Anda di halaman 1dari 11

Analytica Chimica Acta 556 (2006) 69–79

Review

Advanced analytical tools in proteomics


Resmi C. Panicker a , Souvik Chattopadhaya b , Shao Q. Yao a,b,∗
a Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore
b Department of Biological Science, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore

Received 27 March 2005; received in revised form 16 May 2005; accepted 26 May 2005
Available online 1 July 2005

Abstract

Proteomics deals with the study of proteins, their structures, localizations, posttranslational modifications, functions and interactions with
other proteins. The mapping of protein structure–function holds the key to a better understanding of cellular functions under both normal and
disease states, which is critical for modern drug discovery. However, the study of human proteome presents scientists with a task much more
daunting than the human genome project. In fact, the estimated >100,000 different proteins expressed from 30,000 to 40,000 human genes make
it extremely challenging, if not impossible with existing protein analysis techniques, to map the entire cellular functions at the translational
level. Consequently, there have been rapid advances in the techniques and methods capable of large-scale proteomic studies. Among them, the
recently developed high-throughput screening methods have enabled scientists to analyze proteins quickly and efficiently at an organism-wide
scale. Herein, we overview some of these emerging tools for high-throughput protein analysis. In particular, we focus on recent advances in the
bioassay development, which has provided sensitive and selective tools for high-throughput identification and characterizations of enzymes.
Finally, the recently developed bioimaging techniques to visualize and quantify proteins in living cells are also discussed.
© 2005 Elsevier B.V. All rights reserved.

Keywords: Microarray; Peptide; Protein; Enzyme; Proteomics; 2D gel electrophoresis; Protein labeling; Bioimaging

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69
2. Analytical techniques in proteomics—an overview . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 70
3. Enzyme-profiling assays on chips . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71
3.1. Enzyme assays using protein arrays . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71
3.2. Enzyme assays using peptide/small molecule substrate arrays . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 72
3.3. Enzyme assays using other types of arrays . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 74
4. Bioimaging—an emerging technique for protein analysis in living cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 76
5. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78

1. Introduction functions and interactions with other proteins. The mapping


of protein structure–function holds the key to a better under-
Proteomics deals with the study of proteins, their standing of cellular functions under both normal and disease
structures, localizations, posttranslational modifications, states, which is critical for modern drug discovery. However,
the study of human proteome presents scientists with a task
∗ Corresponding author. Tel.: +65 68741683; fax: +65 67791691. much more daunting than the human genome project [1].
E-mail address: chmyaosq@nus.edu.sg (S.Q. Yao). In fact, the estimated >100,000 different proteins expressed

0003-2670/$ – see front matter © 2005 Elsevier B.V. All rights reserved.
doi:10.1016/j.aca.2005.05.060
70 R.C. Panicker et al. / Analytica Chimica Acta 556 (2006) 69–79

from 30,000 to 40,000 human genes make it extremely within these complex mixtures with liquid chromatography
challenging, if not impossible with existing protein analysis and mass spectrometry [35]. More recently, the gel-based
techniques, to map the entire cellular functions at the transla- ICAT has also been reported [36]. The ICAT approach is
tional level. Consequently, there have been rapid advances in able to simplify the analysis of the proteome mixture by two
the techniques and methods capable of large-scale proteomic orders of magnitude based on the occurrence of cysteine in
studies. Among them, the recently developed high- proteins (1.7%). However, the strategy is limited to probing
throughput screening methods have enabled scientists to known proteins containing cysteine residues and depends on
analyze proteins quickly and efficiently at an organism-wide non-specific binding to CAT reagents.
scale. Herein, we overview some of these emerging tools for From the very beginning of the proteomic era, mass spec-
high-throughput protein analysis. In particular, we focus on trometry (MS) has played a major role in the high-throughput
recent advances in the bioassay development, which has pro- identification of proteins following separation techniques
vided sensitive and selective tools for high-throughput iden- such as 2D-GE, ICAT, etc. With the advent of the ‘soft’ ioniza-
tification and characterizations of enzymes [2–29]. Finally, tion techniques such as ESI and MALDI coupled with various
the recently developed bioimaging techniques to visualize mass analyzers including the ion trap, time-of-flight (TOF),
and quantify proteins in living cells are also discussed. quadrupole and Fourier transform ion cyclotron (FT-MS),
the analysis of large intact protein complexes is now possible
[37], thus providing complementary data to that obtained by
2. Analytical techniques in proteomics—an overview well-established methods in structural biology such as elec-
tron microscopy, X-ray crystallography and NMR. Another
The conventional two-dimensional gel electrophoresis MS-based technology for quantitative analysis of protein
(2D-GE), in combination with advanced mass spectrometric mixtures is known as the surface-enhanced laser desorption
techniques, has facilitated the rapid characterization of thou- ionization-time-of-flight (SELDI-TOF) [38]. This technique
sands of proteins in a single polyacrylamide gel. The ability of utilizes stainless steel or aluminum-based supports, or chips,
this technique to separate thousands of proteins in a specific engineered with chemical or biological bait surfaces that
cell or tissue, including their posttranslationally modified allow differential capture of proteins (based on the intrinsic
forms, has enabled it to become a major separation technique properties of proteins). After the removal of non-specifically
in protein analysis and, thus, is well suited for the global adhered proteins, the bound proteins are laser-desorbed and
analysis of protein expression in an organism. This, together ionized for MS analysis. This field is currently developed
with the newly developed activity-based profiling approaches as a prominent technique when combined with advances in
that target different classes of proteins, has now allowed the protein chip technology.
study of protein functions based on their intrinsic enzymatic While DNA microarray has enabled the high-throughput
activities [5,9]. However, 2D-GE suffers from a number of analysis of genetic materials in a miniaturized format
long-standing problems, including low throughput, a limited [39], the development of similar devices for the analysis
dynamic detection range, poor reproducibility, low sensitiv- of proteins has largely been hindered till the end of 20th
ity, as well as its difficulties in analyzing hydrophobic, small, century, primarily due to the more delicate nature of protein
and very basic or acidic proteins. Incremental improvements structures. Early attempts had focused mainly on the use
in the 2D-GE technology, including the use of sensitive stain- of peptide libraries generated in situ using methods such
ing methods and higher-resolving gels, and sample fractiona- as SPOTTM synthesis [40], light-directed parallel synthesis
tion prior to 2D-GE, have alleviated, but not eliminated, some using photolithographic methods [41] and so forth. In
of these problems [30,31]. Differential gel electrophoresis 2000, MacBeath and Schreiber published their landmark
(DIGE) [32] and Multiplexed proteomics Approach (MP paper and put forward, for the first time, the feasibility of
approach) [33] are some other recent developments related to using microarray-based technologies for high-throughput
2D-GE. Yates and co-workers introduced a multidimensional protein analysis [42]. Ever since, many more examples of
protein identification technology (MudPIT) [34]. In this tech- array-based proteomic tools have been demonstrated, which
nique, multiple types of columns are coupled together to allow the elucidation of multiple proteins by their different
separate proteins using different physiochemical properties in biological properties, i.e. expression, activity and protein
addition to molecular weights and charges, thereby extending interactions, etc. [2,6,10]. Like DNA microarray, the beauty
the analytical range to proteins having high or low molecular of protein microarray lies in the fact that it allows the simul-
weights, as well as low-abundance and insoluble proteins. taneous determination, in a single experiment, of thousands
MudPIT also allows the flexibility of incorporating a suit- of proteins under a variety of parameters with only minute
able affinity column to enable selective analysis of protein amounts of samples. Most of the microarray-based screening
complexes and protein–protein interactions. Among differ- methods developed so far have relied on the detection of
ent isotope-based proteomics techniques, the isotope-coded protein functions by virtue of the non-covalent binding of
affinity tagging (ICATTM ) approach utilizes stable isotope a protein with its ligand(s) thereby limiting the screening to
labeling to perform quantitative analysis of paired protein protein–protein, protein–ligand or antigen–antibody interac-
samples, followed by separation and identification of proteins tions. As a result, these approaches essentially preclude the
R.C. Panicker et al. / Analytica Chimica Acta 556 (2006) 69–79 71

screening of enzymes, which play vital roles in all cellular


processes and metabolic transformations. Traditionally,
enzymatic assays are carried out in a microplate format,
i.e. in an ELISA, where enzymes are mixed separately with
their substrates in individual wells, and their activities read
out by the measurement of substrate turnovers [10]. The
wells function as individual compartments to confine each
enzymatic assay, thus preventing its cross contaminations
from neighboring reactions. However, with the emergence
of protein microarray and related technologies, recent efforts
have been focused on the further miniaturization of enzyme
assays which may analyze multiple enzyme activities in a
microarray format, both reproducibly and quantitatively.
This will be discussed in the following paragraphs.

3. Enzyme-profiling assays on chips

Enzyme assays not only detect enzymes but also can be


used to profile enzymes in terms of their substrate preference
to generate “fingerprint” profiles that are unique for each
enzyme [43]. Such analysis reveals the type of chemical
entities accepted by the enzyme as its potential substrates,
and thereby helps in a better understanding of its catalytic
mechanism and properties. Similarly, the unique pattern
generated for an unknown enzyme using a set of known
substrates may possibly be used to delineate its identity. Con-
ceptually, enzyme-profiling assays on chips may be carried
out with any of the three existing microarray-based platform
technologies—namely the protein array, the peptide array
and the small molecule array [2,4,6]. Immobilization of these
biomolecules to solid supports is a crucial step to retain the
specific activity and has been reviewed extensively [3]. As a
viable and global assay format, the major detection schemes
to track enzyme activities and on-chip assays for enzyme
identification, profiling and the kinetic measurements are key
developments. Different types of enzyme assays have been
developed which are primarily based on well-known detec-
tion methods, i.e. fluorescence, radioactivity and chemilumi-
nescence [43]. Among them, fluorescence-based methods are
most suitable because of their operational simplicity, reliabil-
ity and greater sensitivity for low enzyme turnover reactions.
Fig. 1. (a) Principle of activity-based detection of enzymes in a microarray;
3.1. Enzyme assays using protein arrays (b) mechanism-based probes used for detection of phosphatases (PT-Cy3),
cysteine (VS-Cy3) and serine (FP-Cy3) proteases; (c) enzymatic screening
With the introduction of protein microarray as the plat- using PT-Cy3 (Panel A), VS-Cy3 (Panel B) and FP-Cy3 (Panel C) of 12
form technology for high-throughput enzyme screenings, the commercial enzymes on a microarray.
critical issue is to develop strategies that allow the simulta-
neous assessment of the activity of individual proteins while enzyme assays, including enzyme substrates, from freely dif-
minimizing cross contaminations from neighboring proteins fusing across the whole array surface thus makes it difficult,
immobilized on the same surface. As mentioned earlier, if not impossible, for microplate-based enzymatic assays to
enzyme assay in a microplate format is straightforward, as be transferred directly to a microarray format. To address
each well serves as an individual compartment to confine this problem, we recently developed a novel activity-based
each enzymatic reaction. In a protein microarray, thousands strategy that allows sensitive detection of enzymes immobi-
of protein spots are immobilized on a single flat surface. The lized in a protein microarray (Fig. 1a) [15]. Small molecule
lack of any physical barrier which prevents reagents used in probes derived from mechanism-based suicide inhibitors
72 R.C. Panicker et al. / Analytica Chimica Acta 556 (2006) 69–79

(Fig. 1b) had previously been reported only in gel-based was also used to probe the enzyme specificity and inhibition
methods for the global analysis of enzyme expression and using different peptide substrates. Ellman et al. recently
functions [5]. By covalent modifications of target enzymes developed an elegant approach for potential high-throughput
in a highly selective manner, these probes facilitate the easy profiling of proteases in a microarray format [53]. They made
identification and/or purification of proteins from complex use of combinatorial libraries of peptidyl coumarin deriva-
proteomes. Several types of probes have been developed tives immobilized site-specifically in a peptide microarray.
that target different classes of proteins on the basis of their 7-amino-4-carbamoylmethyl coumarin (ACC) was used
enzymatic activities [23–29]. More recently, affinity-based as the fluorescence reporter. We independently developed
approaches have also been developed to profile other classes a similar approach for high-throughput identifications of
of enzymes previously inaccessible by activity-based profil- hydrolytic enzymes including esterases, lipases, proteases
ing approaches [22]. As depicted in Fig. 1a, we successfully and epoxide hydrolases [21]. In our method, a fluorogenic
extended this activity-based enzyme profiling approach to coumarin derivative was suitably modified to generate a
protein microarray. Our studies showed that these probes are series of substrates that were immobilized on a glass slide to
suitable for array-based detection of enzyme activities and yield a small molecule array. Upon enzymatic cleavage of the
inhibitions [15]. Incubation of these probes with an enzyme head/substrate, the fluorescent coumarin was “unmasked”
array leads to its reaction with the immobilized enzymes thereby allowing for enzymatic activity to be translated to
by virtue of their activity against the inhibitors. Because the fluorescent readouts (Fig. 2). Besides these studies, the use
probes are pre-labeled with a fluorescent dye, the formation of fluorescently tagged affinity ligand as a means to quantita-
of covalent enzyme-inhibitor adducts renders the enzymes tively measure on-chip bindings of reversible and irreversible
detectable. Our strategy was successfully carried out with inhibitors to proteases has been reported by Miyake and
a protein microarray immobilized with different classes of coworkers [54].
enzymes, including phosphatases, cysteine proteases, serine For profiling kinases, peptide phage display [55], on-
hydrolases etc. using a panel of different mechanism-based bead peptide libraries [56], or SPOTTM technology [57]
probes as given in Fig. 1c. are the early-developed methods. The earliest functional
kinase assay in a microarray format was in the example of
3.2. Enzyme assays using peptide/small molecule Kemptide phosphorylation catalyzed by cAMP-dependent
substrate arrays protein kinase A (PKA) [42]. In a more pervasive study, Zhu
et al. tested 119 of the 122 putative yeast kinases against
Peptide/small molecule substrate arrays were used to a panel of 17 different proteins immobilized in a nanowell
identify the enzymatic activity of proteases and other format. Peptides as kinase substrates were at first used by
hydrolytic enzymes, kinases and Carbohydrate-modifying Falsey et al. who showed the incorporation of 33 P into
enzymes. Proteases are highly discriminating for substrate an immobilized peptide by p60c-src tyrosine kinase [58].
cleavage sequences. Though a number of approaches, such Alternatively, Houseman et al. developed self-assembled
as the use of FRET peptides [44], substrate phage libraries monolayer (SAM)-based peptide microarrays for the rapid
[45], positional-scanning libraries [46,47], mixture-based and quantitative evaluation of kinase activity [59–61].
and oriented peptide libraries [48,49] have been described A recent addition to the identification of peptidic kinase
to determine the substrate specificity of proteases, they do substrates has been the use of “phospho-site” collection
not possess the degree of throughput normally provided arrays, which consist of libraries of peptides derived from
for by microarray-based technologies. Consequently, the annotated phosphorylation sites in human proteins [62,63].
complete specificity profile of a protease, as well as the In order to avert the drawbacks of radioactivity-based detec-
kinetic evaluation of all of its potential substrates, is not tion methods used in the aforementioned studies without
easily attainable. To address these shortcomings, a number the loss in sensitivity, our group developed an alternative
of microarray-based protease profiling methods have been detection scheme based on the use of fluorescently labeled
developed [50–53]. By using a solid support for peptide anti-phosphoamino antibodies as depicted in Fig. 3a [19].
immobilization, Kiyonaka et al. studied protease activities In recent years, another fluorescence-based method for
using fluorogenic peptides trapped in a 3D supra-molecular the detection of phosphorylated peptides/proteins has also
hydrogel [50]. Gosalia and Diamond recently described become commercially available, which makes use of the
the use of a liquid-phase microarray that utilizes nanoliter dye ProQ DiamondTM [64,65]. To further expand the utility
sample volumes for protease screenings [51]. Such fluid of microarray-based techniques in the profiling of kinase
phase reactions are advantageous as it is possible to tailor substrate specificities, we developed a combinatorial method
optimized reaction conditions at each individual position on to characterize kinase substrates (Fig. 3b) [20]. Traditional
the array. A chromogenic assay for detecting hydrolases was microarrays employ a ‘one-spot, one-compound’ approach
described by Park and Clark [52]. In this method, hydrolases for the direct identification of positive hits. This, however,
were encapsulated in sol–gel microstructures to create the limits the number of compounds that can be studied in an
‘solzyme’ array and hydrolysis was monitored by the color array to what may be synthesized, thus severely limiting
change of a generic indicator, bromothymol blue. The array throughput. In order to study a greater diversity of compounds
R.C. Panicker et al. / Analytica Chimica Acta 556 (2006) 69–79 73

Fig. 2. (a) Strategies for detection of hydrolytic enzymes on microarrays (route I—for proteases and phosphatases, route II—for epoxide hydrolases and
esterases); (b) chemical structure of the fluorogenic substrates (1–4) used for the detection of the different hydrolases; (c) fluorescence profiles generated on
microarray following treatment with (A) epoxide hydrolase from Rhodococcus rhodochrous, (B) acetylcholine esterase from Electrophorus electricus, (C)
trypsin (D) alkaline phosphatase. 7-hydroxy-4-carboxymethylcoumarin was used as control for all the experiments.

more rapidly and efficiently, it is imperative that alternative have shown that, by spotting libraries with various combina-
strategies like combinatorial peptide synthesis be suitably tions of peptide sequences, it is possible to draw conclusions
employed together with array-based methods to facilitate about positive hits or substrate specificity without generating
the development of next-generation peptide microarrays. We large numbers of peptides sequences individually.
74 R.C. Panicker et al. / Analytica Chimica Acta 556 (2006) 69–79

Fig. 3. (a) Overall strategy for on-chip detection of kinase substrates using anti-phosphoamino antibodies; (b) intensity profiles and image obtained with the
different combinatorial libraries following treatment with p60c-src kinase: (1–6) deletion libraries, (7–13) alanine-scanning libraries, (16–25) positional-scanning
libraries, (14) full combinatorial mixture peptide libraries, (15) original putative substrate. All peptides were synthesized with a CGG linker at the N-terminus
and immobilized on PEGylated thioester slides.

A major impediment in glycobiology has been the lack of specificity of lectins against an array of monosaccharide-thiol
convenient and effective tools for the synthesis and analysis conjugates immobilized onto a self-assembled monolayer
of complex and structurally diverse carbohydrates [66]. Sig- (SAM)-derivatized with maleimide groups [60]. Park et
nificant advances in chemical and enzymatic methods for the al. reported the fabrication of carbohydrate arrays by
solid-phase synthesis of complex carbohydrates in the past immobilization of maleimide-terminated carbohydrates on
few years have eased the development of tools to explore new thiol-coated glass slides [70]. Using these arrays, the group
vistas in carbohydrate biology [67]. On the technology front, demonstrated both qualitative and quantitative bindings of
carbohydrate arrays have been investigated in a series of lectins to ␣-, ␤- and N-linked carbohydrates.
reports [68,69]. The usefulness of these arrays for functional
enzymatic studies has also been demonstrated. Houseman 3.3. Enzyme assays using other types of arrays
and Mrksich reported a carbohydrate microarray in which
Diels–Alder reaction was used for carbohydrate immobiliza- Schultz and colleagues reported the development of a pep-
tion [59]. They subsequently used the “carbochip” to profile tide nucleic acid (PNA)-tagged, small molecule array that, in
the binding specificities between rhodamine-labeled lectins conjunction with the standard DNA microarray technology,
and different monosaccharides. They also characterized can be used to monitor the level and activities of enzymes in
the substrate specificity of ␤-1,4-galactosyltransferase a proteomic scale [71,72]. Using the split-pool combinatorial
and quantitatively estimated the inhibitory concentration of method, a small molecule library based on mechanism-based
␣-methyl mannose for concanavalin A in competitive binding inhibitors of cysteine proteases was synthesized, with each
studies. In a related study, the same group profiled the binding member of the library having a PNA tag. The PNA tags did not
R.C. Panicker et al. / Analytica Chimica Acta 556 (2006) 69–79 75

Fig. 4. (a) Schematic representation of expression display; (b) structure of the activity-based probe specific for identification of protein tyrosine phosphatases
(PTPs); (c) hybridization of Cy3-labeled reverse transcripts following in vitro selection using activity-based probe onto a “decoding” DNA microarray containing
384 yeast ORFs (in duplicates). The spotting pattern is indicated by white circles. Zoomed regions represent positive hits.

alter the activity/selectivity of these small molecules and, at to the small molecule, the approach may potentially be used
the same time served as molecular “barcodes” which could be to guide the design of drugs against diseased phenotypes.
used to decode the library by hybridization to a suitable DNA Our own inputs in the area of genome-wide identification
microarray. Alternatively, the utility of this method to monitor of enzyme activities using the DNA microarray technology
enzymatic activities in a proteomic scale was demonstrated have been through the development of a technique termed
by the detection and characterization of the caspase-3 activity “expression display” as shown in Fig. 4a which combines the
in crude extracts of cells undergoing apoptosis triggered by advantages of three different techniques—ribosome display,
Granzyme B. Since the enzyme activity governs its binding DNA microarray and activity-based enzyme profiling [11].
76 R.C. Panicker et al. / Analytica Chimica Acta 556 (2006) 69–79

In this method, a mixture of proteins was expressed, in a some of the small molecule-based protein labeling tech-
single vessel, from their cDNA library by ribosome display, niques developed in recent years, which holds great promises
which ensured that each protein member was linked and in future high-throughput protein analysis in live cells.
thus encoded by its own mRNA sequence, via the formation A few groups, including our own, have developed novel
of a stable mRNA–ribosome–protein ternary complex. strategies for specific labeling of proteins with small molecu-
Subsequent functional selections using an activity-based lar probes in live cells. Different reported strategies are based
small molecule probe enriched only complexes that contain on unique chemistries and bio-interactions. Of different
proteins having the desired enzymatic activity as given in small molecule probes used, a number of criteria should
Fig. 4b. The identity of these enzymes was then decoded be carefully considered, including their cell permeability,
in a high-throughput manner, following hybridization of cytotoxicity, specific reactivity, good fluorescent properties,
their tethered mRNA to a DNA microarray. So far we have etc. The first innovative method for the site-specific labeling
demonstrated that this approach could be used to isolate and of recombinant protein with small organic molecules within
identify, in a single experiment, representative yeast tyrosine live cells was developed by Tsien et al. [76]. Their method
phosphatases from a large pool of unrelated proteins as exploits the well-known specificity of organoarsenicals with
depicted in Fig. 4c. We envision that, in future, this work pairs of thiols. Here a short peptide sequence CCXXCC (in
might be extended to the high-throughput screening of other which X is a non-cysteine amino acid) was genetically fused
enzyme classes. to the protein of interest, which was subsequently recognized
and labeled by a cell permeable fluorescein derivative called
FlAsH as shown in Fig. 5a. The free FlAsH label is virtually
4. Bioimaging—an emerging technique for protein non-fluorescent, but acquiring bright fluorescence only upon
analysis in living cells binding to CCXXCC motif present in the target protein [76].
Tsien et al. chose ECFP (enhanced cyan fluorescent protein)
While most enzyme assays are performed with proteins as a model protein and genetically fused a CCXXCC motif
either purified to homogeneity or present in a crude cell to its C-terminus. Upon transient expression in HeLa cells
lysate, it is much more meaningful if one can assess a and treatments of the cells with FlAsH, the formation of
target enzyme directly under its own cellular environments. a specifically labeled protein–small molecule complex
In a broader sense, analytical techniques that allow the was successfully detected by monitoring the fluorescence
visualization and quantitation of proteins, including resonance energy transfer (FRET) from ECFP to FlAsH.
enzymes, in living cells is essential to study their structure The age and life cycle of protein molecules could be studied
and functions within the biological context. With the advent by sequential labeling with differently colored biarsenical
of fluorescent proteins and other molecular labels, cell labels. Gaietta et al. applied this strategy to monitor the
biologists and protein chemists have witnessed a better translocation of connexin in and out of gap junctions [77].
understanding of intracellular proteins and subcellular events. Another protein-labeling method was recently developed by
Together with the advancement in fluorescence and confocal Kepplar et al., which covalently labels a target protein fused
laser scanning microscopies (CLSM), 3D imaging of the to the human DNA repair protein O6 -alkylguanine-DNA
cellular events and protein dynamics can now be detected in alkyltransferase, or hAGT [78]. Specific covalent labeling
real time within living cells [73]. However, the main draw- of the target protein with a small molecule was achieved
backs of GFP-like proteins include their large sizes, obligate through the highly specific enzymatic reaction between
oligomerization, and sometimes slow or incomplete matura- hAGT and the small molecule. The cellular role of hAGT is to
tion. Ideally, a good protein labeling strategy should satisfy irreversibly transfer the alkyl group from O6 -alkylguanine-
the following criteria: (1) possesses a high signal-to-noise DNA to one of its reactive cysteine residues, which results
ratio (i.e. high specificity for target protein); (2) upholds the in the repair of the alkylated DNA. The authors brilliantly
integrity of the labeled protein; (3) does not interfere with the exploited this chemistry to label the hAGT fusion protein
biochemical functions or cellular localization of the labeled expressed in live mammalian cells as depicted in Fig. 5b.
protein; (4) has minimal perturbation to the normal cellular Our group has developed a novel strategy for site-specific
processes. Many of these could be met by small molecule- covalent labeling of proteins in vivo. In our strategy, a pro-
based labeling strategies, which in recent years have become tein of interest bearing an N-terminal cysteine is expressed
increasingly feasible for bioimaging experiments, as a result inside a live cell by intein-mediated protein splicing. We
of advances in the synthesis of novel fluorescent dyes, as selected the 17 kDa Ssp DnaB mini intein, whose splicing
well as the development of new bioconjugation strategies activity was genetically engineered to occur efficiently under
which allow for highly specific and efficient incorporation of physiological conditions, to generate an N-terminal cysteine
molecular probes to proteins expressed in live cells [74,75]. in the target protein. Chemoselective native chemical lig-
Since probes used for protein labeling in these strategies are ation reaction between the thioester containing probe and
typically small, they likely do not perturb the function of the the N-terminal cysteine of the protein under physiological
labeled protein, thus making them attractive over FP-based cellular conditions results in the specific protein labeling
strategies. In the following paragraphs, we will summarize as given in Fig. 5c. Our strategy provides a simple way of
R.C. Panicker et al. / Analytica Chimica Acta 556 (2006) 69–79 77

Fig. 5. (a) Site-specific labeling of tetracysteine motif by FlAsH; (b) mechanism of labeling of hAGT fusion proteins with BG derivative; (c) labeling mechanism
of thioester probe with the N-terminal cysteine proteins in live cells; (d) NTA probe coordinating to Ni (II) ion; (e) labeling of fusion proteins based on interactions.

site-specifically labeling proteins in live cells, with little mod- and a metal ion chelating to the nitrilotriacetate (NTA) moi-
ification to the original protein apart from the addition of at ety. The labeling is based on the well-known interaction
most a few extra amino acids. We have successfully shown its between the oligohistidine sequence and the NTA moiety.
utility in specific covalent labeling of proteins in bacteria [79], The chemical structure of the NTA probe used for the experi-
as well as in mammalian cells [80]. More recently, our group ments is shown in Fig. 5d. By applying this approach, the
developed a proteomic method that allows identification of authors were able to investigate the structure and plasma
caspases and their associating proteins from live apoptotic membrane distribution of the ionotropic 5HT3 serotonin
cells [29]. We found that efficient in vivo labeling of caspases receptor (5HT3 R). A separate method developed by Tsien’s
expressed inside apoptotic HeLa cells could be achieved group takes advantage of the very high affinity of avidin for its
using fluoromethylketone (fmk)-containing, activity-based small molecule ligand, biotin (10−15 M) as shown in Fig. 5e.
small molecule probes. We anticipate that, given the increas- In their approach, avidin functions as the genetically encoded
ing availability of activity-based probes that target other tag and the derivatized biotin acts as the label. The luminal
enzymes [22,23], our approach may provide a viable tool for pH of specific organelles expressing an avidin-fused pro-
future high-throughput studies of enzyme-associating pro- tein was investigated using a cell-permeable, pH-sensitive
teins. fluorescein-biotin derivative [82]. Another strategy involv-
Recently, Vogel et al. reported a generic method for the ing the selective binding of a chemical ligand to a receptor
site-selective and reversible labeling of membrane proteins protein is that based on the hapten–antibody interaction as
containing a polyhistidine sequence in live cells with small given in Fig. 5e. A membrane-permeant phenyloxazolinone
molecular probes [81]. This probe consists of a chromophores hapten conjugated to a pH-dependant fluorescent dye was
78 R.C. Panicker et al. / Analytica Chimica Acta 556 (2006) 69–79

trapped by a single chain antibody that had been expressed in Acknowledgements


the lumen of the organelles and used to study pH regulation
in different compartments along the cellular secretory path- Funding support was provided by the National University
way [83]. The noncovalent interaction between dihydrofolate of Singapore (NUS) and the Agency for Science, Technology
reductase (DHFR) and methotrexate (Mtx)-containing small and Research (A*STAR) of Singapore. S.Q.Y. is the recip-
molecule derivates is a good alternative to avidin–biotin and ient of the 2002 Young Investigator Award (YIA) from the
hapten–antibody interactions as depicted in Fig. 5e. The Cor- Biomedical Research Council (BMRC).
nish group very recently exploited this binding as a generic
method for in vivo labeling of DHFR-fused proteins using References
fluorescent, small molecule probes conjugated to Mtx [84].
They were able to successfully label DHFR-fused proteins [1] C. Ezzell, Sci. Am. 283 (2000) 64.
targeted to the nucleus and plasma membrane in DHFR- [2] M. Uttamchandani, D.P. Walsh, S.Q. Yao, Y.T. Chang, Curr. Opin.
deficient CHO mammalian cells. Chem. Biol. 9 (2005) 4–13.
[3] D.S.Y. Yeo, R.C. Panicker, L.P. Tan, S.Q. Yao, Comb. Chem. High
Inteins are internal polypeptide sequences that are Throughput Screen. 7 (2004) 213.
posttranslationally excised from a protein precursor by [4] R.C. Panicker, X. Huang, S.Q. Yao, Comb. Chem. High Throughput
a self-catalyzed protein-splicing reaction. The result is Screen. 7 (2004) 547.
the concurrent ligation of the two flanking polypeptides [5] Y. Hu, X. Huan, G.Y.J. Chen, S.Q. Yao, Mol. Biotechnol. 28 (2004)
sequences to generate a mature, functional protein. In 63.
[6] M. Uttamchandani, X. Huang, G.Y.J. Chen, L.P. Tan, S.Q. Yao, Mol.
recent years, intein-based labeling methods have become an Biotechnol. 28 (2004) 227.
important technique in protein engineering [85]. Recently, [7] R.Y.P. Lue, G.Y.J. Chen, Q. Zhu, M.L. Lesaicherre, S.Q. Yao, Meth.
Muir’s group developed a novel strategy to specifically Mol. Biol. 264 (2004) 85.
incorporate a synthetic molecule into a protein in vivo by [8] M. Uttanchandani, G.Y.J. Chen, M.L. Lesaicherre, S.Q. Yao, Meth.
utilizing trans-splicing inteins, in which the intein is split Mol. Biol. 264 (2004) 191.
[9] X. Huang, E.L.P. Tan, G.Y.J. Chen, S.Q. Yao, Appl. Genomics Pro-
into two pieces and its activity is subsequently regained teomics 2 (2003) 225.
upon reconstitution of these fragments [86]. By exploiting [10] G.Y.J. Chen, M. Uttamchandani, R.Y.P. Lue, M.L. Lesaicherre, S.Q.
this technique, his group was able to successfully label GFP Yao, Curr. Top. Med. Chem. 3 (2003) 705.
with a FLAG epitope inside live mammalian CHO cells. [11] Y. Hu, G.Y.J. Chen, S.Q. Yao, Angew. Chem. Int. Ed. 44 (2005)
Pioneered by the Schultz group, the suppressor tRNA 1048.
[12] L.P. Tan, R.Y.P. Lue, G.Y.J. Chen, S.Q. Yao, Bioorg. Med. Chem.
technology was an ingenious and promising technique that Lett. 14 (2004) 6067.
enables the incorporation of unnatural amino acids into pro- [13] M. Uttamchandani, D.P. Walsh, S.M. Khersonsky, X. Huang, S.Q.
teins during in vitro transcription/translation reactions [87]. Yao, Y.T. Chang, J. Comb. Chem. 6 (2004) 862.
Recently, Zhang et al. designed and utilized an orthogonal [14] R.Y.P. Lue, G.Y.J. Chen, Y. Hu, Q. Zhu, S.Q. Yao, J. Am. Chem.
tRNA–TyrRS pair that selectively and efficiently incorpo- Soc. 126 (2004) 1055.
[15] G.Y.J. Chen, M. Uttamchandani, Q. Zhu, G. Wang, S.Q. Yao, Chem.
rated the unnatural amino acid, m-acetyl-(l)-phenylalanine, Biochem. 4 (2003) 336.
into a protein overexpressed in E. coli, and successfully intro- [16] Y. Hu, G. Wang, G.Y.J. Chen, X. Fu, S.Q. Yao, Electrophoresis 24
duced a “ketone handle” into the target protein [88]. This pro- (2003) 1458.
tein was subsequently modified, in living cells, by hydrazide- [17] M.L. Lesaicherre, R.Y.P. Lue, G.Y.J. Chen, Q. Zhu, S.Q. Yao, J.
containing small molecules using the ketone/hydrazide Am. Chem. Soc. 124 (2002) 8768.
[18] M.L. Lesaicherre, M. Uttamchandani, G.Y.J. Chen, S.Q. Yao, Bioorg.
chemistry, demonstrating for the first time the potential use Med. Chem. Lett. 12 (2002) 2079.
of tRNA technology for live cell bioimaging purposes. The [19] M.L. Lesaicherre, M. Uttamchandani, G.Y.J. Chen, S.Q. Yao, Bioorg.
labeling reaction was selective, and in general proceeded with Med. Chem. Lett. 12 (2002) 2085.
relatively good yields (e.g. >75% ligation product). [20] M. Uttamchandani, E.W.S. Chan, G.Y.J. Chen, S.Q. Yao, Bioorg.
Med. Chem. Lett. 13 (2003) 2997.
[21] Q. Zhu, M. Uttamchandani, D.B. Li, M.L. Lesaicherre, S.Q. Yao,
Org. Lett. 5 (2003) 1257.
5. Concluding remarks [22] E.W.S. Chan, S. Chattopadhaya, R.C. Panicker, X. Huang, S.Q. Yao,
J. Am. Chem. Soc. 126 (2004) 14435.
As new roles of enzymes emerge, many more novel appli- [23] Q. Zhu, A. Girish, S. Chattopadhaya, S.Q. Yao, Chem. Commun.
cations of enzyme assays can be envisioned. Rapid develop- (2004) 1512.
[24] Q. Zhu, X. Huang, G.Y.J. Chen, S.Q. Yao, Tetrahedron Lett. 45
ments in enzymatic assays will continue to be fueled by the (2004) 707.
identification of new enzymes stemming from the growth in [25] Q. Zhu, X. Huang, G.Y.J. Chen, S.Q. Yao, Tetrahedron Lett. 44
genome information which has provided a sequence-based (2003) 2669.
framework for data mining. With new analytical techniques [26] G. Wang, M. Uttamchandani, G.Y.J. Chen, S.Q. Yao, Org. Lett. 5
for high-throughput protein analysis becoming increasing (2003) 737.
[27] G. Wang, M. Uttamchandani, G.Y.J. Chen, S.Q. Yao, Org. Lett. 5
available, it is time now time to look ahead that in the coming (2003) 737.
years, it will be entirely possible that one may be able track [28] M.L. Liau, R.C. Panicker, S.Q. Yao, Tetrahedron Lett. 44 (2003)
enzymes en masse in their own cellular environments. 1043.
R.C. Panicker et al. / Analytica Chimica Acta 556 (2006) 69–79 79

[29] E.L.P. Tan, R.C. Panicker, G.Y.J. Chen, S.Q. Yao, Chem. Commun. [60] B.T. Houseman, M. Mrksich, Chem. Biol. 9 (2002) 443.
(2005) 596. [61] B.T. Houseman, E.S. Gawalt, M. Mrksich, Langmuir 19 (2003)
[30] T. Rabilloud, Proteomics 2 (2002) 3. 1522.
[31] C. Gauss, M. Kalkum, M. Lowe, M.H. Lehrach, J. Klose, Elec- [62] U. Schutkowski, S. Reimer, L. Panse, J.M. Dong, D.R. Lizcano, J.
trophoresis 20 (1999) 575. Alessi, M. Schneider, Angew. Chem. Int. Ed. 43 (2004) 2671.
[32] M. Unlu, M.E. Morgan, J.S. Minden, Electrophoresis 18 (1997) [63] J.M. Lizcano, M. Deak, N. Morrice, A. Kieloch, C.J. Hastie, L.
2071. Dong, M. Schutkowski, U. Reimer, D.R. Alessi, J. Biol. Chem. 271
[33] T.H. Steinberg, K. Pretty, K.N. Berggren, C. Kemper, L. Jones, Z. (2002) 27839.
Diwu, R.P. Haugland, W.F. Pattonet, Proteomics 1 (2001) 841. [64] S. Fukui, T. Feizi, C. Galustian, A.M. Lawson, W. Chai, Nat.
[34] M.P. Washburn, D. Wolters, J.R. Yates, Nat. Biotechnol. 19 (2001) Biotechnol. 20 (2002) 1011.
242. [65] D. Wang, S. Liu, B.J. Trummer, C. Deng, A. Weng, Nat. Biotechnol.
[35] S.P. Gygi, B. Rist, S.A. Gerber, F. Turecek, M.H. Gelb, R. Aebersold, 20 (2002) 275.
Nat. Biotechnol. 17 (1999) 994. [66] K. Martin, T.H. Steinberg, L.A. Cooley, K.R. Gee, J.M. Beechem,
[36] M. Smolka, H. Zhou, R. Aebersold, Mol. Cell. Proteomics 1 (2002) W.F. Patton, Proteomics 3 (2003) 1244.
19. [67] P. Sears, C.-H. Wong, Science 291 (2001) 2344.
[37] J.M. Daniel, S.D. Friess, S. Rajagopalan, S. Wendt, R. Zenobi, Int. [68] O.J. Plante, E.R. Palmacci, P.H. Seeberger, Science 291 (2001)
J. Mass Spectrom. 216 (2002) 1. 1523.
[38] H.J. Issaq, T.P. Conrads, D.A. Prieto, R. Tirumalai, T.D. Veenstra, [69] D.M. Ratner, E.W. Adams, M.D. Disney, P.H. Seeberger, Chem.
Anal. Chem. 75 (2003) 148A. Biochem. 5 (2004) 1375.
[39] D.J. Lockhart, E.A. Winzeler, Nature 405 (2000) 827. [70] S. Park, M.-R. Lee, S.-J. Pyo, I. Shin, J. Am. Chem. Soc. 126 (2004)
[40] R. Frank, Tetrahedron 48 (1992) 9217. 4812.
[41] S.P. Fodor, J.L. Read, M.C. Pirrung, L. Stryer, A.T. Lu, D. Solas, [71] N. Winssinger, J.L. Harris, B.J. Backes, P.G. Schultz, Angew. Chem.
Science 251 (1991) 767. Int. Ed. 40 (2001) 3152.
[42] G. MacBeath, S.L. Schreiber, Science 289 (2000) 1760. [72] N. Winssinger, S. Ficarro, P.G. Schultz, J.L. Harris, Proc. Natl. Acad.
[43] J.P. Goddard, J.L. Reymond, Trends Biotechnol. 22 (2004) 363. Sci. U.S.A. 99 (2002) 11139.
[44] S. Mizukami, K. Kikuchi, T. Higuchi, Y. Urano, T. Mashima, T. [73] G.T. Hermanson, Bioconjugate Techniques, Academic Press, San
Tsuruo, T. Nagano, FEBS Lett. 453 (1999) 356. Diego, CA, 1996.
[45] D.J. Matthews, J.A. Wells, Science 260 (1993) 1113. [74] R. Srinivasan, S.Q. Yao, D.S.Y. Yeo, Comb. Chem. High Throughput
[46] B.J. Backes, J.L. Harris, F. Leonetti, C.S. Craik, J.A. Ellman, Nat. Screen. 7 (2004) 597.
Biotechnol. 18 (2000) 187. [75] D.S.Y. Yeo, R. Srinivasan, G.Y.J. Chen, S.Q. Yao, Chem. Eur. J. 10
[47] J.L. Harris, P.B. Alper, J. Li, M. Rechsteiner, B.J. Backes, Chem. (2004) 4664.
Biol. 8 (2001) 1131. [76] B.A. Griffin, S.R. Adams, R.Y. Tsein, Science 28 (1998) 269.
[48] B.E. Turk, L.C. Cantley, Methods 32 (2004) 398. [77] G. Gaietta, T.J. Deerinck, S.R. Adams, J. Bouwer, O. Tour, D.W.
[49] B.E. Turk, L.L. Huang, E.T. Piro, L.C. Cantley, Nat. Biotechnol. 19 Laird, G.E. Sosinsky, R.Y. Tsien, M.H. Ellisman, Science 296 (2002)
(2001) 661. 503.
[50] S. Kiyonaka, K. Sada, I. Yoshimura, S. Shinkai, N. Kato, I. Hamachi, [78] A. Keppler, S. Gendreizig, T. Gronemeyer, H. Pick, H. Vogel, K.
Nat. Mater. 3 (2004) 58. Johnsson, Nat. Biotechnol. 21 (2003) 86.
[51] D.N. Gosalia, S.L. Diamond, Proc. Natl. Acad. Sci. U.S.A. 100 [79] D.S.Y. Yeo, R. Srinivasan, M. Uttamchandani, G.Y.J. Chen, Q. Zhu,
(2003) 8721. S.Q. Yao, Chem. Commun. (2003) 2870.
[52] C.B. Park, D.S. Clark, Biotechnol. Bioeng. 78 (2002) 229. [80] D.S.Y. Yeo, R. Srinivasan, G.Y.J. Chen, S.Q. Yao, unpublished
[53] C.M. Salisbury, D.J. Maly, J.A. Ellman, J. Am Chem. Soc. 124 results.
(2002) 14868. [81] E.G. Guignet, R. Hovius, H. Vogel, Nat. Biotechnol. 22 (2004) 440.
[54] J. Eppinger, D.P. Funeriu, M. Miyake, L. Denizot, J. Miyake, Angew. [82] J. Llopis, J.M. McCaffery, A. Miyawaki, M.G. Farquhar, R.Y. Tsien,
Chem. Int. Ed. 43 (2004) 3806. Proc. Natl. Acad. Sci. U.S.A. 95 (1998) 6803.
[55] G. Cassirini, FEBS Lett. 307 (1992) 66. [83] J. Farinas, A.S. Verkman, J. Biol. Chem. 274 (1999) 7603.
[56] K.S. Lam, M. Renil, Curr. Opin. Chem. Biol. 6 (2002) 353. [84] L.W. Miller, J. Sable, P. Goelet, M.P. Sheetz, V.W. Cornish, Angew.
[57] W.R.G. Dostmann, M.S. Taylor, C.K. Nickl, J.E. Brayden, R. Frank, Chem. Int. Ed. 43 (2004) 1672.
W.J. Tegge, Proc. Natl. Acad. Sci. U.S.A. 97 (2000) 14772. [85] T.M. Muir, Annu. Rev. Biochem. 72 (2003) 249.
[58] J.R. Falsey, M. Renil, S. Park, S. Li, K.S. Lam, Bioconj. Chem. 12 [86] I. Giriat, T.W. Muir, J. Am. Chem. Soc. 125 (2003) 7180.
(2001) 346. [87] J.A. Ellman, D. Mendal, P.G. Schultz, Science 255 (1992) 197.
[59] B.T. Houseman, J.H. Huh, S.J. Kron, M. Mrksich, Nat. Biotechnol. [88] Z. Zhang, B.A. Smith, L. Wang, A. Brock, C. Cho, P.G. Schultz,
20 (2002) 270. Biochemistry 4 (2003) 6735.

Anda mungkin juga menyukai