2008;134(5):866–876. 192. Raju K, Doulias PT, Evans P, et al. Regulation of brain glutamate
171. Dix MM, Simon GM, Cravatt BF. Global identification of caspase metabolism by nitric oxide and S-nitrosylation. Sci Signal. 2015;8
substrates using PROTOMAP (protein topography and migration (384):ra68.
analysis platform). Methods in molecular biology. 2014;1133:61–70. 193. Duan J, Gaffrey MJ, Qian WJ. Quantitative proteomic characteriza-
172. Segura V, Garin-Muga A, Guruceaga E, et al. Progress and pitfalls in tion of redox-dependent post-translational modifications on pro-
finding the ‘missing proteins’ from the human proteome map. tein cysteines. Mol Biosyst. 2017;13(5):816–829.
Expert Rev Proteomics. 2017;14(1):9–14. 194. McDonagh B. Detection of ROS Induced Proteomic Signatures by
173. Baker MS, Ahn SB, Mohamedali A, et al. Accelerating the search for Mass Spectrometry. Front Physiol. 2017;8:470.
the missing proteins in the human proteome. Nat Commun. 195. Nagumo K, Tanaka M, Chuang VT, et al. Cys34-cysteinylated human
2017;8:14271. serum albumin is a sensitive plasma marker in oxidative stress-
174. Lange PF, Huesgen PF, Nguyen K, et al. Annotating N Termini for related chronic diseases. PLoS One. 2014;9(1):e85216.
the Human Proteome Project: N Termini and Nα-Acetylation Status 196. Delobel J, Prudent M, Crettaz D, et al. Cysteine redox proteomics of
Differentiate Stable Cleaved Protein Species from Degradation the hemoglobin-depleted cytosolic fraction of stored red blood
Remnants in the Human Erythrocyte Proteome. J Proteome Res. cells. Proteomics Clin Appl. 2016;10(8):883–893.
2014;13(4):2028–2044. 197. Sonego G, Abonnenc M, Tissot JD, et al. Redox Proteomics and
175. Eckhard U, Marino G, Abbey SR, et al. The Human Dental Pulp Platelet Activation: Understanding the Redox Proteome to Improve
Proteome and N-Terminome: Levering the Unexplored Potential Platelet Quality for Transfusion. Int J Mol Sci. 2017;18:2.
of Semitryptic Peptides Enriched by TAILS to Identify Missing 198. Wojdyla K, Williamson J, Roepstorff P, et al. SNO/SOH TMT strategy
Proteins in the Human Proteome Project in Underexplored for combinatorial analysis of reversible cysteine oxidations. J
Tissues. J Proteome Res. 2015;14(9):3568–3582. Proteomics. 2015;113:415–434.
176. Klein T, Fung S-Y, Renner F, et al. The paracaspase MALT1 cleaves 199. Guo J, Gaffrey MJ, Su D, et al. Resin-assisted enrichment of thiols as
HOIL1 reducing linear ubiquitination by LUBAC to dampen lym- a general strategy for proteomic profiling of cysteine-based rever-
phocyte NF-κB signalling. 2015;6:8777. sible modifications. Nat Protoc. 2014;9(1):64–75.
EXPERT REVIEW OF PROTEOMICS 535

200. García-Santamarina S, Boronat S, Domènech A, et al. Monitoring in 220. Christin C, Hoefsloot HC, Smilde AK, et al. A critical assessment of
vivo reversible cysteine oxidation in proteins using ICAT and mass feature selection methods for biomarker discovery in clinical pro-
spectrometry. Nat Protoc. 2014;9(5):1131–1145. teomics. Molecular & cellular proteomics: MCP. 2013;12(1):263–276.
201. Pan KT, Chen YY, Pu TH, et al. Mass spectrometry-based quantita- 221. Skates SJ, Gillette MA, LaBaer J, et al. Statistical design for biospeci-
tive proteomics for dissecting multiplexed redox cysteine modifica- men cohort size in proteomics-based biomarker discovery and
tions in nitric oxide-protected cardiomyocyte under hypoxia. verification studies. J Proteome Res. 2013;12(12):5383–5394.
Antioxid Redox Signal. 2014;20(9):1365–1381. 222. Diz AP, Carvajal-Rodriguez A, Skibinski DO. Multiple hypothesis
202. Forrester MT, Thompson JW, Foster MW, et al. Proteomic analysis of testing in proteomics: a strategy for experimental work. Molecular
S-nitrosylation and denitrosylation by resin-assisted capture. Nat & cellular proteomics: MCP. 2011;10(3):M110 004374.
Biotechnol. 2009;27(6):557–559. 223. Loroch S, Trabold K, Gambaryan S, et al. Alterations of the platelet
203. Wolhuter K, Whitwell HJ, Switzer CH, et al. Evidence against Stable proteome in type I Glanzmann thrombasthenia caused by different
Protein S-Nitrosylation as a Widespread Mechanism of Post-transla- homozygous delG frameshift mutations in ITGA2B. Thromb
tional Regulation. Mol Cell. 2018;69(3):438–450 e435. Haemost. 2017;117(3):556–569.
204. Chouchani ET, James AM, Methner C, et al. Identification and 224. Kammers K, Cole RN, Tiengwe C, et al. Detecting Significant
quantification of protein S-nitrosation by nitrite in the mouse Changes in Protein Abundance. EuPA open proteomics.
heart during ischemia. J Biol Chem. 2017;292(35):14486–14495. 2015;7:11–19.
205. Ibanez-Vea M, Huang H, Martinez De Morentin X, et al. Characterization 225. Datta S, Satten GA, Benos DJ, et al. An empirical bayes adjustment
of Macrophage Endogenous S-Nitrosoproteome Using a Cysteine- to increase the sensitivity of detecting differentially expressed
Specific Phosphonate Adaptable Tag in Combination with TiO2 genes in microarray experiments. Bioinformatics (Oxford, England).
Chromatography. J Proteome Res. 2018;17(3):1172–1182. 2004;20(2):235–242.
206. Yang J, Gupta V, Tallman KA, et al. Global, in situ, site-specific 226. Wettenhall JM, Smyth GK. limmaGUI: a graphical user interface for
analysis of protein S-sulfenylation. Nat Protoc. 2015;10(7):1022–1037. linear modeling of microarray data. Bioinformatics (Oxford,
207. Poole LB, Karplus PA, Claiborne A. Protein sulfenic acids in redox England). 2004;20(18):3705–3706.
signaling. Annu Rev Pharmacol Toxicol. 2004;44:325–347. 227. Ting L, Cowley MJ, Hoon SL, et al. Normalization and statistical
208. Yang J, Gupta V, Carroll KS, et al. Site-specific mapping and quan- analysis of quantitative proteomics data generated by meta-
tification of protein S-sulphenylation in cells. Nat Commun. bolic labeling. Molecular & cellular proteomics: MCP. 2009;8
2014;5:4776. (10):2227–2242.
209. Li R, Klockenbusch C, Lin L, et al. Quantitative Protein Sulfenic Acid 228. Ritchie ME, Phipson B, Wu D, et al. limma powers differential
Analysis Identifies Platelet Releasate-Induced Activation of Integrin expression analyses for RNA-sequencing and microarray studies.
beta2 on Monocytes via NADPH Oxidase. J Proteome Res. 2016;15 Nucleic Acids Res. 2015;43(7):e47.
(12):4221–4233. 229. Boja ES, Rodriguez H. Proteogenomic convergence for under-
210. Bechtel TJ, Weerapana E. From structure to redox: the diverse func- standing cancer pathways and networks. Clin Proteomics.
tional roles of disulfides and implications in disease. Proteomics. 2014;11(1):22.
2017;17:6. 230. Alfaro JA, Sinha A, Kislinger T, et al. Onco-proteogenomics: cancer
211. Kang DH, Lee DJ, Lee KW, et al. Peroxiredoxin II is an essential proteomics joins forces with genomics. Nature methods. 2014;11
antioxidant enzyme that prevents the oxidative inactivation of (11):1107–1113.
VEGF receptor-2 in vascular endothelial cells. Mol Cell. 2011;44 231. Schwamborn K, Caprioli RM. Molecular imaging by mass spectro-
(4):545–558. metry–looking beyond classical histology. Nat Rev Cancer. 2010;10
212. Lu S, Fan SB, Yang B, et al. Mapping native disulfide bonds at a (9):639–646.
proteome scale. Nat Methods. 2015;12(4):329–331. 232. Domanski D, Murphy LC, Borchers CH. Assay development for the
213. Murdoch CE, Shuler M, Haeussler DJ, et al. Glutaredoxin-1 up- determination of phosphorylation stoichiometry using multiple
regulation induces soluble vascular endothelial growth factor reaction monitoring methods with and without phosphatase treat-
receptor 1, attenuating post-ischemia limb revascularization. J Biol ment: application to breast cancer signaling pathways. Anal Chem.
Chem. 2014;289(12):8633–8644. 2010;82(13):5610–5620.
214. Duan J, Kodali VK, Gaffrey MJ, et al. Quantitative Profiling of Protein 233. The M, Tasnim A, Kall L. How to talk about protein-level false
S-Glutathionylation Reveals Redox-Dependent Regulation of discovery rates in shotgun proteomics. Proteomics. 2016;16
Macrophage Function during Nanoparticle-Induced Oxidative (18):2461–2469.
Stress. ACS Nano. 2016;10(1):524–538. 234. Zahedi RP. Joining forces: studying multiple post-translational
215. McGarry DJ, Chen W, Chakravarty P, et al. Proteome-wide identifi- modifications to understand dynamic disease mechanisms. Expert
cation and quantification of S-glutathionylation targets in mouse Rev Proteomics. 2016;13(12):1055–1057.
liver. Biochem J. 2015;469(1):25–32. 235. Diehl HC, Beine B, Elm J, et al. The challenge of on-tissue diges-
216. Chen R, Mias GI, Li-Pook-Than J, et al. Personal omics profiling tion for MALDI MSI- a comparison of different protocols to
reveals dynamic molecular and medical phenotypes. Cell. improve imaging experiments. Anal Bioanal Chem. 2015;407
2012;148(6):1293–1307. (8):2223–2243.
217. Parker CE, Borchers CH. Mass spectrometry based biomarker dis- 236. Kopczynski D, Coman C, Zahedi RP, et al. Multi-OMICS: a critical
covery, verification, and validation–quality assurance and control of technical perspective on integrative lipidomics approaches.
protein biomarker assays. Mol Oncol. 2014;8(4):840–858.. Biochim Biophys Acta. 2017;1862(8):808–811.
• Review article about biomarker discovery with MS. 237. Conrads TP, Petricoin EF 3rd. The Obama Administration’s Cancer
218. Geyer PE, Holdt LM, Teupser D, et al. Revisiting biomarker discovery Moonshot: a Call for Proteomics. Clinical cancer research: an official
by plasma proteomics. Mol Syst Biol. 2017;13(9):942. journal of the American Association for Cancer Research. 2016;22
219. Hernandez B, Parnell A, Pennington SR. Why have so few proteomic (18):4556–4558.
biomarkers “survived” validation? (Sample size and independent vali- 238. Aelion CM, Airhihenbuwa CO, Alemagno S, et al. The US Cancer
dation considerations). Proteomics. 2014;14(13-14):1587–1592. Moonshot initiative. Lancet. Oncol. 2016;17(5):e178–180.

You might also like