Sie sind auf Seite 1von 20

Lucia Monteoliva

is currently a visiting scientist


Differential proteomics:
(postdoctoral researcher) at
the proteomics facility of the
National Center for
An overview of gel and non-
Biotechnology, Madrid, Spain.
Juan Pablo Albar
gel based approaches
is a senior research scientist at Lucia Monteoliva and Juan Pablo Albar
the Spanish Council for Date received (in revised form): 18th October 2004
Scientific Research (CSIC) and
Head of the proteomics facility
at the National Center for Abstract
Biotechnology, Madrid, Spain.
The comprehensive analysis of gene expression in complex biological systems has demanded
the development of new technologies to study the cell transcriptome and the cell proteome.
Each approach has advantages and disadvantages from both the conceptual and the
methodological viewpoints. Differential proteomics, the comparison of distinct proteomes
(eg normal versus diseased cells, diseased versus treated cells etc) is of paramount importance.
Several approaches can be used and these typically involve electrophoresis and/or
chromatography combined with chemical or metabolic labelling and mass spectrometry. These
Keywords: Differential approaches aim to identify molecular targets, namely proteins, involved in different
proteomics, quantitative
physiopathological states. Incorporating this knowledge with knowledge from other
proteomics, DIGE, SILAC,
ICAT, SELDI-TOF technologies lays the foundations of active principles at the molecular level. Here, the various
gel- and non-gel-based approaches that are used in a wide range of biological systems for the
study of differentially expressed proteins will be reviewed.

INTRODUCTION sequencing projects, and the need for


The science of proteomics, one of the deciphering this information, the focus is
most important areas of research in the now on proteins, their structure, function,
post-genomic era, is not new in terms of interactions and modifications. Functional
its experimental foundations. It has, genomics focuses on the characterisation
nonetheless, profited from unprecedented of subcellular interactions between
advances in genome sequencing, proteins, as well as the assessment of
bioinformatics and the development of macromolecular complex components.
robust, sensitive, reliable and reproducible Elucidation of protein function is
analytical techniques. approached by characterising the
Genomics projects have produced a interactions that take place within cells;
large number of DNA sequences from a this approach helps to clarify the concept
wide range of organisms, including that proteins tend to form large
humans and mammals. The complete complexes rather than acting
genomes for 215 organisms were available independently.
by August 2004; of these, 28 were Classical proteomics work involves a
eukaryotes. Together with the study of separation step — usually two-
RNA expression levels, proteomics is dimensional gel electrophoresis (2-DE) —
associated with the analysis of global followed by an identification step, usually
protein expression in cells, organisms, mass spectrometry (MS).3 Proteins
Juan Pablo Albar,
Proteomics Facility, tissues and organelles.1,2 Proteomics is a resolved by 2-DE can be identified by in-
Centro Nacional de Biotecnologia research field that gathers environmental gel trypsin digestion via peptide mass
(CSIC),
UAM Campus Cantoblanco,
and genetic factors, and the ‘proteome’ fingerprinting (PMF) using MS or tandem
Darwin, 3, represents the functional status of a mass spectrometry (MS/MS).4 Alternative
Madrid, E-28049, Spain biological compartment. strategies currently involve the use of: (1)
Tel: +34 91 585 4696
In view of the vast amount of in-solution protein extract digestion; (2)
Fax: +34 91 585 4506 information generated by genome peptide fractionation by liquid

2 2 0 & HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004


Differential proteomics

chromatography or multidimensional flight mass spectrometry (TOF-MS)


liquid chromatography (Mud-LC) enables differential protein expression
coupled to electrospray ionisation ion trap profiling of complex protein mixtures
tandem mass spectrometry (ESI-MS/MS); separated by on-chip retentate
and (3) peptide sequencing obtained chromatography. In this paper, the use of
2-DE, LC-ESI-MS and through analysis of fragmentation spectra these and other related alternative
SELDI MS as tools for (MS/MS) — proteins are then identified approaches for the differential protein
differential proteomics using computer algorithms and database expression analysis will be discussed.11
searches.5 Other recent approaches
compare different proteomes (‘differential GEL-BASED APPROACHES
expression proteomics’) and involve Differential display via two-
electrophoresis and/or chromatography dimensional gel electrophoresis
combined with metabolic or chemical Classical proteomics work involves: (1) a
labelling. separation step in which proteins of
2-DE is a relatively simple visual interest are separated by 2-DE (isoelectric
method for mapping differences in focusing [IEF] followed by separation as a
protein expression. There are certain function of molecular mass); (2) protein
limitations to the universal use of this visualisation and image analysis; (3)
technology, such as low detection excising the spots to be analysed; (4) in-
sensitivity and linearity, poor solubility of gel digestion of proteins and pooling of
membrane proteins, limited loading the released peptides; (5) analysis of the
capacity of gradient pH strips, peptide mass fingerprint (PMF) for every
reproducibility of gels, relatively low digested protein through matrix-assisted
throughput and low linear range of laser desorption/ionisation TOF-MS
visualisation procedures.6 It is, (MALDI-TOF MS); (6) matching peptide
nevertheless, currently the most rapid masses against protein databases to obtain
method for direct targeting of protein candidate proteins;3 and (7) validating
expression differences. Some of the identification by acquisition of MS/MS
drawbacks can be circumvented using the spectra of selected peptides to confirm
differential in-gel electrophoresis (DIGE) their sequences (Figure 1).4,12 Since
system developed by Amersham immobilised pH gradient strips (IPG)
Biosciences;7,8 with this technology, were developed (for a review, see refs. 13
fluorescent labelling of cell extracts with and 14), variability in experimental
one of three fluorescent dyes (Cy2, Cy3 conditions has decreased; this is now
or Cy5) is done prior to gel separation undoubtedly the most widespread strategy
and cellular protein levels can be for comparing distinct states of two
compared within a single gel. proteomes.
Another approach, multidimensional Different protein visualisation methods
(ionic exchange, affinity and reverse- are available. Radiolabelling is very
phase) nano- or capillary chromatography sensitive, but is hazardous and expensive.
in conjunction with MS/MS, provides Colloidal Coomassie blue is an easy to
unprecedented tools that enable use, low-cost staining agent, but has poor
Differential protein comparison of differentially expressed detectability and sensitivity and a small
display has traditionally proteins. Labelling techniques such as linear dynamic range. Silver staining is
been based on 2-DE isotope-coded affinity tags (ICATs),9,10 the most commonly used technique, as it
silver-stained gels which combine differential chemical detects as small a quantity as a single
isotope tagging of cell lysates, nanogram, but its dynamic range is also
multidimensional capillary restricted to a single order of magnitude
chromatography and MS/MS, have been scale; in addition, silver staining is not
used extensively for differential display quantitative, as different proteins tend to
analysis. Surface-enhanced laser interact with silver ions differently.
desorption/ionisation (SELDI) time-of- Finally, despite of its high cost,

& HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004 2 2 1


Monteoliva and Albar

Figure 1: Differential
display via 2-DE: classical
workflow. Samples A
and B are resolved by 2-
DE in replicate gels and
silver stained. 2-D
images are analysed by
specific software and
differentially expressed
protein spots are
proteolytically digested
and analysed by MALDI-
TOF MS. The peptide
mass fingerprint (PMF) is
matched against genomic
or protein databases to
obtain candidate
proteins. Tandem mass
spectrometry (MS/MS)
and peptide sequencing
by analysis of
fragmentation spectra
can assist in identifying
peptides when ambiguity
remains after MALDI-
TOF analysis

fluorescent detection is gaining in Computer programs are continuously


popularity because of its sensitivity and being developed and improved — eg
Deep into visualisation wider linear dynamic range.15 After Progenesis (Nonlinear Dynamics), Image
methods and image
protein separation and staining, further Master 2D Platinum and Melanie
analysis
computer-based analysis is needed to Software (Amersham Biosciences) or
detect differentially expressed proteins. PDQuest (Bio-Rad) — but image

2 2 2 & HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004


Differential proteomics

analysis remains a time-consuming range, with neutralacidic isoelectric


process. points. Basic or very basic proteins (above
Protein identification is based on pH 9.5) are rather difficult to focus. 2-D
matching peptide experimental masses of gels also rarely display hydrophobic
proteolytically digested proteins (PMF) proteins, and only highly abundant
versus the theoretical masses obtained in proteins from total cell lysates are
the in silico digestion of all proteins in a currently visualised. Low abundance
specific database. The result is a list of proteins of physiological relevance, such
candidate proteins with different as regulators or signalling proteins, are
Use of 2-DE silver-
stained gels in different confidence levels. Despite its limitations, difficult to detect (for review, see ref. 33).
biological systems for years this proteomic strategy has Due to these drawbacks, a further protein
proved to be very useful in the fractionation step is needed prior to IEF
identification of differentially expressed to reduce complexity. This pre-
proteins in all areas of biological research. fractionation can be achieved using
In cancer research, it was implemented protocols based on differential solubility
for the detection of tumour-associated or by established procedures such as liquid
proteins in liver carcinoma,16 lung chromatography or free flow
adenocarcinoma,17 fibrosarcoma,18 electrophoresis. Organelle enrichment or
lymphoma,19 breast cancer20 and prostate membrane fraction preparations also
cancer.21 Several putative tumour markers reduce complexity and allow assessment
have been reported, although validation of protein location in the cell. The use of
for clinical purposes — one step further narrow, overlapping pH gradient strips
on — was not always fully accomplished. also increases the amount of protein
This technology has also been applied to loaded, detection of low abundance
the study of such organ-specific human facilitating the proteins.6
pathologies as thrombosis22 or heart
failure, sometimes studied in animal Fluorescent two-dimensional
models,23,24 and to address a variety of difference gel electrophoresis:
new biological challenges such as the cell 2-D DIGE
biology of symbiotic, opportunistic or Classical experimental design:
pathogenic bacteria25–29 and fungi,30 or Cy3 versus Cy5
virus–host cell interactions.31 As mentioned above, an important
Toxicological studies and drug-induced shortcoming of classical 2-DE approaches
differential protein expression are also is the intrinsic gel to gel variation that
among the important application areas.32 requires several replicate gels of each
This is just a short description of the work sample that are not directly overlapped.
published using so-called classical This can be circumvented using
expression proteomics. multiplexing methods such as fluorescent
2-DE workflow Despite the potential and resolution of two-dimensional difference in-gel
drawbacks 2-DE, it remains a labour-intensive electrophoresis (2-D DIGE),7 which
technique that requires qualified personal substantially reduces variability by
to obtain reproducible results. To displaying two or more complex protein
2-DE DIGE overcomes overcome gel to gel or intrinsic biological mixtures labelled with different
gel-to-gel variation and sample variations, it is considered that, for fluorescent dyes in a single 2D gel.
improves quantitation this type of expression proteomics study, Fluorescent labelling also renders 2-D
at least nine different gels are required for DIGE much more quantitative than
each cell state (three different gels of three colorimetric methods. It has a linear
different samples of the same biological dynamic range of four or five orders of
state). In any case, total proteome magnitude, by contrast with the
coverage by 2-DE is experimentally approximately one- or two-order range of
limited to proteins with molecular colloidal Coomassie and silver stains.34 As
weights in the 10120 kilodalton (kD) regards sensitivity, 1 ng of standard

& HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004 2 2 3


Monteoliva and Albar

protein is detected with Cy3/Cy5 homogenates, control versus treated)


fluorescent labelling.35 determined the quantitative variation of
2-D DIGE (marketed by Amersham the process. In this study, inter-animal
Biosciences) is based on the use of the response variability was about nine times
different, spectrally resolvables N- that contributed by the 2-D DIGE
hydroxysuccinimide derivative process.34
fluorescent dyes Cy3 and Cy5 (for a Several reports have been published in
review, see ref. 36). These dyes label the different fields using the two-dye 2-D
-amine groups of protein lysines DIGE technology. Differential gel
specifically and covalently to form an electrophoresis combined with other
amide. Control and treated protein techniques has thus been used to analyse
2-DE DIGE minimal mixtures are labelled independently with insect resistance to Bacillus thuringiensis
labelling features and Cy3 or Cy5 derivatives. The dyes are (Bt) Cry toxins. Changes were examined
workflow designed to have the same molecular in gut proteins from the larvae of an
weight and charge to ensure that proteins Indian meal moth (IMM; Plodia
common to both samples have the same interpunctella) colony showing resistance to
relative 2-DE mobility, regardless of the Bt. This study revealed a number of
dye used to tag them. Samples are changes in the levels of specific mid-gut
minimally labelled (only about 3 per cent proteins that indicate increased
of the total amount of each protein is glutathione utilisation, elevation in
tagged), then mixed and resolved in a oxidative metabolism and differential
single 2-D gel. The protein spot patterns maintenance of energy balance within the
are visualised by alternately illuminating mid-gut epithelial cells of the resistant
the gel with the excitation wavelengths larva model.37
for each of the two fluorescent dyes. The Borner et al.38 reported proteomic (2-
2-D images can be analysed by specific D DIGE-based) and genomic analysis of
software, such as DeCyder (Amersham glycosylphosphatidylinositol-anchored
Biosciences), to detect differentially proteins (GAP) extracted with
expressed protein spots. The nature of the phospholipase C (PLC) from Arabidopsis.
minimal labelling method results in The Pi (phosphatidylinositol-specific)-
populations of labelled and unlabelled PLC-treated and control aqueous phases
species for each protein. At the low were labelled with different fluorescent
molecular weight ranges, some positional Cy dyes. This revealed 30 proteins
discrepancy (shifts) between the low levels specifically enriched in the Pi-PLC-
of the labelled protein and the bulk of treated fraction and some background
unlabelled protein has been observed. In proteins present in both fractions.
order to maximise the amount of protein 2-D DIGE has also been applied to
available for MS, the total protein should biomedical studies. In a study of renal
be visualised using a post-staining method physiology, it was used to identify inner
(SYPRO ruby or silver staining). Selected medullary collecting duct proteins with
spots are then robotically excised from the different expression in cortical or outer
Use of two fluorescent
gel (a preparative scale gel, if necessary) cells.39 In addition, proteomic analysis of
dyes in 2-DE DIGE
applications and subjected to MS for identification. long-term vasopressin action in the inner
This 2-D DIGE workflow is outlined in medullary collecting duct of the
Figure 2A. Brattleboro rat showed 43 proteins that
The reproducibility and sensitivity of differ in abundance or in mobility; these
2-D DIGE were initially established by results were confirmed by semi-
Unlu et al.7 using protein extracts from quantitative immunoblotting and
two different Drosophila embryo samples. immunohistochemical approaches.40
Validation of 2-D DIGE as a tool for 2-D DIGE has also been used in the
toxicological applications (paracetamol search for cancer-specific protein markers
hepatotoxicity in mouse liver — eg to quantify differences in protein

2 2 4 & HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004


Differential proteomics

Figure 2: Schematic overview of 2-D fluorescent difference in-gel electrophoresis (2-D


DIGE) technology. (A) Two fluorescent dyes design. Samples A and B are fluorescently labelled
with either Cy3 or Cy5. The labelled samples are mixed and resolved in a single 2-D gel. The
protein spot patterns are visualised by alternately illuminating the gel with Cy3 and Cy5
excitation wavelengths. The 2-D images are analysed by specific software. Differentially
expressed protein spots, visualised with SYPRO ruby or silver post-staining, can be identified
by mass spectrometry. (B) Three fluorescent dyes design with pooled internal standard.
Samples A and B are fluorescently labelled with either Cy3 or Cy5, and a pooled internal
standard (with equal amounts of all the samples in the experiment) is labelled with Cy2.
Samples are mixed and resolved in a single 2-DE gel. Protein spot patterns are visualised by
alternately illuminating the gel at the excitation wavelengths of each dye. 2-D images are
analysed by specific software and the internal standard is used for normalisation. Statistical
analysis is performed and differentially expressed protein spots can be identified by MS

expression between laser capture infiltrating ductal breast carcinoma


microdissection-procured oesophageal demonstrated that 2-D DIGE portraits
carcinoma cells and normal epithelial might be a reflection of histological and
cells.41 A preliminary study of human pathological status.42

& HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004 2 2 5


Monteoliva and Albar

Among neuroscience-related together with randomised criss-cross


applications, identification of proteins experimental design in which control and
with age-related expression in cat primary treated samples are each tagged with Cy3
visual cortex has been reported. Protein or Cy5, provides statistical confidence in
extracts from the visual cortical area of 17 the detection and quantitation of subtle
adult cats and 30-day-old kittens were variations in protein expression.48
compared.43 Analysis of the brain This three-dye internal standard
proteome of genetically altered mice technique, outlined in Figure 2B, has
revealed alterations of stress-related been applied in a wide range of biological
proteins.44 areas. Liver toxicity in model animals has
Only three protein changes were been analysed in two studies: paracetamol
detected in the mitochondrial proteome toxicity49 and hydrazine toxicity as a
from mouse heart when double knockout model of multivariate data analysis.50
mice for two isoforms of creatine kinase Using a genetic neurokinin 1 receptor
and C57BL/6 mouse heart mitochondria knockout mouse model system, eight
were compared.45 In another study, six differentially expressed proteins were
protein changes were implicated in identified in cerebral cortex tissues.51 The
ischaemia reperfusion injury when 2-D three-dye method has also been applied in
DIGE was used to compare normal, cancer research, using a breast cancer cell
ischaemic and ischaemia-reperfused rat model system, to identify differentially
hearts.46 expressed proteins in a growth factor
2-D DIGE has also been applied to the stimulation time course.35 It has also been
study of D. melanogaster immune used to identify 52 unique proteins with
responses. Comparative analysis of the altered abundance in the proteome of
haemolymph proteome of 2,000 third- human colorectal tumour cells relative to
instar Drosophila larvae showed ten the adjacent normal mucosa of six
different proteins that appear very soon different patients.52
after an immune challenge with In microbiology, Gade et al.53
lipopolysaccharides.47 evaluated the use of 2-D DIGE, including
an internal standard, to detect and
Internal standard: Cy3 versus Cy5 quantify proteins specific for glucose or
versus Cy2 N-acetylglucosamine metabolism in the
A novel 2-D DIGE experimental design marine bacterium Pirellula sp.; 24 proteins
The international includes the use of a special ‘internal differing in abundance were identified
standard and standard’ created by ‘pooling aliquots’ of with high statistical confidence.
appropriate all samples in the experiment labelled
experimental design
allow normalisation and
with a third spectrally resolvable dye, Saturated labelling approach
statistical analysis in Cy2. This pooled internal standard is run Although 2-D DIGE enables increased
2-DE DIGE experiments in the same gel as control and treated confidence in the detection of protein
samples previously labelled with Cy3 and differences, only 35 per cent of a given
Cy5. The Cy2-labelled internal standard protein is tagged by the minimal labelling
is composed of equal amounts of each method. To increase sensitivity,
control and treated sample, and is Amersham Pharmacia developed a new set
Application of 2-DE included in all gels to normalise protein of DIGE Cy3 and Cy5 fluorescent dyes.
DIGE using three abundance measurements across multiple Protein cysteine residues are fully labelled
fluorescent dyes gel experiments. The use of this internal with these new maleimide cyanine dyes,
standard reduces gel to gel variation over enhancing sensitivity for low abundance
conventional 2-D DIGE and facilitates samples. In addition, protein spots are
the use of image analysis software excised directly from the saturation-
(DeCyder) for automated and accurate labelled gel, eliminating the need for post-
spot quantitation, gel to gel matching and stained preparative gels.54 Nevertheless,
statistical analysis. This improvement, only two dyes are currently commercially

2 2 6 & HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004


Differential proteomics

available, and the 2D-spot pattern is followed by MS), other methods have
modified. Expression differences in gained popularity, such as Mud-LC on-
motility-regulating proteins from murine line with ESI-MS/MS.5,60 In these
primitive haematopoietic cell populations approaches, complex mixtures of proteins
have been identified using this approach.55 are digested in solution. The resulting
peptide mixture is fractionated by one or
Other gel-based approaches several steps of capillary chromatography
Few reports have been published and analysed in a data-dependent manner
describing the use of alternative by MS/MS. These techniques share the
Less frequently used multiplexing methods for differential limitations of 2-DE for a dynamic range
multiplexing methods expression studies. In 1983, Goldman et of analysis (usually 103 –105 ) and
al.56 described a method that made use of identification of low concentration
metabolic labelling of proteins using proteins is achieved through pre-
radioactive isotope-labelled amino acids, fractionation techniques in a similar
2-DE and recording on colour negative manner to pre-fractionation methods used
film by radiographic exposure. A sub- in 2-DE gel analysis. Although classically
proteome differential display method that one of the limitations of LC/MS-based
uses radiolabelled proteins from one methods is the difficulty in performing
source and silver-stained proteins from a differential display analysis, several reports
second source, mixed in a gel in a 1:100 have recently appeared showing the
ratio, allowed precise discrimination feasibility of relative peptide
between members of each sub-proteome quantification with these strategies.61–64
(chromatographic fractions) using Alternative or complementary MS-
commonly available software.57 based approaches have been developed for
Combination of radiolabelling and differential protein expression
SYPRO ruby staining of the same gels measurements and are currently being
allows precise quantitation of the protein improved. They are based on the
amount as well as of the 35 S incorporated. differential labelling of perturbed and
This quantitative proteome profiling was non-perturbed protein extracts with
developed by Gerner and co-workers to different stable isotopes (12 C/ 13 C,14 N/
15
determine absolute values of cell protein N and 1 H/ 2 H). In this way, the same
amounts, as well as synthesis and turnover peptide from two different samples will
rates.58 The same method was recently show the same chemical behaviour, with
used to compare quiescent human T cells, a difference in mass detectable by MS
phytohaemagglutinin-stimulated T cells techniques. Peptide peak intensities can
and Jurkat cells, and to study human be used for relative quantification of these
umbilical vein endothelial cells treated for peptides. The workflow for this
6 hours with vascular endothelial growth methodology (outlined in Figure 3) is as
factor.59 follows: (1) differential isotopic labelling;
Because in vivo radioactive protein (2) digestion of combined protein samples
labelling is not always feasible, the to obtain peptide mixtures; (3)
fluorescence-based methods are more chromatographic fractionation of mixed
widely distributed. peptide samples; (4) analysis of the
separated peptides by MS/MS; and (5)
processing of the MS results to obtain
NON-GEL APPROACHES: relative protein abundance as well as
MS-BASED APPROACHES protein identification by database
Quantitative proteomics based searching.
on stable isotope tagging Several recent papers have reviewed in
Isotope tagging allows and MS detail the different chemical, metabolic
differential proteomics Because of the limitations that arise from and enzymatic labelling techniques used
based on LC-MS classical proteomics approaches (2-DE to date. The basis of these strategies, the

& HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004 2 2 7


Monteoliva and Albar

Figure 3: Quantitative proteomics based on stable isotope tagging and MS workflow. (A)
Stable isotope labelling with amino acids in cell culture (SILAC). Cell samples are cultured in
the presence of a stable isotopically labelled amino acid (aa). Cell samples are mixed, lysed and
digested. Subsequently, they are subjected to chromatographic fractionation and analysed by
tandem MS (MS/MS) to obtain relative protein abundance and protein identification by
database searching. (B) Isotope-coded affinity tag (ICAT). Protein samples are labelled with the
light or heavy version of the ICAT reagent, mixed and proteolytically digested. Labelled
peptides are isolated by avidin affinity chromatography and subjected to multidimensional liquid
chromatography ‘online’ with MS/MS. Peptides are identified by matching the MS/MS spectra
against databases. Protein abundance can be determined by integrating the peak areas of the
extracted ion chromatograms (XIC) for each isotope-coded peptide

2 2 8 & HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004


Differential proteomics

specific characteristics of each kind of phosphorylated peptide of the EGF


labelling, as well as advantages and receptor) and ‘control’ (non-
limitations have been discussed phosphorylated) forms were used as bait
extensively.65–69 Here, the focus will be in affinity pull-down experiments.
on two of the most used non-gel MS- SILAC differential labelling was also
based quantitative proteomic approaches used before membrane disorganisation to
and some of their recent applications will identify lipid raft proteins, to distinguish
be described. real raft components from non-raft
contamination. A set of 241 authentic
Metabolic isotopic labelling: SILAC lipid raft proteins was obtained, including
Addition of stable isotope labels to culture a large proportion of signalling molecules
media during cell growth yields isotope- (kinases and phosphatases or G
SILAC metabolic labelled proteins. This approach was first proteins).77 SILAC application to clinical
labelling has been described using Saccharomyces cerevisiae studies includes a recent prostate cancer
applied to identify
differentially expressed
grown in either 14 N minimal media or study.78 Sixty proteins were found with a
15 >3-fold increase in basal levels in the
proteins as well as N-enriched minimal media to
protein-interactions quantitate protein expression.70 highly metastatic cells, whereas levels of
(with proteins, peptides The use of a specific isotopic amino 22 other proteins were reduced. Western
or RNA) acid (eg deuterated L-leucine or 13 C- blot experiments confirmed the results
labelled arginine), rather than the entire obtained using the SILAC approach.
pool of amino acids described above, was
termed stable isotope labelling by amino Chemical labelling: ICAT
acids in cell culture (SILAC) by Mann Metabolic approaches based on the
and colleagues (Figure 3A).65,71 In this biological incorporation of isotope-
case, the mass difference between labelled labelled amino acids are not practical for
and unlabelled peptides is predictable, and any kind of biological sample, as only
MS/MS spectra interpretation becomes proteins extracted from growing cells can
easier. The technique was first applied to be used. Chemical incorporation of
the relative quantitation of changes in isotopic tags after protein extraction is
protein expression during the process of thus the best protein-tagging alternative
muscle cell differentiation, and was when metabolic labelling is not feasible.
described as a simple, inexpensive and The most commonly used method in
accurate method. Pratt et al.72,73 found the quantitative proteomics is based on the
use of isotope-labelled amino acids useful use of isotope-coded affinity tag (ICAT)
as an aid to protein identification in reagents developed by the Aebersold
peptide mass fingerprinting and for the group.9
study of protein turnover in yeast. The ICAT reagent-based analysis
SILAC has also been applied, in very workflow (Figure 3B) comprises the
elegant approaches, to the characterisation following steps: (1) protein extraction and
of protein–protein interactions in the reduction of control and treated samples;
ICAT chemical labelling study of the epidermal growth factor (2) chemical labelling of protein sulph-
is useful for all kinds of
(EGF) pathway74 and of proteins that hydryl groups with the ‘light’ and ‘heavy’
protein samples
participate in early stages of cell spreading versions of the ICAT reagent; (3) mixing
by interacting with focal adhesion of both samples and digestion by an
proteins.75 RNA and RNA-binding endoprotease; (4) isolation of labelled
protein involvement in cell spreading peptides; (5) peptide fractionation usually
through the spreading initiation centre using Mud-LC — strong-cation exchange
(SIC), a previously undescribed structure, (SCX) liquid chromatography followed by
were identified. Peptideprotein reversed-phase (RP) microcapillary liquid
interactions in the EGF pathway were chromatography; (6) MS/MS analysis with
also screened using a SILAC approach.76 an ESI-tandem mass spectrometer; (7)
Synthetic peptides in ‘active’ (tyrosine- automated database searching to identify

& HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004 2 2 9


Monteoliva and Albar

peptide sequences (and, thus, the proteins Myc oncoprotein expression in human B
from which they are derived); and (8) lymphocytes.81
determination of relative protein Two other very informative studies of
abundance from the MS data. The peak macromolecular complexes using the
areas of the extracted ion chromatograms ICAT reagent-based technology are the
(representing total ion currents for each study of Ste12 protein complexes from
peptide eluted from the column at a given yeast cells in different states82 and the
time) for each isotope-coded peptide are dynamic changes in transcription factor
used to determine relative peptide (and, complexes during erythroid
hence, protein) abundance. differentiation.83 In the former study,
Strategies in which peptides eluted (d0/d8)-ICAT was useful not only to
from the chromatography column are determine relative abundance changes in
spotted directly onto a MALDI target the composition of isolated complexes,
have also been used (see refs. 65, 66 and but also to distinguish specific complex
68 for detailed reviews). components from co-purified proteins.
First and second The first ICAT reagent, reported by Based on ICAT strategy, Brand et al.83
generation ICAT Aebersold and colleagues, was composed reported an interesting model of
reagent features of three parts: a reactive group specific for activation and repression of -globin gene
thiol groups, a linker and a biotin moiety expression during erythroid
for affinity chromatography purification differentiation.
using immobilised avidin. The linker has Cleavable (C 12 /C 13 ) ICAT has shown
either eight deuterium atoms in the heavy its potential in other biological
(d8) form or is undeuterated in the light applications, such as the study of the
(d0) form.9 Use (or not) of deuterium redox state of proteins84 and identification
causes differential chromatographic of metalloproteinase substrates in breast
elution profiles of the heavy and light carcinoma cells.85
forms of a given differentially labelled ICAT reagent-based quantitative
peptide, yielding inaccurate proteomics analysis for differential
measurements of abundance. The expression studies of total proteomes are
relatively large size of the biotin tag makes also reported. The study of whole
interpretation of the MS/MS spectra proteome changes in the opportunistic
difficult; a new generation of ICAT bacterial pathogen, Pseudomonas aeruginosa,
reagents was thus developed with nine cultured under conditions that induce
13
C or 12 C atoms at the linker moiety and expression of virulence factors, identified
a cleavable spacer to allow biotin removal several conserved Gram-negative proteins
(Applied Biosystems). involved in that process. The comparative
Several applications have been reported ICAT analyses of membrane versus whole
using the ICAT technology. The use of cell proteins allowed the detection of
cleavable ICAT led to development of a protein changes in subcellular
method to determine the subcellular compartmentalisation.86 ICAT
location of membrane proteins through a technology was applied to the study of
series of pairwise comparisons of gradient changes in the global proteome of yeast as
fractions. This method allows assignment a model eukaryotic system in two studies:
of proteins to a specific compartment in protein changes as a consequence of salt
Arabidopsis thaliana without the need to stress87 and the comparison with an upf1
obtain pure organelles.79 Shiio and co- mutant.88 Eukaryotic samples of higher
workers, besides using (d0/d8) ICAT complexity were analysed using ICAT
ICAT applications in
subcellular protein
reagents to compare the global protein reagent and Mud-LC. Examples include
location, expression pattern in rat myc-null cells the analysis of protein expression changes
macromolecular versus myc-plus cells,80 developed a induced in murine MC3T3 osteoblast
complexes and redox method to identify and quantify cells,89 mouse neurones (in the analysis of
state of proteins chromatin-associated proteins induced by DNA damage-induced neuronal death),90

2 3 0 & HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004


Differential proteomics

cystic fibrosis,91 androgen-stimulated and anionic, hydrophobic, hydrophilic, ion


unstimulated LNcap prostate cancer metal chelating). Each surface is designed
cells92 and intestinal epithelial cells in to retain proteins according to a general
response to an enteropathogenic or specific physicochemical property of
Escherichia coli strain.93 the protein, yielding an on-surface
Taken together, these studies show chromatographic protein separation. The
that, despite the enormous potential of second step is MS spectra acquisition —
ICAT studies of this technology developed for the after washing, the immobilised proteins
complex proteomes:
simultaneous analysis of complex are co-crystallised with a matrix on a
only a small percentage
of the total proteome is proteomes, we are far from being able to target surface and MS spectra are acquired
identified generate a picture of a whole proteome. by a specific mass analyser, eg a SELDI-
In yeast, for example, between 56087 and TOF mass spectrometer. Low-resolution
70088 proteins have been identified, protein patterns or retentate maps of the
which represents about 10 per cent of the proteins bound to each chromatographic
yeast genome; 2,501 were found (about 9 surface are thus generated. Unlike LC-
per cent) in murine osteoblast cells89 and MS, which is based on elution, SELDI-
over 2,000 in human polarised intestinal TOF MS combines retention with MS. In
epithelial monolayers.93 An important most instances, it does not allow direct
disadvantage of the ICAT method is that identification of proteins that may be
proteins lacking cysteines are not potential disease biomarkers. Finally, in
detected. data analysis and interpretation/
ICAT labelling A new ICAT variant based on the use evaluation, peak comparisons are made
combined with 2-DE of ICAT reagent to tag samples from two using multifactorial bioinformatic
cell types, combination of the two software (tree classifiers, neural networks,
samples and separation by 2-DE in a cluster analysis, test statistics) (Figure 4).
single gel, protein detection by MS- The technique is best for relatively small
compatible stains and identification and (,20 kD) proteins and protein fragments.
quantification by MS has been reported. This method constitutes a rapid,
The abundance ratio of the proteins in the reproducible analytical tool that enables
spot is determined by the signal intensity the comparative analysis of protein
ratio for the two isotopic forms of the expression profiles in the low fmol range,
label peptide.94 A similar approach although it has the same dynamic range
requiring metabolic labelling has been limitations as other MS technologies.
described for Plasmodium falciparum,95 rat Proteins from complex biological
hepatocytes,96 the multicellular organisms specimens such as serum, plasma,
Caenorhabditis elegans and D. melanogaster 97 intestinal fluid, urine, cell lysates and cell
and for the quantitation of protein secretion products have been profiled
expression and site-specific using SELDI-TOF MS. Serum proteomic
phosphorylation in yeast.98 profiling using SELDI-TOF MS is a
promising new approach for cancer
SELDI-TOF: protein profiling diagnostics.99 The central hypothesis is as
SELDI-TOF MS enables analysis of follows: protein or protein fragments
SELDI-TOF combines
chromatographic complex protein mixtures separated by produced by cancer cells or their
differential protein on-chip retentate chromatography.11 The microenvironment may eventually enter
retention and TOF MS analytical procedure involves a few the general circulation. The different
common steps, beginning with protein/fragment patterns could then be
chromatographic separation. The analysed by MS and used for diagnostic
biological fluid of interest is pre- purposes, in combination with a
fractionated or loaded directly onto bioinformatic algorithm.100 The cancer
several chemically treated supports — types that have been examined include
SELDI and detection of protein chip arrays — with specific ovarian,101 prostate,102 breast,103
cancer biomarkers chromatographic features (cationic, bladder104 and renal.105 Apparent

& HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004 2 3 1


Monteoliva and Albar

Figure 4: Differential
protein expression
profiling by SELDI-TOF
MS. Pre-fractionated
complex mixtures are
chromatographically
separated using protein
chips arrays; the chips
consist of eight or 16
spots of a specific
chromatographic surface
(hydrophobic, cation
exchange, anion
exchange, metal affinity).
MS spectra of bound
proteins are obtained by
SELDI-TOF MS. Output
data from MS are
displayed as trace, gel
and map views (top to
bottom). Finally,
univariate and/or
multivariate data analysis
is performed with the
appropriate software to
determine differentially
expressed proteins

Are SELDI-TOF
successes of this technology have recently The SELDI-TOF technology is
patterns useful as
diagnostic or prognastic been extensively reviewed.100,106–108 marketed by Ciphergen Biosystems.
markers? Clinical trials are now underway and will More than 200 papers have already been
reveal whether these data can be published on the use of this method. In
reproduced and if the platform is general, it has been suggested that this
sufficiently robust for clinical use. The technique could show much higher
discriminatory peaks, if positively diagnostic sensitivity and specificity
identified, may represent molecules that (approaching 100 per cent) compared
could be measured with simpler and with classical cancer biomarkers.109
cheaper techniques for the purpose of These sensitivities/specificities are far
diagnosing cancer. superior to those obtained using classical

2 3 2 & HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004


Differential proteomics

techniques to detect cancer biomarkers. frequency to obtain an integrated view of


Some papers110,111 have questioned the cell physiology; genomic and proteomic
validity of the results obtained with this data have proven to be
technology and propose experiments to complementary.122,123
investigate these questions in detail The wide dynamic range of
before clinical use of the technique. intracellular or serum protein
Several statistical analyses have been concentration, the much more reduced
performed on the original ovarian cancer range detectable by bioanalytical
work. This analysis uncovered numerous techniques and the lack of protein
problems with study designs and sample amplification procedures are issues that
collection and handling.112,113 This calls proteomics strategies must address. To
into question the original claims of improve global proteome coverage, cell
SELDI patterns as diagnostic or fractionation is a mandatory step in
prognostic markers. Nevertheless, Zhang overcoming these important drawbacks.
et al.114 have recently reported a Traditional approaches for differential
coordinated multicentre study, using 503 display protein analysis based on 2-DE
serum samples from ovarian cancer and colourimetric staining are still very
patients at stages I/II and III/IV of the useful, and have produced many
disease and 142 healthy women, contributions even considering their
specifically designed to alleviate the intrinsic sensitivity- and reproducibility-
impact of these factors and to show the associated limitations. Nevertheless, the
validity of this technological platform as uniqueness of 2-DE for easy visualisation
well as the potential of the identified of protein isoforms, using two physical
biomarkers to improve the detection of parameters such as isoelectric point and
SELDI applications early-stage ovarian cancer.114 The molecular weight, renders this technology
other than cancer
method must clearly be thoroughly itself extremely informative.
validated before clinical implementation Quantitative differential proteomics has
is warranted. The most important required the use of alternative approaches
criticism of the use of this methodology to expand the analytical range offered by
is based on the fact that, since proteins classical 2-DE. Among them, gel- and
are not usually identified, it is difficult to non-gel-based methods such as 2-D
support the central hypothesis that these DIGE, ICAT reagent-based methods,
differential protein features come from SILAC and SELDI-TOF are notable. The
cancer cells or their microenvironment. introduction of fluorescent 2-D DIGE
Without identification, it cannot be with the use of pooled internal standards,
known whether a protein peak is related together with sample pre-fractionation
to a disease state or not. approaches, has greatly improved the
The potential of this method has been efficiency of 2-DE methods as a truly
extended to conditions such as quantitative differential display
Alzheimer’s disease, Creutzfeldt-Jakob technology. Stable isotope tagging
disease and renal allograft rejection.115,116 strategies and MS/MS have also allowed
It has also been used in targeted studies to quantitative differential proteomics by
characterise protein–protein and protein– focusing on peptide comparison as a
DNA interactions,117,118 as well as to ‘reflection/image’ of the proteins from
characterise phosphorylated and which they are derived and, hence, their
glycosylated proteins119,120 or proteomes. Gel- and non-gel-based
Gel- and non-gel-based transcription factors.121 approaches provide closely related but
approaches are
complementary in
distinct information about proteins,
differential proteomics CONCLUDING REMARKS suggesting that they are complementary,
In studying a biological system, both or at least supplementary, methods.
genomics and proteomics approaches are The recent introduction of SELDI-
being implemented with increasing TOF MS has allowed the acquisition of

& HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004 2 3 3


Monteoliva and Albar

protein patterns from complex protein spectrometry and proteomics’, Curr. Opin.
mixtures, mainly body fluids such as Chem. Biol., Vol. 4, pp. 489–494.
plasma or serum associated with many 11. Merchant, M. and Weinberger, S. R. (2000),
human diseases. Before this technique can ‘Recent advancements in surface-enhanced
laser desorption/ionization-time of flight-
be applied to clinical use, it must be mass spectrometry’, Electrophoresis, Vol. 21,
validated extensively, but it may, pp. 1164–1177.
nonetheless, be the most promising 12. Molloy, M. P. and Witzmann, F. A. (2002),
approach for biomarker discovery in ‘Proteomics: technologies and applications’,
clinical proteomics. Brief. Funct. Genom. Proteom., Vol. 1,
pp. 23–39.
13. Gorg, A., Obermaier, C., Boguth, G. et al.
ACKNOWLEDGMENTS (2000), ‘The current state of two-dimensional
We gratefully acknowledge Dr Alberto Paradela for electrophoresis with immobilized pH
critical reading of the manuscript and Cathy Mark gradients’, Electrophoresis, Vol. 21, pp.
for editorial assistance. 1037–1053.
14. Lilley, K. S., Razzaq, A. and Dupree, P.
(2002), ‘Two-dimensional gel
References electrophoresis: recent advances in sample
1. Anderson, N. G. and Anderson, N. L. preparation, detection and quantitation’,
(1996), ‘Twenty years of two-dimensional Curr. Opin. Chem. Biol., Vol. 6, pp. 46–50.
electrophoresis: past, present and future’, 15. Patton, W. F. (2002), ‘Detection
Electrophoresis, Vol. 17, pp. 443–453. technologies in proteome analysis’, J.
2. Pandey, A. and Mann, M. (2000), Chromatogr. B Analyt. Technol. Biomed. Life
‘Proteomics to study genes and genomes’, Sci., Vol. 771, pp. 3–31.
Nature, Vol. 405, pp. 837–846. 16. Jungblut, P. R., Zimny-Arndt, U., Zeindl-
3. Andersen, J. S. and Mann, M. (2000), Eberhart, E. et al. (1999), ‘Proteomics in
‘Functional genomics by mass spectrometry’, human disease: cancer, heart and infectious
FEBS Lett., Vol. 480, pp. 25–31. diseases’, Electrophoresis, Vol. 20, pp.
2100–2110.
4. Corthals, G. L., Gygi, S. P., Aebersold, R.
and Patterson, S. P. (1999), ‘Identification of 17. Chen, G., Gharib, T. G., Wang, H. et al.
proteins by mass spectrometry’, in Rabilloud (2003), ‘Protein profiles associated with
T. (Ed.), ‘Proteome Research: Two–Dimensional survival in lung adenocarcinoma’, Proc. Natl.
Gel Electrophoresis and Identification Methods’, Acad. Sci. USA, Vol. 100, pp. 13537–13542.
Springer Verlag, New York, NY, 18. Sinha, P., Hutter, G., Kottgen, E. et al.
pp. 197–231.
(1999), ‘Search for novel proteins involved in
5. Link, A. J., Eng, J., Schieltz, D. M. et al. the development of chemoresistance in
(1999), ‘Direct analysis of protein complexes colorectal cancer and fibrosarcoma cells in
using mass spectrometry’, Nat. Biotechnol., vitro using two-dimensional electrophoresis,
Vol. 17, pp. 676–682. mass spectrometry and microsequencing’,
Electrophoresis, Vol. 20, pp. 2961–2969.
6. López, J.A., Bernard, A. and Albar, J. P.
(2004), ‘Protein expression profiling analysis 19. Antonucci, F., Chilosi, M., Parolini, C. et al.
in hematopoietic stem cells: phenotypic (2003), ‘Two-dimensional molecular
characterization of mesenchymal stem cells’, profiling of mantle cell lymphoma’,
in Sanchez, J. C., Corthals, G. and Electrophoresis, Vol. 24, pp. 2376–2385.
Hochstrasser, D. F. (Eds), ‘Biomedical
20. Giometti, C. S., Williams, K. and Tollaksen,
Applications of Proteomics’, WILEY-VCH
S. L. (1997), ‘A two-dimensional
Verlag, Weinheim, Germany, pp. 155–171.
electrophoresis database of human breast
7. Unlu, M., Morgan, M. E. and Minden, J. S. epithelial cell proteins’, Electrophoresis, Vol.
(1997), ‘Difference gel electrophoresis: a 18, pp. 573–581.
single gel method for detecting changes in
21. Ahram, M., Best, C. J., Flaig, M. J. et al.
protein extracts’, Electrophoresis, Vol. 18, pp.
(2002), ‘Proteomic analysis of human prostate
2071–2077.
cancer’, Mol. Carcinogen., Vol. 33, pp. 9–15.
8. URL:
22. Gelfi, C., Vigano, A., Ripamonti, M. et al.
http://www.amershambiosciences.com.
(2004), ‘A proteomic analysis of changes in
9. Gygi, S. P., Rist, B., Gerber, S. A. et al. prothrombin and plasma proteins associated
(1999), ‘Quantitative analysis of complex with the G20210A mutation’, Proteomics, Vol.
protein mixtures using isotope-coded affinity 4, pp. 2151–2159.
tags’, Nat. Biotechnol., Vol. 17, pp. 994–999.
23. Sawicki, G. and Jugdutt, B. I. (2004),
10. Gygi, S. P. and Aebersold, R. (2000), ‘Mass ‘Detection of regional changes in protein

2 3 4 & HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004


Differential proteomics

levels in the in vivo canine model of acute 35. Gharbi, S., Gaffney, P., Yang, A. et al. (2002),
heart failure following ischemia-reperfusion ‘Evaluation of two-dimensional differential
injury: functional proteomics studies’, gel electrophoresis for proteomic expression
Proteomics, Vol. 4, pp. 2195–2202. analysis of a model breast cancer cell system’,
Mol. Cell. Proteom., Vol. 1, pp. 91–98.
24. Schwertz, H., Langin, T., Platsch, H. et al.
(2002), ‘Two-dimensional analysis of 36. Van den Bergh, G. and Arckens, L. (2004),
myocardial protein expression following ‘Fluorescent two-dimensional difference gel
myocardial ischemia and reperfusion in electrophoresis unveils the potential of gel-
rabbits’, Proteomics, Vol. 2, pp. 988–995. based proteomics’, Curr. Opin. Biotechnol.,
Vol. 15, pp. 38–43.
25. Djordjevic, M. A. (2004), ‘Sinorhizobium
meliloti metabolism in the root nodule: a 37. Candas, M., Loseva, O., Oppert, B. et al.
proteomic perspective’, Proteomics, Vol. 4, pp. (2003), ‘Insect resistance to Bacillus
1859–1872. thuringiensis: alterations in the indianmeal
moth larval gut proteome’, Mol. Cell.
26. Arevalo-Ferro, C., Hentzer, M., Reil, G. et Proteom., Vol. 2, pp. 19–28.
al. (2003), ‘Identification of quorum-sensing
regulated proteins in the opportunistic 38. Borner, G. H., Lilley, K. S., Stevens, T. J.
pathogen Pseudomonas aeruginosa by and Dupree, P. (2003), ‘Identification of
proteomics’, Environ. Microbiol., Vol. 5, pp. glycosylphosphatidylinositol-anchored
1350–1369. proteins in Arabidopsis. A proteomic and
genomic analysis’, Plant Physiol., Vol. 132,
27. Morales, G., Linares, J. F., Beloso, A. et al. pp. 568–577.
(2004), ‘The Pseudomonas putida Crc global
regulator controls the expression of genes 39. Hoffert, J. D., van Balkom, B. W., Chou,
from several chromosomal catabolic pathways C. L. and Knepper, M. A. (2004),
for aromatic compounds’, J. Bacteriol., Vol. ‘Application of difference gel electrophoresis
186, pp. 1337–1344. to the identification of inner medullary
collecting duct proteins’, Am. J. Physiol. Ren.
28. Cases, I., Lopez, J. A., Albar, J. P. and Physiol., Vol. 286, pp. F170–F179.
de Lorenzo, V. (2001), ‘Evidence of multiple
regulatory functions for the PtsN (IIANtr ) 40. van Balkom, B. W., Hoffert, J. D., Chou,
protein of Pseudomonas putida’, J. Bacteriol., C. L. and Knepper, M. A. (2004), ‘Proteomic
Vol. 183, pp. 10321037. analysis of long-term vasopressin action in the
inner medullary collecting duct of the
29. Agudo, D., Mendoza, M. T., Castanares, C. Brattleboro rat’, Am. J. Physiol. Ren. Physiol.,
et al. (2004), ‘A proteomic approach to study Vol. 286, pp. F216–F224.
Salmonella typhi periplasmic proteins altered
by a lack of the DsbA thiol:disulfide 41. Zhou, G., Li, H., DeCamp, D. et al. (2002),
isomerase’, Proteomics, Vol. 4, pp. 355–363. ‘2D differential in-gel electrophoresis for the
identification of esophageal scans cell cancer-
30. Pitarch, A., Sanchez, M., Nombela, C. and specific protein markers’, Mol. Cell. Proteom.,
Gil, C. (2002), ‘Sequential fractionation and Vol. 1, pp. 117–124.
two-dimensional gel analysis unravels the
complexity of the dimorphic fungus Candida 42. Somiari, R. I., Sullivan, A., Russell, S. et al.
albicans cell wall proteome’, Mol. Cell. (2003), ‘High-throughput proteomic analysis
Proteom., Vol. 1, pp. 967–982. of human infiltrating ductal carcinoma of the
breast’, Proteomics, Vol. 3, pp. 1863–1873.
31. Alfonso, P., Rivera, J., Hernaez, B. et al.
(2004), ‘Identification of cellular proteins 43. Van den Bergh, G., Clerens, S., Cnops, L.
modified in response to African swine fever et al. (2003), ‘Fluorescent two-dimensional
virus infection by proteomics’, Proteomics, difference gel electrophoresis and mass
Vol. 4, pp. 2037–2046. spectrometry identify age-related protein
expression differences for the primary visual
32. Cecconi, D., Astner, H., Donadelli, M. et al. cortex of kitten and adult cat’, J. Neurochem.,
(2003), ‘Proteomic analysis of pancreatic Vol. 85, pp. 193–205.
ductal carcinoma cells treated with 5-aza-29-
deoxycytidine’, Electrophoresis, Vol. 24, pp. 44. Skynner, H. A., Rosahl, T. W., Knowles,
4291–4303. M. R. et al. (2002), ‘Alterations of stress
related proteins in genetically altered mice
33. Corthals, G. L., Wasinger, V. C., revealed by two-dimensional differential in-
Hochstrasser, D. F. and Sanchez, J. C. (2000), gel electrophoresis analysis’, Proteomics, Vol.
‘The dynamic range of protein expression: a 2, pp. 1018–1025.
challenge for proteomic research’,
Electrophoresis, Vol. 21, pp. 11041115. 45. Kernec, F., Unlu, M., Labeikovsky, W. et al.
(2001), ‘Changes in the mitochondrial
34. Tonge, R., Shaw, J., Middleton, B. et al. proteome from mouse hearts deficient in
(2001), ‘Validation and development of creatine kinase’, Physiol. Genomics, Vol. 6,
fluorescence two-dimensional differential gel pp. 117–128.
electrophoresis proteomics technology’,
Proteomics, Vol. 1, pp. 377–396. 46. Sakai, J., Ishikawa, H., Kojima, S. et al.

& HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004 2 3 5


Monteoliva and Albar

(2003), ‘Proteomic analysis of rat heart in ‘Sub-proteome differential display: single gel
ischemia and ischemia-reperfusion using comparison by 2D electrophoresis and mass
fluorescence two-dimensional difference gel spectrometry’, J. Mol. Biol., Vol. 318, pp.
electrophoresis’, Proteomics, Vol. 3, pp. 21–31.
1318–1324.
58. Gerner, C., Vejda, S., Gelbmann, D. et al.
47. Vierstraete, E., Verleyen, P., Baggerman, G. (2002), ‘Concomitant determination of
et al. (2004), ‘A proteomic approach for the absolute values of cellular protein amounts,
analysis of instantly released wound and synthesis rates, and turnover rates by
immune proteins in Drosophila melanogaster quantitative proteome profiling’, Mol. Cell.
hemolymph’, Proc. Natl. Acad. Sci. USA, Vol. Proteom., Vol. 1, pp. 528–537.
101, pp. 470–475.
59. Traxler, E., Bayer, E., Stockl, J. et al. (2004),
48. Alban, A., David, S. O., Bjorkesten, L. et al. ‘Towards a standardized human proteome
(2003), ‘A novel experimental design for database: quantitative proteome profiling of
comparative two-dimensional gel analysis: living cells’, Proteomics, Vol. 4, pp.
two-dimensional difference gel 1314–1323.
electrophoresis incorporating a pooled
internal standard’, Proteomics, Vol. 3, 60. Washburn, M. P., Wolters, D. and Yates III,
pp. 36–44. J. R. (2001), ‘Large-scale analysis of the yeast
proteome by multidimensional protein
49. Ruepp, S. U., Tonge, R. P., Shaw, J. et al. identification technology’, Nat. Biotechnol.,
(2002), ‘Genomics and proteomics analysis of Vol. 19, pp. 242–247.
acetaminophen toxicity in mouse liver’,
Toxicol. Sci., Vol. 65, pp. 135–150. 61. Bondarenko, P. V., Chelius, D. and Shaler,
T. A. (2002), ‘Identification and relative
50. Kleno, T. G., Leonardsen, L. R., Kjeldal, H. quantitation of protein mixtures by enzymatic
O. et al. (2004), ‘Mechanisms of hydrazine digestion followed by capillary reversed-phase
toxicity in rat liver investigated by proteomics liquid chromatography-tandem mass
and multivariate data analysis’, Proteomics, Vol. spectrometry’, Anal. Chem., Vol. 74, pp.
4, pp. 868–880. 4741–4749.
51. Knowles, M. R., Cervino, S., Skynner, H. A. 62. Wang, W., Zhou, H., Lin, H. et al. (2003),
et al. (2003), ‘Multiplex proteomic analysis by ‘Quantification of proteins and metabolites
two-dimensional differential in-gel by mass spectrometry without isotopic
electrophoresis’, Proteomics, Vol. 3, pp. labeling or spiked standards’, Anal. Chem.,
1162–1171. Vol. 75, pp. 4818–4826.
52. Friedman, D. B., Hill, S., Keller, J. W. et al. 63. Stewart, I. I., Zhao, L., Le Bihan, T. et al.
(2004), ‘Proteome analysis of human colon (2004), ‘The reproducible acquisition of
cancer by two-dimensional difference gel comparative liquid chromatography/tandem
electrophoresis and mass spectrometry’, mass spectrometry data from complex
Proteomics, Vol. 4, pp. 793–811. biological samples’, Rapid Comm. Mass
53. Gade, D., Thiermann, J., Markowsky, D. and Spectrom., Vol. 18, pp. 1697–1710.
Rabus, R. (2003), ‘Evaluation of two- 64. Liu, H., Sadygov, R. G. and Yates III, J. R.
dimensional difference gel electrophoresis for (2004), ‘A model for random sampling and
protein profiling. Soluble proteins of the estimation of relative protein abundance in
marine bacterium Pirellula sp. strain 1’, J. Mol. shotgun proteomics’, Anal. Chem., Vol. 76,
Microbiol. Biotechnol., Vol. 5, pp. 240–251. pp. 4193–4201.
54. Shaw, J., Rowlinson, R., Nickson, J. et al. 65. Ong, S. E., Foster, L. J. and Mann, M.
(2003), ‘Evaluation of saturation labelling (2003), ‘Mass spectrometric-based approaches
two-dimensional difference gel in quantitative proteomics’, Methods, Vol. 29,
electrophoresis fluorescent dyes’, Proteomics, pp. 124–130.
Vol. 3, pp. 1181–1195.
66. Flory, M. R., Griffin, T. J., Martin, D. and
55. Evans, C. A., Tonge, R., Blinco, D. et al. Aebersold, R. (2002), ‘Advances in
(2004), ‘Comparative proteomics of primitive quantitative proteomics using stable isotope
hematopoietic cell populations reveals tags’, Trends Biotechnol., Vol. 20, pp.
differences in expression of proteins S23–S29.
regulating motility’, Blood, Vol. 103, pp.
3751–3759. 67. Lill, J. (2003), ‘Proteomic tools for
quantitation by mass spectrometry’, Mass
56. Goldman, R. C., Trus, B. L. and Leive, L. Spectrom. Rev., Vol. 22, pp. 182–194.
(1983), ‘Quantitative double-label
radiography of two-dimensional protein gels 68. Goshe, M. B. and Smith, R. D. (2003),
using colour negative film and computer ‘Stable isotope-coded proteomic mass
analysis’, Eur. J. Biochem., Vol. 131, pp. spectrometry’, Curr. Opin. Biotechnol., Vol.
473–480. 14, pp. 101–109.
57. Spandidos, A. Ò. and Rabbitts, T. H. (2002), 69. Aebersold, R. and Mann, M. (2003), ‘Mass

2 3 6 & HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004


Differential proteomics

spectrometry-based proteomics’, Nature, Vol. 82. Ranish, J. A., Yi, E. C., Leslie, D. M. et al.
422, pp. 198–207. (2003), ‘The study of macromolecular
complexes by quantitative proteomics’, Nat.
70. Washburn, M. P., Ulaszek, R., Deciu, C. Genet., Vol. 33, pp. 349355.
et al. (2002), ‘Analysis of quantitative
proteomic data generated via 83. Brand, M., Ranish, J. A., Kummer, N. T.
multidimensional protein identification et al. (2004), ‘Dynamic changes in
technology’, Anal. Chem., Vol. 74, pp. transcription factor complexes during
1650–1657. erythroid differentiation revealed by
quantitative proteomics’, Nat. Struct. Mol.
71. Ong, S. E., Blagoev, B., Kratchmarova, I. et Biol., Vol. 11, pp. 73–80.
al. (2002), ‘Stable isotope labeling by amino
acids in cell culture, SILAC, as a simple and 84. Sethuraman, M., McComb, M. E., Heibeck,
accurate approach to expression proteomics’, T. et al. (2004), ‘Isotope-coded affinity tag
Mol. Cell. Proteomics, Vol. 1, pp. 376–386. approach to identify and quantify oxidant-
sensitive protein thiols’, Mol. Cell. Proteomics,
72. Pratt, J. M., Robertson, D. H., Gaskell, S. J., Vol. 3, pp. 273–278.
et al. (2002), ‘Stable isotope labelling in vivo as
an aid to protein identification in peptide 85. Tam, E. M., Morrison, C. J., Wu, Y. I. et al.
mass fingerprinting’, Proteomics, Vol. 2, pp. (2004), ‘Membrane protease proteomics:
157–163. isotope-coded affinity tag MS identification
of undescribed MT1-matrix
73. Pratt, J. M., Petty, J., Riba-Garcia, I. et al. metalloproteinase substrates’, Proc. Natl. Acad.
(2002), ‘Dynamics of protein turnover, a Sci. USA, Vol. 101, pp. 6917–6922.
missing dimension in proteomics’, Mol. Cell.
Proteomics, Vol. 1, pp. 579–591. 86. Guina, T., Wu, M., Miller, S. I. et al. (2003),
‘Proteomic analysis of Pseudomonas aeruginosa
74. Blagoev, B., Kratchmarova, I., Ong, S. E. grown under magnesium limitation’, J. Am.
et al. (2003), ‘A proteomics strategy to Soc. Mass Spectrom., Vol. 14, pp. 742–751.
elucidate functional protein-protein
interactions applied to EGF signaling’, Nat. 87. Li, J., Steen, H. and Gygi, S. P. (2003),
Biotechnol., Vol. 21, pp. 315–318. ‘Protein profiling with cleavable isotope-
coded affinity tag (cICAT) reagents: the yeast
75. De Hoog, C. L., Foster, L. J. and Mann, M. salinity stress response’, Mol. Cell. Proteomics,
(2004), ‘RNA and RNA binding proteins Vol. 2, pp. 1198–1204.
participate in early stages of cell spreading
through spreading initiation centers’, Cell, 88. Parker, K. C., Patterson, D., Williamson, B.
Vol. 117, pp. 649–662. et al. (2004), ‘Depth of proteome issues: a
yeast isotope-coded affinity tag reagent
76. Schulze, W. X. and Mann, M. (2004), ‘A study’, Mol. Cell. Proteomics, Vol. 3, pp.
novel proteomic screen for peptide–protein 625–659.
interactions’, J. Biol. Chem., Vol. 279, pp.
10756–10764. 89. Conrads, K. A., Yu, L. R., Lucas, D. A. et al.
(2004), ‘Quantitative proteomic analysis of
77. Foster, L. J., De Hoog, C. L. and Mann, M. inorganic phosphate-induced murine
(2003), ‘Unbiased quantitative proteomics of MC3T3-E1 osteoblast cells’, Electrophoresis,
lipid rafts reveals high specificity for signaling Vol. 25, pp. 1342–1352.
factors’, Proc. Natl. Acad. Sci. USA, Vol. 100,
pp. 5813–5818. 90. Johnson, M. D., Yu, L. R., Conrads, T. P.
et al. (2004), ‘Proteome analysis of DNA
78. Everley, P. A., Krijgsveld, J., Zetter, B. R. damage-induced neuronal death using high
and Gygi, S. P. (2004), ‘Quantitative cancer throughput mass spectrometry’, J. Biol.
proteomics: stable isotope labeling with Chem., Vol. 279, pp. 26685–26697.
amino acids in cell culture (SILAC) as a tool
for prostate cancer research’, Mol. Cell. 91. Hansen, K. C., Schmitt-Ulms, G., Chalkley,
Proteomics, Vol. 3, pp. 729–735. R. J. et al. (2003), ‘Mass spectrometric
analysis of protein mixtures at low levels
79. Dunkley, T. P., Dupree, P., Watson, R. B. using cleavable 13 C-isotope-coded affinity tag
and Lilley, K. S. (2004), ‘The use of isotope- and multidimensional chromatography’, Mol.
coded affinity tags (ICAT) to study organelle Cell. Proteomics, Vol. 2, pp. 299–314.
proteomes in Arabidopsis thaliana’, Biochem.
Soc. Trans., Vol. 32, pp. 520–523. 92. Meehan, K. L. and Sadar, M. D. (2004),
‘Quantitative profiling of LNCaP prostate
80. Shiio, Y., Donohoe, S., Yi, E. C. et al. cancer cells using isotope-coded affinity tags
(2002), ‘Quantitative proteomic analysis of and mass spectrometry’, Proteomics., Vol. 4,
Myc oncoprotein function’, EMBO J., Vol. pp. 1116–1134.
21, pp. 5088–5096.
93. Hardwidge, P. R., Rodriguez-Escudero, I.,
81. Shiio, Y., Eisenman, R. N., Yi, E. C. et al. Goode, D. et al. (2004), ‘Proteomic analysis
(2003), ‘Quantitative proteomic analysis of of the intestinal epithelial cell response to
chromatin-associated factors’, J. Am. Soc. enteropathogenic Escherichia coli’, J. Biol.
Mass Spectrom., Vol. 14, pp. 696–703. Chem., Vol. 279, pp. 20127–20136.

& HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004 2 3 7


Monteoliva and Albar

94. Smolka, M., Zhou, H. and Aebersold, R. 106. Rai, A. J., Zhang, Z., Rosenzweig, J. et al.
(2002), ‘Quantitative protein profiling using (2002), ‘Proteomic approaches to tumor
two-dimensional gel electrophoresis, isotope- marker discovery’, Arch. Pathol. Lab. Med.,
coded affinity tag labeling, and mass Vol. 126, pp. 1518–1526.
spectrometry’, Mol. Cell. Proteomics, Vol. 1,
pp. 19–29. 107. Issaq, H. J., Conrads, T. P., Prieto, D. A.
et al. (2003), ‘SELDI-TOF MS for diagnostic
95. Nirmalan, N., Sims, P. F. and Hyde, J. E. proteomics’, Anal. Chem., Vol. 75, pp.
(2004), ‘Quantitative proteomics of the 148A–155A.
human malaria parasite Plasmodium falciparum
and its application to studies of development 108. Wulfkuhle, J. D., Paweletz, C. P., Steeg,
P. S. et al. (2003), ‘Proteomic approaches to
and inhibition’, Mol. Microbiol., Vol. 52, pp.
the diagnosis, treatment, and monitoring of
11871199.
cancer’, Adv. Exp. Med. Biol., Vol. 532, pp.
96. Jaleel, A. and Nair, K. S. (2004), 59–68.
‘Identification of multiple proteins whose
109. Powell, K. (2003), ‘Proteomics delivers on
synthetic rates are enhanced by high amino
promise of cancer biomarkers’, Nat. Med.,
acid levels in rat hepatocytes’, Am. J. Physiol.
Vol. 9, p. 980.
Endocrinol. Metab., Vol. 286, pp. E950–E957.
110. Diamandis, E. P. (2004), ‘Mass spectrometry
97. Krijgsveld, J., Ketting, R. F., Mahmoudi, T.
as a diagnostic and a cancer biomarker
et al. (2003), ‘Metabolic labeling of C. elegans
discovery tool: opportunities and potential
and D. melanogaster for quantitative
limitations’, Mol. Cell. Proteomics, Vol. 3, pp.
proteomics’, Nat. Biotechnol., Vol. 21, pp.
367–378.
927–931.
111. Diamandis, E. P. (2004), ‘Analysis of serum
98. Oda, Y., Huang, K., Cross, F. R. et al.
proteomic patterns for early cancer diagnosis:
(1999), ‘Accurate quantitation of protein
drawing attention to potential problems’, J.
expression and site-specific phosphorylation’,
Natl. Cancer Inst., Vol. 96, pp. 353–356.
Proc. Natl. Acad. Sci. USA, Vol. 96, pp.
6591–6596. 112. Baggerly, K. A., Morris, J. S. and Coombes,
K. R. (2004), ‘Reproducibility of SELDI-
99. Service, R. F. (2003), ‘Genetics and
TOF protein patterns in serum: comparing
medicine. Recruiting genes, proteins for a
datasets from different experiments’,
revolution in diagnostics’, Science, Vol. 300,
Bioinformatics, Vol. 20, pp. 777–785.
pp. 236–239.
113. Sorace, J. M. and Zhan, M. (2003), ‘A data
100. Petricoin, E. F., Zoon, K. C., Kohn, E. C.
review and re-assessment of ovarian cancer
et al. (2002), ‘Clinical proteomics: translating
serum proteomic profiling’, Bioinformatics,
benchside promise into bedside reality’, Nat.
Vol. 4, p. 24.
Rev. Drug Discov., Vol. 1, pp. 683–695.
114. Zhang, Z., Bast Jr, R. C., Yu, Y. et al.
101. Petricoin, E. F., Ardekani, A. M., Hitt, B. A.
(2004), ‘Three biomarkers identified from
et al. (2002), ‘Use of proteomic patterns in
serum proteomic analysis for the detection of
serum to identify ovarian cancer’, Lancet, Vol.
early stage ovarian cancer’, Cancer Res., Vol.
359, pp. 572–577.
64, pp. 58825890.
102. Grizzle, W. E., Adam, B. L., Bigbee, W. L.
115. Carrette, O., Demalte, I., Scherl, A. et al.
et al. (2003), ‘Serum protein expression
(2003), ‘A panel of cerebrospinal fluid
profiling for cancer detection: validation of a
potential biomarkers for the diagnosis of
SELDI-based approach for prostate cancer’,
Alzheimer’s disease’, Proteomics, Vol. 3, pp.
Dis. Markers, Vol. 19, pp. 185–195.
1486–1494.
103. Paweletz, C. P., Trock, B., Pennanen, M.
116. Clarke, W., Silverman, B. C., Zhang, Z. et al.
et al. (2001), ‘Proteomic patterns of nipple
(2003), ‘Characterization of renal allograft
aspirate fluids obtained by SELDI-TOF:
rejection by urinary proteomic analysis’, Ann.
potential for new biomarkers to aid in the
Surg., Vol. 237, pp. 660–664.
diagnosis of breast cancer’, Dis. Markers, Vol.
17, pp. 301–307. 117. Forde, C. E. and McCutchen-Maloney, S. L.
(2002), ‘Characterization of transcription
104. Zhang, Y. F., Wu, D. L., Guan, M. et al. factors by mass spectrometry and the role of
(2004), ‘Tree analysis of mass spectral urine SELDI-MS’, Mass Spectrom. Rev., Vol. 21, pp.
profiles discriminates transitional cell 419–439.
carcinoma of the bladder from noncancer
patient’, Clin. Biochem., Vol. 37, pp. 118. Bane, T. K., LeBlanc, J. F., Lee, T. D. and
772–779. Riggs, A. D. (2002), ‘DNA affinity capture
and protein profiling by SELDI-TOF mass
105. Won, Y., Song, H. J., Kang, T. W. et al. spectrometry: effect of DNA methylation’,
(2003), ‘Pattern analysis of serum proteome Nucleic Acids Res., Vol. 30, p. e69.
distinguishes renal cell carcinoma from other
urologic diseases and healthy persons’, 119. Espina, V., Dettloff, K. A., Cowherd, S. et al.
Proteomics, Vol. 3, pp. 2310–2316. (2004), ‘Use of proteomic analysis to monitor

2 3 8 & HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004


Differential proteomics

responses to biological therapies’, Expert Commun., Vol. 290, pp. 1328–1335.


Opin. Biol. Ther., Vol. 4, pp. 83–93.
122. Schmidt, F., Donahoe, S., Hagens, K. et al.
120. Carter, D., Douglass, J. F., Cornellison, C. D. (2004), ‘Complementary analysis of the
et al. (2002), ‘Purification and Mycobacterium tuberculosis proteome by two-
characterization of the mammaglobin/ dimensional electrophoresis and isotope-
lipophilin B complex, a promising diagnostic coded affinity tag technology’, Mol. Cell.
marker for breast cancer’, Biochemistry, Vol. Proteomics, Vol. 3, pp. 24–42.
41, pp. 6714–6722.
123. Tian, Q., Stepaniants, S. B., Mao, M. et al.
121. Forde, C. E., Gonzales, A. D., Smessaert, J. (2004), ‘Integrated genomic and proteomic
M. et al. (2002), ‘A rapid method to capture analyses of gene expression in mammalian
and screen for transcription factors by SELDI cells’, Mol. Cell. Proteomics, Vol. 3, pp.
mass spectrometry’, Biochem. Biophys. Res. 960–969.

& HENRY STEWART PUBLICATIONS 1473-9550. B R I E F I N G S I N F U N C T I O N A L G E N O M I C S A N D P R O T E O M I C S . VOL 3. NO 3. 220–239. NOVEMBER 2004 2 3 9

Das könnte Ihnen auch gefallen