Está en la página 1de 10

Special issue paper

Published online in Wiley Online Library

(wileyonlinelibrary.com) DOI 10.1002/bmc.3157

Recent advances in proteomics: towards the


human proteome
Kui Wanga,b, Canhua Huanga and Edouard Niceb*
ABSTRACT: After the successful completion of the Human Genome project in 2003, the next major challenge was to
understand when and where the encoded proteins were expressed, and to generate a map of the complex, interconnected
pathways, networks and molecular systems (the human proteome) that, taken together, control the workings of all cells,
tissues, organs and organisms. Proteomics will be fundamental for such studies. This review summarizes the key discoveries
that laid down the foundations for proteomics as we now know it, and describes key recent technological advances that will
undoubtedly contribute to achieving the initial goal of the Human Proteome Organization of identifying and characterizing
at least one protein product and representative post-translational modifications, single amino acid polymorphisms and splice
variant isoforms from the 20,300 human protein-coding genes within the next 10 years. Successful unraveling of the human
proteome will undoubtedly improve our understanding of human biology at the cellular level and lay the foundations for
improved diagnostic, prognostic, therapeutic and preventive medical outcomes as we enter the era of personalized medicine.
Copyright © 2014 John Wiley & Sons, Ltd.

Keywords: Proteomics; human proteome; HUPO; protein MS; automated sample preparation; validated antibodies

Introduction et al., 1989; Hu et al., 2005). Such mass spectrometers provide


excellent sensitivity and mass resolving power (Hardman and
The foundations for proteomics as we now know it were laid Makarov, 2003). The use of peptide sequence tags (Mann and
down more than 30 years ago with the development by Wilm, 1994), whereby MS/MS data is used for matching peptides
Macfarlane and Torgerson (1976) of Cf252 plasma-desorption to their cognate protein, facilitated identification.
mass spectrometry (PDMS) with time of flight analysis (later Coupled with these advances in mass spectrometry,
commercialized as the Bio-Ion in 1982), which was used to developments in the separation sciences were also required for
successfully detect insulin (Hakansson et al., 1982), followed pre-purification and enrichment of low abundance proteins in
closely by the development of fast atom bombardment mass complex biological mixtures. However, while N-terminal sequence
spectroscopy (FABMS) techniques for protein and peptide analysis using Edman Chemistry (Edman, 1950), which had been
analysis (Barber et al., 1981; Dell and Morris, 1982). However, it the method used for protein characterization prior to the develop-
was perceived that both these technologies had limitations in ment of mass spectrometry, required purification to homogeneity,
both the mass range (<25 kDa) and sensitivity (picomole) that for MS only partial purification is required.
would ultimately be required for comprehensive analysis of Foremost among the protein separation techniques used for
proteins and peptides in biological samples (Roepstorff, 1996), proteomic analysis has been two-dimensional polyacrylamide
and, in the case of the Bio-Ion, the lifetime of the Cf252 source
(half-life 2.64 years).
The next major breakthroughs came in 1988 when two
techniques, which remain the cornerstone of many proteomic
* Correspondence to: Edouard Nice; Department of Biochemistry and Molecular
studies to this day, were published: ESI and MALDI. John Fenn from Biology, Monash University, Clayton, Victoria, 3800, Australia. Email: ed.
Yale University described a new ionization technique, termed nice@monash.edu
electrospray ionization (ESI), for protein analysis (Meng et al.,
a
1988), and Michael Karas and Franz Hillenkamp, from the Univer- The State Key Laboratory of Biotherapy, West China Hospital, Sichuan University,
Chengdu, 610041, People’s Republic of China
sity of Munster in Germany, presented a new ionization technique,
matrix-assisted laser desorption/ionization (MALDI; with which b
Department of Biochemistry and Molecular Biology, Monash University,
they analyzed lysozyme (Mr 14,306), β-lactoglobulin (Mr 18,277), Clayton, Victoria, 3800, Australia
trypsin (Mr 23,463) and bovine serum albumen (Mr ~67,000; Fig. 1).
Abbreviations used: 2D-PAGE, two-dimensional polyacrylamide gel
John Fenn was to win the Nobel Prize in Chemistry with Koichi
electrophoresis; ABP, albumin-binding protein; ESI, electrospray ionization;
Tanaka in 2002 for their work on soft desorption ionization for FABMS, fast atom bombardment mass spectroscopy; FASP, filtered aided
mass spectrometric analyses of biological macromolecules. sample preparation; FFPE, formalin-fixed, paraffin-embedded; HUPO,
More recent advances in mass spectrometry include the Human Proteome Organization; IMS, imaging mass spectrometry; MALDI,
development of the Orbitrap and the use of ESI. Invented by matrix-assisted laser desorption/ionization; Mr, molecular weight; MRM,
multiple reaction monitoring; MWCO, molecular weight cut-off; OxICAT, ox-
Alexander Marakov in 1999 (Makarov 2000), the Orbitrap uses idative isotope-coded affinity tags; PDMS, plasma-desorption mass spec-
the principle of radially trapping ions about a central spindle trometry; prEST, protein epitope signature tag; PTM, post-translational
electrode and it can be equipped with an ESI source (Fenn
848

modification; QQQ, triple quadrupole; SRM, selected reaction monitoring.

Biomed. Chromatogr. 2014; 28: 848–857 Copyright © 2014 John Wiley & Sons, Ltd.
Recent advances in proteomics

a multidimensional mode, giving high purification factors, for


effective sample workup prior to proteomics analysis (Nice
et al., 2007). In addition, depletion strategies (e.g. Agilent MARS;
Agilent Technologies, Inc., Santa Clara, CA, USA) and Sigma
ProteoPrep® 20 (Sigma-Aldrich Co., St Louis, MO, USA) were
developed to remove the high abundance ‘housekeeping pro-
teins’, particularly in serum, which tended to mask low-level
components during MS analysis, although it was quickly realized
that some lower abundance proteins could ‘piggy back’ on these
high abundance proteins during depletion and therefore be lost.
In addition, the peptide chromatography needed to be opti-
mized for MS compatibility, and capillary columns (<1 mm i.d.)
were developed for sample delivery to the mass spectrometer
at low flow rate (Tong et al., 1997). Currently, to achieve the
separation efficiency required for the separation of complex
peptide mixtures, high efficiency nano HPLC columns
Figure 1. The first MALDI spectrum of bovine serum albumin with smaller i.d. (10–150 μm) operated at very low flow rate
(reproduced by permission of Karas and Hillenkamp, 1988). (10–1000 nL/min) are being widely used (Karlsson and Novotny,
1988). Recently, Mechtler’s group developed a novel nano LC
method in which ultralong gradients (Acclaim PepMap C18; 2 cm ×
gel electrophoresis (2D-PAGE) and microchromatography. 100 μm × 5 μm, 100 Å; Dionex, Sunnyvale, CA, USA) were used to
Patrick O’Farrell developed high-resolution methods for 2D-gel analyze the whole-cell protein extracts. In this study, 2761 proteins
electrophoresis in the early 1970s (O’Farrell, 1975; Fig. 2) which from Hela cell lysate were identified (Kocher et al., 2012) in a single
are still widely used, although he apparently never originally run. Likewise, Mann’s group (Thakur et al., 2011) has shown
envisaged the impact that this technology would have on how the use of long chromatographic run times (gradient times
understanding modern systems biology (O’Farrell 2008). Today, up to 6 h) can result in deep and highly sensitive coverage of the
over 10,000 spots can be separated in a single run using large yeast proteome without prior fractionation.
format gels (Klose and Kobalz, 1995). Clearly, advances in proteomics have, as summarized above, like
In the chromatography arena, early work on the development advances in genomics, been largely driven by technological
of micropreparative HPLC systems was initially driven largely by innovation. Progress in the proteomics field is exemplified by the
the need to purify low microgram quantities of low abundance exponential increase in the number of proteomics publications
proteins such as growth factors, their receptors and other (Fig. 3A) and the number of human protein sequences deposited
signaling molecules from complex biological matrices in high in the databases (Fig. 3B, 3C). Until fairly recently many people
yield for N-terminal sequence analysis. It was quickly realized thought that proteomics had underperformed for the large
that the use of ‘conventional’ 4.6 mm i.d. columns gave vanish- amount of capital invested, particularly in the fields of biomarker
ingly low protein recovery levels owing largely to on-column discovery and validation and clinical proteomics (Poste, 2011;
losses by nonspecific absorption, and that column dimensions Service, 2008; Zolg, 2006). However, the tide now appears to be
had to be carefully tuned to the sample load applied. By contrast turning (Nice, 2013), and technology once again is leading the way.
the use of short (3–10 cm) columns of reduced i.d. (1–2.1 mm)
gave excellent recovery at high concentration of microgram
quantities of protein (Nice, 1990) when operated at the The role of the Human Proteome
equivalent linear flow velocity. These columns could be used in
Organization
It is now realized (Hood et al., 2012; Poste, 2012) that big-science
projects like the Human Proteome Project require careful coordi-
nation to bring together the requisite global multidisciplinary
teams and infrastructure (e.g. protein chemists, peptide
chemists, organic chemists, molecular biologists, physicists,
clinicians, bioinformaticians, statisticians, computer scientists,
engineers, regulatory experts, instrument companies, diagnostic
companies and well curated biobanks) required for success. The
Human Proteome Organization (HUPO) was founded in 2001
with the goals of promoting the field of proteomics and its
application through international cooperation and research
collaborations and, importantly, by fostering the development
of new technologies and techniques and the necessary reagents
(Omenn, 2013).
HUPO has identified three working pillars – mass spec-
trometry, antibodies and an integrated knowledge base
Figure 2. An early (27 May 1974) 2D-PAGE separation of Escherichia coli (bioinformatics) – and has structured discovery around
14
proteins labeled with C-amino-acids (reproduced with permission of chromosome-centric (C-HPP) and disease and biology-based
O’Farrell, 2008). approaches (B/DHPP; Fig. 4; Legrain et al., 2011). The primary
849

Biomed. Chromatogr. 2014; 28: 848–857 Copyright © 2014 John Wiley & Sons, Ltd. wileyonlinelibrary.com/journal/bmc
K. Wang et al.

Figure 3. Proteomics publications and human protein sequences deposited in the databases. The number of proteomics publications per annum. The
data was generated by performing a Pubmed search with the term ‘proteome’ or ‘proteomics’ in the title or abstract (A). The number of human protein
sequences that has been deposited to Swiss-Prot (B) or TrEMBL (C). The primary data was downloaded from http://www.uniprot.org/

sample (typically using trypsin) are introduced by HPLC into


the first quadrupole of the QQQ instrument where proteotypic
precursor ions are preselected and, following collision-induced
dissociation of the precursor ions in the second quadrupole,
selected product ions are monitored in the third sector. MRM
has the advantage over antibody-based techniques that it is
generic and thus obviates the need to generate expensive
reagents for each individual assay. The complexity of the resulting
MRM spectra is significantly reduced (Fig. 5), facilitating the detec-
tion and measurement of low abundance proteins. If scheduled
MRM is used, in which selected ions are only monitored during
the time window in which they elute from the HPLC (Lange
et al., 2008), multiplex analysis of hundreds of targets is possible.
Selectivity is afforded from the mass of the specific proteotypic
Figure 4. The Human Proteome Organization (HUPO) operating structure peptide chosen, the corresponding product ions and the charac-
(reproduced by permission of Legrain et al., 2011). Note the three pillars:
teristic HPLC elution time. The sensitivity is now approaching that
mass spectrometry (MS), antibodies (Abs) and knowledge base (KB).
of ELISA assays, particularly if specific enrichment techniques are
used (see later).
Quantitative MRM methods have been developed for analysis
goal of HUPO is to identify and characterize at least one protein of samples from a number of biological matrices, including
product for each of the 20,300 human protein-coding genes blood (Anderson and Hunter, 2006; Domanski et al., 2012;
including examples of post-translational modifications (PTMs), Stahl-Zeng et al., 2007), urine (Chen et al., 2012) and feces (Ang
single amino acid polymorphisms and splice variant isoforms. At and Nice, 2010; Ang et al., 2011). In the case of the fecal samples
present about 5000 (25%) of the predicted human protein-coding it was possible to analyze carcinoembryonic antigen, a low level
genes lack any experimental evidence at the protein level (Gaudet component, in trifluoroacetic acid fecal extracts (ng/mg stool),
et al., 2013). Providing confirmatory evidence of these putative without prior sample fractionation (Ang et al., 2011).
proteins is an initial HUPO priority. Some of the key recent technol-
ogy advances that will assist in the unraveling of the human
proteome are summarized below, with examples from the litera- SWATH
ture and our own laboratories.
Another important technological advance that will facilitate
consistent and accurate proteome analysis is SWATH (Sequential
Windowed data independent Acquisition of the Total High-reso-
Multiple reaction monitoring lution mass spectra; Gillet et al., 2012). In SWATH mode, instead
The development of targeted sensitive and specific label-free of the Q1 quadrupole transmitting a narrow selected mass range
quantitative techniques compatible with protein and peptide through to the collision cell, a wider window containing multiple
analysis such as multiple reaction monitoring (MRM, also known analytes is allowed through. This generates a complex MS/MS
as selected reaction monitoring, SRM), capable of the multiplex product ion spectrum containing data on all the analytes
analysis of large numbers of targets in a single MS run, has contained within the Q1 m/z window (Fig. 6). High-quality
significantly advanced our understanding of the human extracted ion chromatograms (XICs) can be then generated
proteome, especially for discovery of biomarkers of human post-acquisition to produce MRM-like data. The Q1 window
disease. Various types of instruments capable of two stages of can be stepped across the full mass range (25 m/z unit
mass filtering can be used for MRM assays, although typically a increments), with collection of full-scan composite MS/MS
triple quadrupole (QQQ) instrument is used. Specific proteotypic spectra at each step, within an LC compatible cycle time,
peptides generated by enzymatic degradation of a protein enabling a data-independent LC workflow. Importantly, the
850

wileyonlinelibrary.com/journal/bmc Copyright © 2014 John Wiley & Sons, Ltd. Biomed. Chromatogr. 2014; 28: 848–857
Recent advances in proteomics

Top-down proteomics
Another area that is rapidly advancing is top-down proteomics for
the analysis of intact proteins. Improvement of both the speed
and the quality in the individual steps required for top down
proteomics analysis now allows robust implementation for the
analysis of proteins below 70 kDa. Thus, improved intact protein
separations coupled to state-of-the-art high-performance instru-
mentation (e.g. Fourier transform ion cyclotron resonance or
Orbitrap instruments) are enhancing both the quality and the
number of proteins and protein isoforms that can be detected.
In a recent study 1220 proteins (yielding over 5000 proteoforms)
were identified from a human lung carcinoma cell line, including
300 membrane proteins (Catherman et al., 2013). Importantly,
unlike bottom-up protocols where, owing to enzymatic digestion,
detailed information on PTMs and sequence variants is
compromised, in top-down approaches intact proteins are
analyzed, allowing unequivocal identification and location of
specific modifications.

Antibody-based techniques
Good quality, validated antibody reagents, both polyclonal and
monoclonal, are an essential part of the modern proteomics
toolbox, although unfortunately many researchers have been
badly burnt by reagents of inferior quality supplied by some
manufacturers (Colwill and Graslund, 2011). It must be remem-
bered, however, that antibodies are a case of ‘horses for courses’,
and an antibody that is excellent for immunoprecipitation, for
Figure 5. A comparison of Total Ion Chromatogram (TIC) and multiple example, may not work for immunohistochemistry. Luckily,
reaction monitoring (MRM) analysis of the same RP-HPLC purified fecal
there are now efforts to ensure that validated antibodies against
sample from a patient with colorectal cancer. The proteins targeted for
a defined target can be chosen for a specific application.
the MRM analysis, in order of elution, were hemoglobin, haptoglobin and
carcinoembryonic antigen.
Antibodypedia is a free, open-access, resource for data and
commentary on antibodies from both commercial and academic
providers (http://www.antibodypedia.com/). The November
data can be re-mined ad libitum for alternative targets, or using 2012 release catalogs more than 500,000 individual antibodies,
different product ions or peptides if there is signal overlap. covering 88% of the proteins encoded by the human genome.
SWATH has recently been used to quantitate N-linked glycopro- Antibodypedia also details nearly 145,000 experiments
teins in human plasma with low ng/ml sensitivity (Liu et al., 2013), performed with these antibodies, as well as 32,000 links to
using a hydrazine-based sample preparation protocol for glyco- publications related to their use. Side by side comparison of
protein enrichment. the key properties of antibodies to specific targets can be made.

Figure 6. Schematic of SWATH Workflow (courtesy of Dr Christie Hunter, AbSciex). Unlike MRM, in which a narrow window is set for Q1, in SWATH a
wider window containing multiple analytes is passed. This produces a MS/MS spectrum that is a composite of all the analytes within the Q1 m/z
window. High-quality extracted ion chromatograms (XICs) can be generated post-acquisition to produce the MRM-like data. The Q1 window can be
stepped across the mass range, collecting full-scan composite MS/MS spectra at each step, enabling data-independent LC workflow.
851

Biomed. Chromatogr. 2014; 28: 848–857 Copyright © 2014 John Wiley & Sons, Ltd. wileyonlinelibrary.com/journal/bmc
K. Wang et al.

In collaboration with the HUPO effort, The Human Protein Atlas, proteomics, particularly the use of immunoprecipitation, pull-
launched in 2005, was set up to allow for a systematic explora- downs, epitope tagging (which circumvents the need for spe-
tion of the human proteome using an antibody-based approach cific antibodies against each target), tandem affinity purification
(Uhlen et al., 2010). The goal was to generate at least one (Puig et al., 2001) and chemical cross linking (Rigaut et al., 1999),
antibody to all approximately 20,300 genes in the human are now becoming predominant (Braun 2013). Several early
genome by the end of 2014. Version 11.0, which was released proteomics studies focussed on yeast as a model organism
on 11-03-13, now covers 15,156 genes with protein expression (Gavin et al., 2002, 2006; Ho et al., 2002; Krogan et al., 2006).
profiles based on 18707 antibodies. Key to this approach is the Collectively, these studies covered almost 50% of the yeast
use of protein epitope signature tags (prESTs) (Agaton et al., proteome (Lievens et al., 2010). Importantly, the global land-
2003; Larsson et al., 2006) as immunogens. prESTs contain scape of the membrane interactome in Saccharomyces cerevisiae
selected proteotypic fragments, typically 25–150 residues, of has been characterized (Babu et al., 2012). More recently multi-
the target protein. They are produced in Escherichia coli as fusion ple studies have focussed on the human interactome
proteins with an N-terminal His6-ABP tag: the His6 tag allows (Havugimana et al., 2012). However, it was frequently noted that
single-step protein purification under denaturing conditions the overlap between datasets was poor (Braun, 2013; Lievens
while the albumin-binding protein (ABP) both helps promote et al., 2010), possibly owing to the heterogeneity of both the
an immune response and facilitate quantitation following experimental systems and the processing pipelines.
purification. Polyclonal antibodies are raised in rabbits and Fortunately, it has now been shown (Varjosalo et al., 2013)
immunopurified against the corresponding prEST. It is now that the use of carefully designed standard operating proce-
generally recognized that the rate-limiting step in the high- dures and bioinformatics approaches can overcome this lack
throughput antibody purification is the generation of the target of reproducibility, even between different laboratories. In this
antigen (either as a native protein, recombinant protein or study, the interactomes of 32 human kinases (which were
synthetic peptide): prESTs obviate this and offer a generic used as baits), containing 377 unique proteins and 488
method to rapidly obtain immunogens against all the proteins interactions (Fig. 7), were determined.
encoded by the human genome. While polyclonal antibodies Lambert et al. (2013) have recently used affinity purification
are cheaper to produce and recognize multiple epitopes that coupled with SWATH to characterize changes in protein–protein
can be advantageous for some applications (e.g. secondary anti- interactions imparted by the hsP90 inhibitor nVP-AuY922 or mel-
bodies in sandwich ELISAs), they suffer the disadvantages of anoma-associated mutations in the human kinase cdK4 using
finite supply with each batch requiring fresh validation. Mono- pull down with Flag M2 magnetic beads followed by on-bead
clonal antibodies, by contrast, are a renewable resource where tryptic digestion.
all batches are identical, generally have lower nonspecific
binding than polyclonals, and have a single epitope. This has
stimulated the development of high-throughput methods
Redox proteomics
capable of meeting the global monoclonal antibody demand Thiol groups on cysteine residues of certain proteins (redox sensors)
(Colwill and Graslund, 2011), including the use of a robotic can be direct targets of reactive oxygen species and can be oxidized
approach (Layton et al., 2013). to sulfenic acids and further to sulfinic acids and sulfonic acids
Antibody-based techniques are complementing MS approaches (Leonard and Carroll, 2011). These redox modifications can act as
(immuno-MS) in two main areas: ensuring highly specific targeting an on/off switch for protein activity controlling many important
and enrichment for enhanced selectivity and sensitivity; and biological events such as gene transcription, translation and protein
supporting targeted interactomics studies (see below). A key folding, cell metabolism, signal transduction, cell cycle and cell
example of the first approach is stable isotope standards and death (Brandes et al., 2009; Paulsen and Carroll, 2010; Wouters
capture by anti-peptide antibodies (SISCAPA; Anderson et al., et al., 2011). Characterization of the redox state of redox sensors is
2004). In this approach specific anti-peptide polyclonal antibodies, urgently required in the understanding of cancer biology (Parri
generated in rabbits, are immobilized onto 100 nL affinity columns and Chiarugi, 2013) and stem cell biology (Wang et al., 2013).
to enrich the corresponding target peptide present in a tryptic di- Oxidative isotope-coded affinity tags (OxICAT), a thiol-trapping
gest, along with the corresponding spiked stable-isotope-labeled proteomic technique, was developed by Ursula Jakob’s group to
internal standard. Following elution from the anti-peptide characterize the redoxome in vivo. In this study, the reduced
antibody supports, mass spectrometry is used to quantitate the thiol groups were labeled with light 12C-ICAT reagents under pro-
peptides (both endogenous and labeled). The method was shown tein-denaturing conditions and oxidized thiol groups were labeled
to be compatible with direct multiplex analysis of peptides with heavier 13C-ICAT. The proteins were then subjected to trypsin
representing 47 high to medium abundance proteins in human digestion, affinity chromatography enrichment and LC/MS/MS
serum using MRM (Anderson and Hunter, 2006). The method has identification (Leichert et al., 2008). Using this method, they success-
now been modified to use rabbit monoclonal antibodies and fully produced a snapshot of the in vivo thiol modification status of
automated solid phase extraction MS/MS with a sample cycle time cellular proteins in E. coli (Leichert et al., 2008) and Caenorhabditis
of approximately 7 s (Razavi et al., 2012). elegans (Knoefler et al., 2012). It is believed that OxICAT will be
widely used to visualize the thiol status of proteins in many different
human disease states. In addition, several other proteomic-based
Interactomics technologies, such as nonreducing/reducing ‘diagonal’ polyacryl-
In living systems most proteins function by interaction with amide gel electrophoresis (Cumming et al., 2004), quantitative gel-
other proteins in either stable or dynamic complexes. The sum based redox proteomics (Izquierdo-Alvarez et al., 2012; Leichert
of these interactions in a specific biological system is the and Jakob, 2004), OxMRM (Held et al., 2010), purification of
interactome. While the yeast two-hybrid system was the original reversibly oxidized proteins (Templeton et al., 2010; Victor
mainstay of this technology (Lievens et al., 2010), advances in et al., 2012) and the use of sulfenic acid-specific antibodies
852

wileyonlinelibrary.com/journal/bmc Copyright © 2014 John Wiley & Sons, Ltd. Biomed. Chromatogr. 2014; 28: 848–857
Recent advances in proteomics

Figure 7. The interactomes of 32 human kinases determined by affinity purification mass spectrometry (AP-MS) (reproduced by permission of
Varjosalo et al., 2013). The bait proteins used for affinity purification are shown in the rectangles. The thick lines correspond to interactions present
in the public databases. Black dashed lines indicate known interactions between the proteins identified by AP-MS.

(Seo and Carroll, 2009) and S-glutathionylation-specific antibodies retrieval techniques, similar to those used in immunohistochem-
(Yusuf et al., 2010), have extended the boundaries for detecting istry (Gustafsson et al., 2010), is a possible solution.
and quantifying redox cysteine modifications in a cellular context. A number of interesting clinical applications of IMS have been
reported. These include studies on the molecular physiology of
the eye, determination of human epidermal growth factor
Imaging mass spectrometry receptor 2 (HER2) status in cancer tissues, neuroscience and
Imaging mass spectrometry (IMS) allows region-specific mea- drug analysis (Norris and Caprioli, 2013).
surement of proteins and peptides directly from biological
specimens (e.g. tissue slices) immobilized on a MALDI target,
allowing detection of analytes in situ without the need for Automated sample preparation
labeling (Cornett et al., 2007). The ability to carry out multiplex
analysis of hundreds of analytes in a single experiment without The need, and the ability, to rapidly analyze large numbers of
prior knowledge of tissue composition is most attractive. Funda- biological samples by mass spectrometry in a high-throughput
mental to this technology is sample preparation. Unique to format have stimulated the need to develop compatible
imaging mass spectrometry compared with other mass spectro- automated methods of rapid sample preparation. Automated
metric techniques is that preparation of the sample and MALDI plate spotting was one of the earlier applications where
acquisition of the MS data must be performed in such a way eluent from HPLC columns was collected directly onto the
as to preserve the spatial integrity of the sample within the limits MALDI plates. Automated techniques have been also been
of the spatial resolution of the measurement (Norris and Caprioli, developed using chromatographic column, microplate and
2013). Archival formalin-fixed, paraffin-embedded (FFPE) tissues magnetic bead-based formats, and multidimensional protocols
offer an invaluable source of potential retrospective proteomic can readily be incorporated. Typically some form of robotic
information for such studies (Lemaire et al., 2007), with the liquid handling platform is utilized. Thus, Kreusch et al. (2008)
associated clinical data readily available. However, analysis of combined size exclusion, anion exchange and lectin affinity
archival FFPE tissues by high-throughput proteomic methods chromatography orthogonally using a robotic system developed
has been hindered by the adverse effects of formaldehyde ‘in house’ to analyze human serum. This system was used to
fixation and subsequent histological processing resulting in search for biomarker candidates in sera of patients with Alport
chemical modification of the target proteins, in particular syndrome and severe inflammation. Twenty-three new marker
methylol (hydroxymethyl) adducts and stable cross-linked candidates were detected for Alport syndrome and 33 new for
Schiff’s bases and methylene bridges (Fowler et al., 2013), and severe inflammation (Baum et al., 2008). In another approach,
significant effort has been made in recent years to overcome Loo et al. (2010) developed a panel of seven Lectin-Dynabeads
these problems. It has been found that the use of antigen- for the effective one-step purification of serum glycoproteins in
853

Biomed. Chromatogr. 2014; 28: 848–857 Copyright © 2014 John Wiley & Sons, Ltd. wileyonlinelibrary.com/journal/bmc
K. Wang et al.

96-well microplate format. This method is also being used for home/software/Skyline/begin.view) for SRM/MRM and SWATH
biomarker discovery. quantitative analysis, freely accessible protein databases of protein
Filtered aided sample preparation (FASP) (Wisniewski et al., sequence and functional information such as UniProt (http://www.
2009), is another technique applicable to high-throughput uniprot.org/) and programs such as KEGG (http://www.genome.jp/
sample preparation, using the principle of protein retention kegg/pathway.html), STRING (http://string-db.org/), Cytoscape
and buffer exchange by ultracentrifugation on molecular weight (http://www.cytoscape.org/) and Ingenuity (http://www.ingenu-
cut-off (MWCO) spin columns to enable protein denaturation, ity.com/) for pathway analysis, and gene ontology analysis tools
reduction, alkylation and digestion without sample transfer. such as the Gene Ontology (http://www.geneontology.org/),
FASP has been multiplexed in 96-well filter plate format (Switzar GOEAST (http://omicslab.genetics.ac.cn/GOEAST/) and DAVID
et al., 2013) making it amenable to robotic automation. Cells or Bioinformatics Resources (http://david.abcc.ncifcrf.gov/).
tissues can be lysed in a 96-well plate and then transferred
directly to a 96-well MWCO filter plate for simultaneous process-
ing. In addition to cells and fresh tissues, a recent study showed Databases/browsers
that proteins extracted from FFPE tissues can also be applied to
FASP (called FFPE-FASP; Ostasiewicz et al., 2010). High-throughput proteomic experiments generate huge amounts
As another alternative, combinatorial hexapeptide ligand of peptide data, much of which has been stored and dissemi-
libraries (CPLL), containing millions of different ligands, have nated in multiple databases, such as PeptideAtlas (Deutsch
been used in an affinity saturation strategy to amplify low abun- et al., 2008b), PRIDE (Vizcaino et al., 2010), Human Proteinpedia
dance proteins using what has been termed ‘equalizer (Kandasamy et al., 2009), GPMDB (Craig et al., 2004), neXtProt
technology’ (Di Girolamo et al., 2011). The library is bound onto (Lane et al., 2012) and Human Protein Atlas (Berglund et al.,
solid-phase porous beads, and is now commercially available 2008). To ensure rapid and open sharing of proteomic data,
(ProteoMiner, Bio-Rad, Bio-Rad Laboratories Inc., Hercules, CA, policies for data deposition are being implemented by the
USA). This approach has recently been coupled with MRM (Di leading journals in the field (Kinsinger et al., 2011). The proteomics
Girolamo et al., 2011) using Saccharomyces cerevisiae as a model community is taking steps to ensure that data are made publicly
system. A set of target proteins covering a broad abundance accessible and are of high quality, a challenging task that requires
range was selected to quantitatively evaluate the precision of the development and deployment of methods for measuring and
the approach and its capability to detect low-abundance proteins. documenting data quality metrics. The plethora of data that
A number of instrument companies are now developing resides in these databases needs to be collated into coherent
platforms for high-throughput sample preparation. Agilent and searchable browsers, that are regularly updated, for their
Technologies have developed their AssayMAP Bravo Platform effective interrogation [e.g. to identify ‘missing proteins’, defined
and affinity cartridges that allow 96 samples to be processed as those proteins which have only transcriptomic evidence and
simultaneously. PhyNexus (PhyNexus, Inc., San Jose, CA, USA) a predicted sequence (or are inferred by homology), or those
have developed a range of ‘in tip’ columns (PhyTip) compatible partially identified proteins, in which transcript evidence for the
with their own micro-extractor automated instrument (Le et al., existence of the corresponding protein is available but lacks
2013) as well as other robotic platforms such as Tecan and convincing MS information (Paik et al., 2012)]. To this end, several
Hamilton. Atoll GmbH (Weingarten, Germany) also produce a browsers are currently being developed as part of the HUPO
range of RoboColumns (www.atoll-bio.com) in 96-array format initiative, including The Proteome Browser (Goode et al., 2013)
which can be packed with any chromatographic support. and CAPER (Guo et al., 2013).
Shimadzu (Tokyo, Japan), taking an automated chromatography
approach, market the Perfinity system that integrates automated
affinity selection, buffer exchange, digestion, desalting and
reversed-phase separation, allowing the generation and
Conclusions
purification of peptides from serum for mass-spectral analysis We are now entering an era where recent advances in
in approximately 10 min. It should be noted that robotics may proteomics, coupled with genomics, transcriptomics and bioin-
also be used to rapidly optimize chromatographic and affinity formatics, will rapidly populate the human genome. This will
elution conditions. lead to improved understanding of human biology, both in the
normal and the diseased state, which will bring with it enormous
economic benefits (Hood et al., 2012). This heralds the arrival of
Bioinformatic tools for data analysis personalized medicine, which will have an increased focus on
Data processing is undoubtedly a critical component in any wellness rather than disease and which will revolutionize
proteomics experiment, and is frequently the most time- treatment decisions guided by the patient’s unique circum-
consuming. Recent years have seen significant advances in the stances and characteristics based on the individual’s specific
field of proteomics-related bioinformatics and, in addition to genome and proteome.
the proprietary software supplied by many of the instrument However, as pointed out in a recent article by George Poste
manufacturers, many free and sophisticated open-source soft- (2012), such a path is not without obstacles. In particular,
ware tools are currently available (Deutsch et al., 2008a). multidisciplinary teams involving multi-institution cooperation
Examples include Proteome Discoverer (Thermo Fisher Scientific will be required. The roles of bodies such as HUPO will be
Inc., Waltham, MA, USA), SIEVE Software (Thermo Fisher Scientific imperative for co-ordinating this. In addition, significant funding
Inc.), Mascot Daemon (Matrix Science, London, UK), MaxQuant will be required to support this big science big data initiative,
(http://www.maxquant.org/), the Trans-Proteomic Pipeline, a suite involving a major paradigm shift in how funds are allocated,
of software tools for the analysis of MS/MS datasets (Deutsch et al., without which the cohesion required for such a ‘big science
2010), Skyline (https://skyline.gs.washington.edu/labkey/project/ big data’ project will not be possible.
854

wileyonlinelibrary.com/journal/bmc Copyright © 2014 John Wiley & Sons, Ltd. Biomed. Chromatogr. 2014; 28: 848–857
Recent advances in proteomics

Dell A and Morris HR. Fast atom bombardment – high field magnet mass
Acknowledgments spectrometry of 6000 dalton polypeptides. Biochemical and Biophysi-
We gratefully acknowledge Dr Alun Jones (University of Queens- cal Research Communications 1982; 106(4): 1456–1462.
land) who performed the assay shown in Fig. 4. Edouard Nice was Deutsch EW, Lam H and Aebersold R. Data analysis and bioinformatics
supported, in part, by NHMRC project grant 603130, 1017078 and tools for tandem mass spectrometry in proteomics. Physiology and
development grant 1017078. In this special issue I would particularly Genomics 2008a; 33(1): 18–25.
Deutsch EW, Lam H and Aebersold R. PeptideAtlas: a resource for target
like to acknowledge both over 35 years of scientific interaction with
selection for emerging targeted proteomics workflows. EMBO Reports
Chang Kee, and, most importantly, our long-term friendship. 2008b; 9(5): 429–434.
Deutsch EW, Mendoza L, Shteynberg D, Farrah T, Lam H, Tasman N, Sun
Z, Nilsson E, Pratt B, Prazen B, Eng JK, Martin DB, Nesvizhskii AI and
References Aebersold R. A guided tour of the Trans-Proteomic Pipeline.
Agaton C, Galli J, Hoiden Guthenberg I, Janzon L, Hansson M, Asplund A, Proteomics 2010; 10(6): 1150–1159.
Brundell E, Lindberg S, Ruthberg I, Wester K, Wurtz D, Hoog C, Di Girolamo F, Boschetti E, Chung MC, Guadagni F and Righetti PG.
Lundeberg J, Stahl S, Ponten F and Uhlen M. Affinity proteomics for ‘Proteomineering’ or not? The debate on biomarker discovery in sera
systematic protein profiling of chromosome 21 gene products in human continues. Journal of Proteomics 2011; 74(5): 589–594.
tissues. Molecular and Cellular Proteomics 2003; 2(6): 405–414. Domanski D, Percy AJ, Yang J, Chambers AG, Hill JS, Freue GV and
Anderson L and Hunter CL. Quantitative mass spectrometric multiple Borchers CH. MRM-based multiplexed quantitation of 67 putative
reaction monitoring assays for major plasma proteins. Molecular cardiovascular disease biomarkers in human plasma. Proteomics
and Cellular Proteomics 2006; 5(4): 573–588. 2012; 12(8): 1222–1243.
Anderson NL, Anderson NG, Haines LR, Hardie DB, Olafson RW and Pear- Edman P. Method for determination of the amino acid sequence in
son TW. Mass spectrometric quantitation of peptides and proteins peptides. Acta Chemica Scandinavica 1950; 4(2): 283–293.
using stable isotope standards and capture by anti-peptide anti- Fenn JB, Mann M, Meng CK, Wong SF and Whitehouse CM. Electrospray
bodies (SISCAPA). Journal of Proteome Research 2004; 3(2): 235–244. ionization for mass spectrometry of large biomolecules. Science 1989;
Ang CS and Nice EC. Targeted in-gel MRM: a hypothesis driven approach 246(4926): 64–71.
for colorectal cancer biomarker discovery in human feces. Journal of Fowler CB, O’Leary TJ and Mason JT. Toward improving the proteomic
Proteome Research 2010; 9(9): 4346–4355. analysis of formalin-fixed, paraffin-embedded tissue. Expert Reviews
Ang CS, Rothacker J, Patsiouras H, Gibbs P, Burgess AW and Nice EC. Use in Proteomics 2013; 10(4): 389–400.
of multiple reaction monitoring for multiplex analysis of colorectal Gaudet P, Argoud-Puy G, Cusin I, Duek P, Evalet O, Gateau A, Gleizes A,
cancer-associated proteins in human feces. Electrophoresis 2011; Pereira M, Zahn-Zabal M, Zwahlen C, Bairoch A and Lane L. neXtProt:
32(15): 1926–1938. organizing protein knowledge in the context of human proteome
Babu M, Vlasblom J, Pu S, Guo X, Graham C, Bean BD, Burston HE, projects. Journal of Proteome Research 2013; 12(1): 293–298.
Vizeacoumar FJ, Snider J, Phanse S, Fong V, Tam YY, Davey M, Gavin AC, Bosche M, Krause R, Grandi P, Marzioch M, Bauer A, Schultz J, Rick
Hnatshak O, Bajaj N, Chandran S, Punna T, Christopolous C, Wong JM, Michon AM, Cruciat CM, Remor M, Hofert C, Schelder M, Brajenovic
V, Yu A, Zhong G, Li J, Stagljar I, Conibear E, Wodak SJ, Emili A and M, Ruffner H, Merino A, Klein K, Hudak M, Dickson D, Rudi T, Gnau V,
Greenblatt JF. Interaction landscape of membrane–protein com- Bauch A, Bastuck S, Huhse B, Leutwein C, Heurtier MA, Copley RR,
plexes in Saccharomyces cerevisiae. Nature 2012; 489(7417): 585–589. Edelmann A, Querfurth E, Rybin V, Drewes G, Raida M, Bouwmeester
Barber M, Bordoli RS, Sedgwick RD and Tyler AN. Fast atom bombard- T, Bork P, Seraphin B, Kuster B, Neubauer G and Superti-Furga G.
ment of solids (Fab) – a new ion-source for mass-spectrometry. Functional organization of the yeast proteome by systematic analysis
Chemical Communications 1981; 7: 325–327. of protein complexes. Nature 2002; 415(6868): 141–147.
Baum A, Pohl M, Kreusch S, Cumme GA, Ditze G, Misselwitz J, Kiehntopf Gavin AC, Aloy P, Grandi P, Krause R, Boesche M, Marzioch M, Rau C,
M, Udby L, Meier-Hellmann A and Rhode H. Searching biomarker Jensen LJ, Bastuck S, Dumpelfeld B, Edelmann A, Heurtier MA, Hoff-
candidates in serum using multidimensional native chromatography. man V, Hoefert C, Klein K, Hudak M, Michon AM, Schelder M, Schirle
II. Method evaluation with Alport syndrome and severe inflammation. M, Remor M, Rudi T, Hooper S, Bauer A, Bouwmeester T, Casari G,
Journal of Chromatography B: Analytical Technology in Biomedicine Drewes G, Neubauer G, Rick JM, Kuster B, Bork P, Russell RB and
and Life Sciences 2008; 876(1): 31–40. Superti-Furga G. Proteome survey reveals modularity of the yeast cell
Berglund L, Bjorling E, Oksvold P, Fagerberg L, Asplund A, Szigyarto CA, machinery. Nature 2006; 440(7084): 631–636.
Persson A, Ottosson J, Wernerus H, Nilsson P, Lundberg E, Sivertsson Gillet LC, Navarro P, Tate S, Rost H, Selevsek N, Reiter L, Bonner R and
A, Navani S, Wester K, Kampf C, Hober S, Ponten F and Uhlen M. A Aebersold R. Targeted data extraction of the MS/MS spectra gener-
genecentric Human Protein Atlas for expression profiles based on ated by data-independent acquisition: a new concept for consistent
antibodies. Molecular and Cellular Proteomics 2008; 7(10): 2019–2027. and accurate proteome analysis. Molecular and Cellular Proteomics
Brandes N, Schmitt S and Jakob U. Thiol-based redox switches in eukaryotic 2012; 11(6): O111 016717.
proteins. Antioxidant and Redox Signal 2009; 11(5): 997–1014. Goode RJ, Yu S, Kannan A, Christiansen JH, Beitz A, Hancock WS, Nice E
Braun P. Reproducibility restored – on toward the human interactome. and Smith AI. The proteome browser web portal. Journal of Proteome
Nature Methods 2013; 10(4): 301, 303. Research 2013; 12(1): 172–178.
Catherman AD, Durbin KR, Ahlf DR, Early BP, Fellers RT, Tran JC, Thomas Guo F, Wang D, Liu Z, Lu L, Zhang W, Sun H, Zhang H, Ma J, Wu S, Li N,
PM and Kelleher NL. Large-scale top-down proteomics of the human Jiang Y, Zhu W, Qin J, Xu P, Li D and He F. CAPER: a chromosome-
proteome: membrane proteins, mitochondria, and senescence. assembled human proteome browsER. Journal of Proteome Research
Molecular and Cellular Proteomics 2013; 12(12): 3465–3473. 2013; 12(1): 179–186.
Chen YT, Chen HW, Domanski D, Smith DS, Liang KH, Wu CC, Chen CL, Chung T, Gustafsson JO, Oehler MK, McColl SR and Hoffmann P. Citric acid antigen
Chen MC, Chang YS, Parker CE, Borchers CH and Yu JS. Multiplexed quan- retrieval (CAAR) for tryptic peptide imaging directly on archived
tification of 63 proteins in human urine by multiple reaction monitoring- formalin-fixed paraffin-embedded tissue. Journal of Proteome
based mass spectrometry for discovery of potential bladder cancer Research 2010; 9(9): 4315–4328.
biomarkers. Journal of Proteomics 2012; 75(12): 3529–3545. Hakansson P, Kamensky I, Sundqvist B, Fohlman J, Peterson P, McNeal CJ and
Colwill K and Graslund S. A roadmap to generate renewable protein Macfarlane RD. I-127-labeled plasma desorption mass-spectrometry of in-
binders to the human proteome. Nature Methods 2011; 8(7): 551–558. sulin. Journal of the American Chemistry Society 1982; 104(10): 2948–2949.
Cornett DS, Reyzer ML, Chaurand P and Caprioli RM. MALDI imaging Hardman M and Makarov AA. Interfacing the orbitrap mass analyzer to an
mass spectrometry: molecular snapshots of biochemical systems. electrospray ion source. Analytical Chemistry 2003; 75(7): 1699–1705.
Nature Methods 2007; 4(10): 828–833. Havugimana PC, Hart GT, Nepusz T, Yang H, Turinsky AL, Li Z, Wang PI,
Craig R, Cortens JP and Beavis RC. Open source system for analyzing, Boutz DR, Fong V, Phanse S, Babu M, Craig SA, Hu P, Wan C, Vlasblom
validating, and storing protein identification data. Journal of Prote- J, Dar VU, Bezginov A, Clark GW, Wu GC, Wodak SJ, Tillier ER,
ome Research 2004; 3(6): 1234–1242. Paccanaro A, Marcotte EM and Emili A. A census of human soluble
Cumming RC, Andon NL, Haynes PA, Park M, Fischer WH and Schubert D. protein complexes. Cell 2012; 150(5): 1068–1081.
Protein disulfide bond formation in the cytoplasm during oxidative Held JM, Danielson SR, Behring JB, Atsriku C, Britton DJ, Puckett RL, Schilling
stress. Journal of Biological Chemistry 2004; 279(21): 21749–21758. B, Campisi J, Benz CC and Gibson BW. Targeted quantitation of site-
855

Biomed. Chromatogr. 2014; 28: 848–857 Copyright © 2014 John Wiley & Sons, Ltd. wileyonlinelibrary.com/journal/bmc
K. Wang et al.

specific cysteine oxidation in endogenous proteins using a differential Lane L, Argoud-Puy G, Britan A, Cusin I, Duek PD, Evalet O, Gateau A,
alkylation and multiple reaction monitoring mass spectrometry Gaudet P, Gleizes A, Masselot A, Zwahlen C and Bairoch A. neXtProt:
approach. Molecular and Cellular Proteomics 2010; 9(7): 1400–1410. a knowledge platform for human proteins. Nucleic Acids Research
Ho Y, Gruhler A, Heilbut A, Bader GD, Moore L, Adams SL, Millar A, Taylor 2012; 40(Database issue): D76–83.
P, Bennett K, Boutilier K, Yang L, Wolting C, Donaldson I, Schandorff S, Lange V, Picotti P, Domon B and Aebersold R. Selected reaction monitor-
Shewnarane J, Vo M, Taggart J, Goudreault M, Muskat B, Alfarano C, ing for quantitative proteomics: a tutorial. Molecular Systems Biology
Dewar D, Lin Z, Michalickova K, Willems AR, Sassi H, Nielsen PA, 2008; 4: 222.
Rasmussen KJ, Andersen JR, Johansen LE, Hansen LH, Jespersen H, Larsson K, Wester K, Nilsson P, Uhlen M, Hober S and Wernerus H.
Podtelejnikov A, Nielsen E, Crawford J, Poulsen V, Sorensen BD, Multiplexed PrEST immunization for high-throughput affinity proteo-
Matthiesen J, Hendrickson RC, Gleeson F, Pawson T, Moran MF, mics. Journal of Immunology Methods 2006; 315(1–2): 110–120.
Durocher D, Mann M, Hogue CW, Figeys D and Tyers M. Systematic Layton D, Laverty C and Nice EC. Design and operation of an automated
identification of protein complexes in Saccharomyces cerevisiae by high-throughput monoclonal antibody facility. Biophysics Reviews
mass spectrometry. Nature 2002; 415(6868): 180–183. 2013; 5(1): 47–55.
Hood LE, Omenn GS, Moritz RL, Aebersold R, Yamamoto KR, Amos M, Le ME, Vizel A and Hutterer KM. Automated sample preparation for CE-
Hunter-Cevera J and Locascio L. New and improved proteomics SDS. Electrophoresis 2013; 34(9–10): 1369–1374.
technologies for understanding complex biological systems: address- Legrain P, Aebersold R, Archakov A, Bairoch A, Bala K, Beretta L, Bergeron
ing a grand challenge in the life sciences. Proteomics 2012; 12(18): J, Borchers CH, Corthals GL, Costello CE, Deutsch EW, Domon B, Han-
2773–2783. cock W, He F, Hochstrasser D, Marko-Varga G, Salekdeh GH, Sechi S,
Hu Q, Noll RJ, Li H, Makarov A, Hardman M and Graham Cooks R. The Snyder M, Srivastava S, Uhlen M, Wu CH, Yamamoto T, Paik YK and
Orbitrap: a new mass spectrometer. Journal of Mass Spectrometry Omenn GS. The human proteome project: current state and future di-
2005; 40(4): 430–443. rection. Molecular and Cellular Proteomics 2011; 10(7): M111 009993.
Izquierdo-Alvarez A, Ramos E, Villanueva J, Hernansanz-Agustin P, Leichert LI and Jakob U. Protein thiol modifications visualized in vivo.
Fernandez-Rodriguez R, Tello D, Carrascal M and Martinez-Ruiz A. PLoS Biology 2004; 2(11): e333.
Differential redox proteomics allows identification of proteins revers- Leichert LI, Gehrke F, Gudiseva HV, Blackwell T, Ilbert M, Walker AK,
ibly oxidized at cysteine residues in endothelial cells in response to Strahler JR, Andrews PC and Jakob U. Quantifying changes in the
acute hypoxia. Journal of Proteomics 2012; 75(17): 5449–5462. thiol redox proteome upon oxidative stress in vivo. Proceedings of
Kandasamy K, Keerthikumar S, Goel R, Mathivanan S, Patankar N, the National Academy of Science USA 2008; 105(24): 8197–8202.
Shafreen B, Renuse S, Pawar H, Ramachandra YL, Acharya PK, Lemaire R, Desmons A, Tabet JC, Day R, Salzet M and Fournier I. Direct
Ranganathan P, Chaerkady R, Keshava Prasad TS and Pandey A. analysis and MALDI imaging of formalin-fixed, paraffin-embedded
Human Proteinpedia: a unified discovery resource for proteomics re- tissue sections. Journal of Proteome Research 2007; 6(4): 1295–1305.
search. Nucleic Acids Research 2009; 37(Database issue): D773–781. Leonard SE and Carroll KS. Chemical ‘omics’ approaches for understand-
Karas M and Hillenkamp F. Laser desorption ionization of proteins with ing protein cysteine oxidation in biology. Current Opinion in Chemis-
molecular masses exceeding 10,000 daltons. Analytical Chemistry try and Biology 2011; 15(1): 88–102.
1988; 60(20): 2299–2301. Lievens S, Eyckerman S, Lemmens I and Tavernier J. Large-scale protein
Karlsson KE and Novotny M. Separation efficiency of slurry-packed liquid interactome mapping: strategies and opportunities. Expert Reviews
chromatography microcolumns with very small inner diameters. in Proteomics 2010; 7(5): 679–690.
Analytical Chemistry 1988; 60(17): 1662–1665. Liu Y, Huttenhain R, Surinova S, Gillet LC, Mouritsen J, Brunner R, Navarro
Kinsinger CR, Apffel J, Baker M, Bian X, Borchers CH, Bradshaw R, Brusniak P and Aebersold R. Quantitative measurements of N-linked glyco-
MY, Chan DW, Deutsch EW, Domon B, Gorman J, Grimm R, Hancock proteins in human plasma by SWATH-MS. Proteomics 2013; 13(8):
W, Hermjakob H, Horn D, Hunter C, Kolar P, Kraus HJ, Langen H, 1247–1256.
Linding R, Moritz RL, Omenn GS, Orlando R, Pandey A, Ping P, Rahbar Loo D, Jones A and Hill MM. Lectin magnetic bead array for biomarker
A, Rivers R, Seymour SL, Simpson RJ, Slotta D, Smith RD, Stein SE, discovery. Journal of Proteome Research 2010; 9(10): 5496–5500.
Tabb DL, Tagle D, Yates JR, 3rd and Rodriguez H. Recommendations Macfarlane RD and Torgerson DF. Californium-252 plasma desorption
for mass spectrometry data quality metrics for open access data mass spectroscopy. Science 1976; 191(4230): 920–925.
(corollary to the Amsterdam Principles). Molecular and Cellular Makarov A. Electrostatic axially harmonic orbital trapping: a high-
Proteomics 2011; 10(12): O111 015446. performance technique of mass analysis. Analytical Chemistry 2000;
Klose J and Kobalz U. Two-dimensional electrophoresis of proteins: an 72(6): 1156–1162.
updated protocol and implications for a functional analysis of the Mann M and Wilm M. Error-tolerant identification of peptides in
genome. Electrophoresis 1995; 16(6): 1034–1059. sequence databases by peptide sequence tags. Analytical Chemistry
Knoefler D, Thamsen M, Koniczek M, Niemuth NJ, Diederich AK and 1994; 66(24): 4390–4399.
Jakob U. Quantitative in vivo redox sensors uncover oxidative stress Meng CK, Mann M and Fenn JB. Applications of electrospray liquid
as an early event in life. Molecule and Cell 2012; 47(5): 767–776. chromatography/mass spectrometry. Zeitschrift für Physik D Atoms,
Kocher T, Pichler P, Swart R and Mechtler K. Analysis of protein mixtures Molecules and Clusters 1988; 10: 361–368.
from whole-cell extracts by single-run nanoLC-MS/MS using ultralong Nice EC. Micropreparative LC of proteins. Nature 1990; 348(6300): 462–463.
gradients. Nature Protocol 2012; 7(5): 882–890. Nice E. Biomarker discovery and validation: the tide is turning. Expert Re-
Kreusch S, Schulze M, Cumme GA, Ditze G, Moore T and Rhode H. views in Proteomics 2013; 10(6): 505–507.
Searching biomarker candidates in serum using multidimensional Nice EC, Rothacker J, Weinstock J, Lim L and Catimel B. Use of
native chromatography. I. Enhanced separation method. Journal of multidimensional separation protocols for the purification of trace com-
Chromatography B: Analytical Technology in Biomedicine and Life ponents in complex biological samples for proteomics analysis. Journal
Sciences 2008; 875(2): 567–572. of Chromatography A 2007; 1168(1–2): 190–210; discussion 189.
Krogan NJ, Cagney G, Yu H, Zhong G, Guo X, Ignatchenko A, Li J, Pu S, Norris JL and Caprioli RM. Analysis of tissue specimens by matrix-assisted
Datta N, Tikuisis AP, Punna T, Peregrin-Alvarez JM, Shales M, Zhang laser desorption/ionization imaging mass spectrometry in biological
X, Davey M, Robinson MD, Paccanaro A, Bray JE, Sheung A, Beattie B, and clinical research. Chemistry Reviews 2013; 113(4): 2309–2342.
Richards DP, Canadien V, Lalev A, Mena F, Wong P, Starostine A, Canete O’Farrell PH. High resolution two-dimensional electrophoresis of pro-
MM, Vlasblom J, Wu S, Orsi C, Collins SR, Chandran S, Haw R, Rilstone JJ, teins. Journal of Biological Chemistry 1975; 250(10): 4007–4021.
Gandi K, Thompson NJ, Musso G, St Onge P, Ghanny S, Lam MH, O’Farrell PH. The pre-omics era: the early days of two-dimensional gels.
Butland G, Altaf-Ul AM, Kanaya S, Shilatifard A, O’Shea E, Weissman Proteomics 2008; 8(23–24): 4842–4852.
JS, Ingles CJ, Hughes TR, Parkinson J, Gerstein M, Wodak SJ, Emili A Omenn GS. The strategy, organization, and progress of the HUPO Human
and Greenblatt JF. Global landscape of protein complexes in the yeast Proteome Project. Journal of Proteomics 2013; S1874-3919(13): 00523-X.
Saccharomyces cerevisiae. Nature 2006; 440(7084): 637–643. doi: 10.1016/j.jprot.2013.10.012.
Lambert JP, Ivosev G, Couzens AL, Larsen B, Taipale M, Lin ZY, Zhong Q, Ostasiewicz P, Zielinska DF, Mann M and Wisniewski JR. Proteome,
Lindquist S, Vidal M, Aebersold R, Pawson T, Bonner R, Tate S and phosphoproteome, and N-glycoproteome are quantitatively pre-
Gingras AC. Mapping differential interactomes by affinity purification served in formalin-fixed paraffin-embedded tissue and analyzable
coupled with data-independent mass spectrometry acquisition. Na- by high-resolution mass spectrometry. Journal of Proteome Research
ture Methods 2013; 10(12): 1239–1245. 2010; 9(7): 3688–3700.
856

wileyonlinelibrary.com/journal/bmc Copyright © 2014 John Wiley & Sons, Ltd. Biomed. Chromatogr. 2014; 28: 848–857
Recent advances in proteomics

Paik YK, Omenn GS, Uhlen M, Hanash S, Marko-Varga G, Aebersold R, Templeton DJ, Aye MS, Rady J, Xu F and Cross JV. Purification of revers-
Bairoch A, Yamamoto T, Legrain P, Lee HJ, Na K, Jeong SK, He F, Binz ibly oxidized proteins (PROP) reveals a redox switch controlling p38
PA, Nishimura T, Keown P, Baker MS, Yoo JS, Garin J, Archakov A, MAP kinase activity. PLoS One 2010; 5(11): e15012.
Bergeron J, Salekdeh GH and Hancock WS. Standard guidelines for Thakur SS, Geiger T, Chatterjee B, Bandilla P, Frohlich F, Cox J and Mann
the chromosome-centric human proteome project. Journal of M. Deep and highly sensitive proteome coverage by LC-MS/MS with-
Proteome Research 2012; 11(4): 2005–2013. out prefractionation. Molecular and Cellular Proteomics 2011; 10(8):
Parri M and Chiarugi P. Redox molecular machines involved in tumor M110 003699.
progression. Antioxidant and Redox Signal 2013; 19(15): 1828–1845. Tong D, Moritz RL, Eddes JS, Reid GE, Rasmussen RK, Dorow DS and
Paulsen CE and Carroll KS. Orchestrating redox signaling networks Simpson RJ. Fabrication of stable packed capillary reversed-phase
through regulatory cysteine switches. ACS Chemistry and Biology columns for protein structural analysis. Journal of Protein Chem.
2010; 5(1): 47–62. 1997; 16(5): 425–431.
Poste G. Bring on the biomarkers. Nature 2011; 469(7329): 156–157. Uhlen M, Oksvold P, Fagerberg L, Lundberg E, Jonasson K, Forsberg M,
Poste G. Biospecimens, biomarkers, and burgeoning data: the imperative Zwahlen M, Kampf C, Wester K, Hober S, Wernerus H, Bjorling L and
for more rigorous research standards. Trends in Molecular Medicine Ponten F. Towards a knowledge-based Human Protein Atlas. Nature
2012; 18(12): 717–722. Biotechnology 2010; 28(12): 1248–1250.
Puig O, Caspary F, Rigaut G, Rutz B, Bouveret E, Bragado-Nilsson E, Wilm Varjosalo M, Sacco R, Stukalov A, van Drogen A, Planyavsky M, Hauri S,
M and Seraphin B. The tandem affinity purification (TAP) method: a Aebersold R, Bennett KL, Colinge J, Gstaiger M and Superti-Furga G.
general procedure of protein complex purification. Methods 2001; Interlaboratory reproducibility of large-scale human protein–complex
24(3): 218–229. analysis by standardized AP-MS. Nature Methods 2013; 10(4): 307–314.
Razavi M, Frick LE, LaMarr WA, Pope ME, Miller CA, Anderson NL and Victor KG, Rady JM, Cross JV and Templeton DJ. Proteomic profile of
Pearson TW. High-throughput SISCAPA quantitation of peptides from reversible protein oxidation using PROP, purification of reversibly
human plasma digests by ultrafast, liquid chromatography–free mass oxidized proteins. PLoS One 2012; 7(2): e32527.
spectrometry. Journal of Proteome Research 2012; 11(12): 5642–5649. Vizcaino JA, Cote R, Reisinger F, Barsnes H, Foster JM, Rameseder J,
Rigaut G, Shevchenko A, Rutz B, Wilm M, Mann M and Seraphin B. A ge- Hermjakob H and Martens L. The Proteomics Identifications database:
neric protein purification method for protein complex characteriza- 2010 update. Nucleic Acids Research 2010; 38(Database issue):
tion and proteome exploration. Nature Biotechnology 1999; 17(10): D736–742.
1030–1032. Wang K, Zhang T, Dong Q, Nice EC, Huang C and Wei Y. Redox
Roepstorff P. Mass spectrometry in the analysis of peptides and proteins, homeostasis: the linchpin in stem cell self-renewal and differentia-
past and present. Methods in Molecular Biology 1996; 61: 1–7. tion. Cell Death and Disease 2013; 4: e537.
Seo YH and Carroll KS. Profiling protein thiol oxidation in tumor cells Wisniewski JR, Zougman A, Nagaraj N and Mann M. Universal sample
using sulfenic acid-specific antibodies. Proceedings of the National preparation method for proteome analysis. Nature Methods 2009;
Academy of Science USA 2009; 106(38): 16163–16168. 6(5): 359–362.
Service RF. Proteomics. Proteomics ponders prime time. Science 2008; Wouters MA, Iismaa S, Fan SW and Haworth NL. Thiol-based redox signal-
321(5897): 1758–1761. ling: rust never sleeps. International Journal of Biochemistry and Cell
Stahl-Zeng J, Lange V, Ossola R, Eckhardt K, Krek W, Aebersold R and Biology 2011; 43(8): 1079–1085.
Domon B. High sensitivity detection of plasma proteins by multiple Yusuf MA, Chuang T, Bhat GJ and Srivenugopal KS. Cys-141
reaction monitoring of N-glycosites. Molecular and Cellular Proteomics glutathionylation of human p53: Studies using specific polyclonal an-
2007; 6(10): 1809–1817. tibodies in cancer samples and cell lines. Free Radical Biology and
Switzar L, van Angeren J, Pinkse M, Kool J and Niessen WM. A high- Medicine 2010; 49(5): 908–917.
throughput sample preparation method for cellular proteomics using Zolg W. The proteomic search for diagnostic biomarkers: lost in transla-
96-well filter plates. Proteomics 2013; 13(20): 2980–2983. tion? Molecule and Cell Proteomics 2006; 5(10): 1720–1726.

857

Biomed. Chromatogr. 2014; 28: 848–857 Copyright © 2014 John Wiley & Sons, Ltd. wileyonlinelibrary.com/journal/bmc

También podría gustarte