William P.
Heal
a,
T. H. Tam
Dang
a and
Edward W.
Tate
*ab
aDepartment of Chemistry, South Kensington Campus, Imperial College, London, SW7 2AZ, UK. E-mail: e.tate@imperial.ac.uk; Fax: +44 (0)20 75941139; Tel: +44 (0)20 75943752
bInstitute of Chemical Biology, South Kensington Campus, Imperial College, London, SW7 2AZ, UK
First published on 1st October 2010
The development and application of chemical technologies enabling direct analysis of enzyme activity in living systems has undergone explosive growth in recent years. Activity-based protein profiling (ABPP) is a key constituent of this broad field, and is among the most powerful and mature chemical proteomic technologies. This tutorial review introduces the essential features of ABPP and the design and application of activity-based probes (ABPs) from drug target elucidation and in vivo visualisation of enzyme activity to comprehensive profiling of the catalytic content of living systems, and the discovery of new biological pathways.
![]() William P. Heal | William Heal received a BSc with honours in chemistry in 1998, and an MSc in chemical process R&D the following year from the University of Liverpool (UK). He stayed in Liverpool to complete a PhD in 2003, studying asymmetric synthesis under the supervision of Professor Stan Roberts, followed by postdoctoral research in medicinal chemistry (prion disease) at the University of Sheffield (UK). He is presently at Imperial College London (UK), undertaking postdoctoral research in the Tate group in the field of chemical biology. He is particularly interested in the design and synthesis of chemical tools for use in studying biological systems. |
![]() T. H. Tam Dang | Tam Dang received a MS in organic chemistry in 2007 from the University of Rostock, Germany, performing her thesis research in the lab of Prof. Peter Langer. She is currently completing her PhD thesis in the group of Dr Ed Tate in the Department of Chemistry, Imperial College London, where she has developed and applied activity-based probes (ABPs) in the pathogenic bacterium Clostridium difficile to track down new proteases and potential drug targets. Outside the lab, Tam enjoys playing the guitar, art, reading and cooking. |
![]() Edward W. Tate | Dr Tate received his PhD in the group of Prof. Steve Ley FRS at the University of Cambridge, and then worked with Prof. Sam Zard at EcolePolytechnique (Paris) as an 1851 Research Fellow. Following postdoctoral research in molecular microbiology at the Pasteur Institute in Paris and in chemical biology at Imperial College London, he was awarded a BBSRC David Phillips Research Fellowship in 2006, and in 2010 he was appointed Senior Lecturer in Chemical Biology in the Department of Chemistry, Imperial College London. His research group currently comprises over 20 researchers engaged in multiple aspects of the design and application of chemical approaches to understanding living systems, with an emphasis on the roles of protein modification in disease. |
Proteomics,1 the quantitative analysis of proteins present in a sample (e.g. a protein complex, a cell or an organ) has enabled many key advances in our understanding of living systems. However, to move this revolutionary technology beyond cataloguing of gene products and into the dissection of the behaviour of living systems we require ways to educe the dynamic properties of proteins that are not directly encoded in the genome. The universal identification, quantification and manipulation of emergent features such as catalytic activity, post-translational modification (PTM, Table 1), and protein complex formation may fairly be described as a ‘grand challenge’ in contemporary science. The application of chemistry to this challenge is proving particularly powerful and has given rise to a vibrant sub-field broadly termed chemical proteomics. In this review we outline the specific challenges posed by measuring catalytic activity in complex living systems, discuss the key role played by chemistry in this rapidly-developing area, and highlight important examples of how chemical tools can been designed and applied.
Catalomics presents some fascinating challenges in chemical biology (Fig. 1). The in vivo activity of enzymes is rarely a simple function of their abundance; it also depends on regulation by substrate co-localisation, post-translational modification, allosteric control, and the co-regulation of endogenous inhibitors. This is perhaps most evident in signalling cascades, where the activity of downstream effectors may be controlled on a sub-second timescale by the activity of upstream regulatory enzymes, which are in turn allosterically activated by receptor complexes. Another important example is the regulated in vivo inhibition of many proteases that is essential to prevent uncontrolled protein cleavage and degradation. These proteases are expressed as an inactive zymogen being rendered catalytically active ‘on demand’ by post-translational modification or allosteric activation.5 This highly emergent in vivo behaviour implies that cell-free experiments with recombinant enzymes can offer only limited, even misleading, information about the biological role of an enzyme. The implications for both basic biology and drug discovery are evident, as is the need for analytical tools that can access the chemistry of enzymes in living systems.
![]() | ||
Fig. 1 The functional catalytic properties of an enzyme in living cells and in vivo are typically modulated by a wide range of phenomena. |
The emerging solution to this challenge is to exploit the incipient in vivo chemistry of enzymes to identify and read out their activity in situ. There are two obvious approaches to achieve this end. The more straightforward is to detect the presence of an active enzyme indirectly through use of a substrate mimic that produces a signal when it is turned over; however, this provides no information on the specific identity of the enzyme or enzyme complex that is responsible for the reaction, and requires a suitable substrate of the enzyme to be known in advance. The more nuanced approach is to detect the active form of the enzyme itself, which has the potential to provide data on both activity and identity. However, this comes at potentially increased risk of disrupting the system under study, and is not always easy to implement. Antibodies, the universal workhorse of bioanalysis, are generally limited to indirect detection of an activating PTM (e.g. protein phosphorylation or proteolytic cleavage).7 Subtle structural changes accompanying allosteric activation are not amenable to antibody detection, and antibodies are generally too cumbersome for use inside cells. The interplay between PTMs and activity can also be very complex, and PTMs are another emergent feature of biological systems that present similar challenges for analysis.8,9 Genetic engineering is also of limited use in isolation since in addition to the shortcomings of timescale noted above, direct labelling of an enzyme by a recombinant tag does not usually provide information on activation and requires the target enzyme to be known in advance. In short, a universal approach to detection and identification of active enzymes requires chemical technologies that can address enzyme chemistry directly in living systems.
![]() | ||
Fig. 2 ABPs label on the basis of activation by the enzyme, whilst AfBPs label on the basis of affinity, stabilised by a non-specific cross-linking. |
An ABP may often be generated directly from a given irreversible inhibitor, which is in turn frequently derived from a substrate mimetic or natural product.12,13 There are also recent instances of ABPs designed in a ‘reverse’ manner by converting a known reversible inhibitor into an irreversible ABP.14 In the case of ABPs, the response to enzyme chemistry is usually encapsulated in a chemical warhead, for example an electrophilic trap such as an epoxide or Michael acceptor. Upon activation by the enzyme the warhead reacts irreversibly to form a new covalent bond to the active site (Fig. 3). By linking enzyme chemistry to ABP activation the warhead typically defines the catalytic class of enzyme targeted enabling distinction, for example, between proteases, phosphatases or glycosidases. The reactivity of the warhead with respect to the enzyme active site depends on both affinity and innate chemical reactivity, and tuning this reactivity offers control of selectivity between enzymes. Highly reactive warheads tend to be promiscuous, whilst others can have negligible reactivity in living systems unless coupled to an element that provides increased affinity, resulting in greatly increased reaction rates for a well-defined set of targets. It is often possible to generate an ABP with either very broad or more selective labelling by including a specificity element that directs the ABP to a particular class of enzymes or to a specific enzyme within a class.
![]() | ||
Fig. 3 Summary of the key features of an activity-based probe (ABP), with some representative examples. |
The key component of an ABP that sets it apart from a common irreversible inhibitor is the presence of a tag or label (Fig. 3). This provides the ‘handle’ with which to manipulate, visualise or quantify the active enzyme: in the act of activating and reacting with the ABP the enzyme labels itself. This handle can range from the simple attachment of a dye molecule directly to the ABP through to advanced labelling technologies that can introduce sets of labels starting from a minimal chemical ‘tag’ by exploiting bioorthogonal ligation or ‘click’ chemistry.15–17 An important caveat is that a label directly appended to the ABP may interfere with the selectivity or activity of the probe, and the use of a ‘clickable’ tag has proven a reliable way to avoid this potential pitfall in many systems.2 ABPs have even been designed to be fluorogenic, switching on their fluorescence in response to enzymatic activation and thereby encompassing certain properties of a fluorogenic substrate. Here we will use the term ‘tag’ to refer to a ‘clickable’ functional group (typically an alkyne) for indirect (‘two-step’) labelling, and ‘label’ to refer to directly labelled ABPs. This distinction between a ‘tag’ and a ‘label’ can be useful, but it is important to note that these semantics are not universally observed by researchers in the field. Recent reports have added a fourth component, a targeting element, which serves to enhance cell uptake or to deliver the ABP to a specific cell type in vivo,18,19 or chemically-cleavable linkers to facilitate selective release of targets.20,21
The modular nature of ABPs (warhead, specificity element, tag/label) lends them to modular synthetic approaches such as solid-phase (peptide) synthesis (SP(P)S). In ideal cases the three components can be varied independently within a single synthetic scheme; this enables rapid access to libraries of variants that can be screened across multiple enzyme classes and exploit orthogonal detection strategies. Click chemistry has also proven a useful tool for the rapid modular assembly of ABPs, as recently reviewed by Yao et al.7
• De novo enzyme discovery: (re)assignment of roles for proteins. This is a key challenge since around 30–40% of gene products of the human genome (and a much larger proportion in many other organisms) currently have no known role.
• Context-independent high-throughput assays for enzyme activity (as opposed to simple expression level). Even where the substrate is unknown and a conventional enzyme activity assay would be difficult to devise, assays using ABPs can enable high-throughput screening.
• Competition assays to determine in vivo selectivity of a reversible or irreversible inhibitor across a particular enzyme class. ABPs enable selectivity to be determined across all potential off-targets, including those with uncharacterised function.
• Fluorogenic probes for enzyme labelling, localisation of enzyme activity by microscopy and whole organism imaging, quantification of enzyme activity in vivo.
• Tools for probing the protein chemistry of enzymes: these probes can also be useful in structural and biochemical studies to manipulate the active site of an enzyme without resorting to mutagenesis.
• In the second part of this review, we will look at three selected case studies that highlight some of these applications.
To start with, a catalomic analysis of a range of aggressive and non-aggressive melanoma, ovarian and breast cancer cell lines was carried out to identify enzymes possessing significant hydrolytic activity (Fig. 4). ABPP using a fluorophosphonate (FP) ABP was carried out, as this functionality is directed towards serine hydrolases,24 which represent one of the largest and most varied groups of hydrolytic enzymes in the human proteome. With roles in proteolysis, signal transduction and lipid processing, it was hoped that hydrolases with modified activity unique to the aggressive lines would be identifiable. The FP functionality reacts to form a covalent bond with the catalytic serine nucleophile of the serine hydrolases, and a FP-biotin probe can distinguish between active and inactive forms of these enzymes. This allows monitoring of enzyme activity, even when this is not related to enzyme expression level. After treatment of the soluble and insoluble cancer cell proteome fractions with the FP-biotin probe, functionalised entities were enriched by a two-stage process of size exclusion chromatography to remove unreacted probe followed by incubation with avidin–agarose beads. Tryptic digestion of enriched enzymes directly from the beads preceded separation of this complex mixture by two-dimensional liquid chromatography on a biphasic (strong cation exchange/reverse phase) capillary column with elution over a 5-step gradient of salt concentration.
![]() | ||
Fig. 4 Strategy for identification of MAGL as a principle component of the metabolic pathways of aggressive cancers. |
Along with analysis of the LC output by ion trap tandem mass spectrometry, this system comprises the so-called multidimensional protein identification technology (MudPIT).25 The mass spectra obtained were analysed against a custom database containing over 20000 unique gene products and the results filtered for serine hydrolases. Of the >50 serine hydrolases detected in this way, two (KIAA1363 and MAGL) exhibited consistently raised activity levels in aggressive lines. Whilst the authors had shown previously that KIAA1363 was involved in lipid signalling in aggressive cancer cell lines,26 little was known about the function of MAGL. Elevated MAGL activity was confirmed by monitoring the hydrolysis of the substrate C20
∶
4 MAG, and upon treatment with a potent and selective MAGL inhibitor (JZL18427) MAG hydrolysis was substantially reduced. The authors demonstrated that highly elevated MAG hydrolysis is an important feature of aggressive cancers that is mostly, if not completely, the result of up-regulation of MAGL activity. MAGL was also found to regulate free fatty acid (FFA) levels in aggressive cancer cells, which is not generally the case in normal cells. Cravatt went on to demonstrate that disruption of MAGL activity (by knock-down and by JZL184 treatment) reduced aggressive cancer pathogenicity, and that inducing over-expression in non-aggressive lines increased their aggressive character.
This work is an elegant example of ABPs in action: starting from a black box of non-aggressive vs. aggressive phenotype, broad-spectrum serine hydrolase probes were used to identify candidate enzymes potentially involved in aggressive cancers de novo from live cells, picking up activity differences that would not have been detected at the expression level. The same probes were used to identify selective inhibitors for MAGL, and to verify modulation of its activity during target validation studies. As a direct result new insights into lipid metabolism in cancer have been gained, and a new drug target has been uncovered for aggressive cancers. In future, detection of enzyme activity using ABPs may be used as an effective biomarker for early detection of aggressive cancers, something that standard transcriptomic or proteomic analyses typically fail to discriminate.
Apoptosis is a programmed and tightly controlled mechanism for cell death, which in normal tissues is an important part of homoestasis and for the destruction of damaged cells. However, incorrect regulation of apoptosis has been estimated to play a role in as much as 70% of human diseases.28 Our understanding of the importance of apoptosis, a form of programmed cell death, in cancer, and of the roles played by proteases in this process, is amongst the most significant breakthroughs in cancer biology in recent years. Members of the cysteine protease family, cysteine-dependent aspartate-directed proteases, or caspases, are the principle executors of the apoptotic process.29 They are involved with activation of DNases (e.g. caspase activated DNase, or CAD), inhibition of DNA repair enzymes (such as poly (ADP-ribose) polymerase, or PARP) and digestion of structural proteins in the nucleus (e.g. lamin, which mediates interactions between chromatin and the nuclear membrane and helps maintain the shape of the nucleus). Each of these functions acts to reduce the genetic and structural integrity of the cell to facilitate its organised non-inflammatory destruction, an essential mechanism by which the body limits carcinogenesis.
Although labelled analogues of annexin V have been reported as probes for apoptosis, they suffer from slow clearance in vivo, resulting in high background.30 Alternatively, Bogyo et al. have shown that acyloxymethyl ketones (AOMK, a chemical warhead) are effective ABPs for labelling caspases in vivo, but they also display significant cross-reactivity with lysosomal cysteine protease legumain and cathepsins.31 However, by iterative optimisation of the specificity of the peptide sequence (using positional scanning libraries, obtained by solid phase peptide synthesis), it proved possible to lower the legumain response and eliminate the cathepsin cross-reactivities altogether. The probe of choice AB50-Cy5 also contains the fluorescent cyanine fluorophore Cy5 (Fig. 5). In addition, the inclusion of well-known cell penetrating peptide trans-activating transcriptional activator (Tat, from HIV-1)32 into the probe produced enhanced uptake and greatly boosted the signal in apoptotic cells and tissues, giving multifunctional probe tAB50-Cy5. The effective inclusion of a targeting element is an excellent example of the versatility possible in ABP design.
To demonstrate the efficacy of these ABPs in probing for caspases Bogyo et al. monitored apoptosis in the CD4+CD8+ thymocytes of mice treated with dexamethasone, a well-characterised in vivo system which has been studied previously with apoptosis markers such as annexin V.33 Mice were monitored over several time points after treatment with dexamethasone and intravenous injection of ABP two hours prior to removal of the thymi for imaging. After imaging, protein was extracted from the tissue and separated by SDS-PAGE, using in-gel fluorescence to visualise labelled proteins. By immunoprecipitation the authors were able to show that caspase-3 and legumain were labelled with probe. Quantification of the total caspase and legumain, and comparison with the data from intact thymi indicated that legumain activity was low and remained so after dexamethasone treatment, whereas caspase-3 was seen to become active rapidly after treatment, peaking at 12 h and dropping to background levels by 24 h. Furthermore, comparing the imaging data from the intact thymi and direct quantification showed that the overall trends are correlated and that whole-organ imaging is a sufficient readout for labelled caspases.
Whilst probing with AB50-Cy5 produced similar labelling patterns, there was a marked increase in fluorescence from intact thymi labelled with tAB50-Cy5. This was not the case in tissue in which apoptosis had not been induced, indicating that the tat peptide was facilitating uptake only in apoptotic cells. Further analysis by flow cytometry showed that, by comparison of fluorescence in apoptotic vs. non-apoptotic cells, the tAB50-Cy5 probe accumulated in the dying cells only. In a further demonstration of the specificity of these probes, intact thymi at 12 h after dexamethasone treatment showed high levels of probe accumulation relative to untreated controls, but with no caspase labelling observable by SDS-PAGE. This demonstrates the significant uptake of probe by cells as they become apoptotic, and precise labelling of caspases dependant on their level of activity.
To demonstrate the utility of these probes in non-invasive imaging, and in a model more closely related to human disease, apoptosis was studied in mice that had undergone human tumour tissue xenografts and in which apoptosis had been induced by the monoclonal antibody Apomab.34 As well as bearing more relevance to human disease, the mechanism of induction of apoptosis of Apomab is entirely distinct from that in dexamethasone treated thymocytes discussed earlier. Initial experiments mirrored those discussed earlier: following dosing with Apomab probe was administered at specific time points, and after a further 1 h whole tumours were excised and imaged ex vivo. This allowed comparison of image data and quantification of caspase labelling by SDS-PAGE. Similarly to the thymi experiments before, fluorescent signal intensity in whole tumour imaging correlated closely with the quantities of labelled caspase, with legumain labelling remaining steady over the time points studied. Peak caspase activity was found to occur 12 h after administration of Apomab. This represents a direct biochemical monitoring of the kinetics of apoptosis in vivo. To optimise clearance (and thus signal to noise), 12 h after treatment with Apomab probes were administered to mice and tumours imaged in a non-invasive manner over several time points. Interestingly, in this study it was found that the probe bearing the tat-peptide, tAB50-Cy5, whilst producing an overall stronger fluorescence signal, exhibited considerably slower clearance than AB50-Cy5. Uptake into tumours was also shown to be apoptosis specific, with SDS-PAGE confirmation ex vivo to show that caspases were labelled specifically in apoptotic tumour tissues.
Taken together, this work presents a highly elegant development of probes that allow very early non-invasive detection of apoptosis along with direct real time kinetics measurements of protease activity, and these tools will clearly be of considerable value in preclinical and possibly also clinical applications. It is also an excellent example of fine-tuning the specificity and targeting elements of an ABP to achieve a desired objective. Taking advantage of the modular nature of ABPs and the power of solid-phase synthesis technology, the authors were able to eliminate an undesired activity whilst moderating a second off-target activity to act as an internal control for labelling, and achieve superior bioavailability of their probes in vivo. A comparative study by the same lab has recently shown that fluorogenic ABPs can be superior to fluorogenic substrates for the detection of enzyme activity, particularly in an in vivo setting.35 This may seem counterintuitive since ABPs do not benefit from signal amplification due to enzyme turnover in the same way as substrates. However ABPs, which tend to be small and drug-like, benefit from better bioavailability, rapid clearance and prolonged retention at the site of action by virtue of remaining covalently bound to the target enzyme. These results in low background combined with a less diffuse signal that remains strictly dependent on enzyme activity.
We decided to take a direct catalomic approach to identify the protease by its activity in vivo, and the first step therefore was to identify a suitable inhibitor as a starting point for ABP development. A live cell assay for SlpA cleavage was used to screen a small library of known protease inhibitors with diverse activities: by measuring the quantity of processed vs. full-length SlpA in the S-layer of treated C. difficile cultures we were able to identify a handful of compounds capable of weak inhibition of S-layer biogenesis. Of these, the natural product E-64 (Fig. 6) was most promising; this compound is a broad-spectrum cysteine protease inhibitor, and typically acts by covalent modification of the unique active site cysteine.37 The warhead in this case is an epoxysuccinyl moiety, and the specificity element is based on L-leucine and a guanidino motif that resembles the side chain of arginine.
![]() | ||
Fig. 6 Inhibitors and ABPs used to target and identify Cwp84, the protease that mediates S-layer biogenesis in C. difficile. |
Taking the E-64 warhead as our starting point, we re-engineered the specificity element initially to Leu-Arg; this inhibitor recapitulated the activity of E-64, and could be accessed using an efficient solid-phase synthetic strategy. Synthesis of a wide range of analogues varying amino acids at both positions demonstrated that only the first position (Leu in E-64) has a strong influence on S-layer processing (Fig. 6). We therefore designed a first generation ABP that incorporated propargyl glycine as the second amino acid, thus introducing a tag into inhibitors for subsequent labelling by click chemistry. Amongst these ABPs we found that those incorporating Tyr, Phe and Arg exhibited greatly improved inhibition over E-64 in a cell-based assay, whilst others lost all activity (e.g. Ala, Ser, Glu). Interestingly, this specificity correlates with the consensus sequence for SlpA cleavage, strongly suggesting that the inhibitors bind to the enzyme in a manner similar to the substrate.
With a spectrum of ABPs in hand, we developed a protocol for the click labelling step; this proved challenging, as the high salt conditions used for sample preparation from C. difficile cultures interfered with the ligation reaction. Indeed, it is important to note from a practical perspective that bioorthogonal ligation can require significant optimisation when applied to the highly complex mixtures obtained from whole organisms, and there remains scope for development of more robust protocols and reagents. Exploiting a novel dual-label reagent we were able to introduce both a fluorescent dye and a biotin moiety onto target proteins (Fig. 6).38 This relatively new development in labelling chemistry provides sensitive, convenient detection and quantification (using in-gel fluorescence), and an affinity handle (biotin) for chemical proteomics. Using these two-stage ABPs, we demonstrated labelling of a protein that correlated with S-layer inhibition activity, along with a degree of background labelling due to off-target ligation. This target protein appeared to be in the surface layer of the bacterium, and we therefore switched to membrane-impermeable ABPs directly-labelled with biotin. These probes enabled very specific labelling of the target protease, and its subsequent isolation and identification by proteomics as the putative cysteine protease Cwp84. Interestingly, we also identified components of a putative processing complex that co-purified with Cwp84 and an ABP that appears to intercept multiple isoforms of the protease, suggesting a complex maturation pathway in vivo.
The work described above is one of a growing number of examples of how ABPs can assist in the dissection of a complex biological process in an organism that is decidedly difficult to manipulate using the tools of molecular biology. Faced with essentially a ‘black box’ in which only the final processed S-layer proteins were open to direct analysis, the authors sought to prove the hypothesis that the S-layer precursor is cleaved by a single specific protease at the cleavage site, as defined by proteomics studies. This is a common challenge in biology: the phenotype (in this case, S-layer assembly) is known, but the mediator (here, a protease) is unknown. The classical genetic approach would generate knockout strains of every gene (or putative protease gene) in the genome, and observe whether cleavage still occurs. However, this approach is practicable only in well-studied model organisms with elaborate tools for genetic engineering, and can be particularly difficult where multiple proteins are involved or where a process is essential, making a knockout strain non-viable. In such cases chemical inhibitors and ABPs are uniquely powerful, providing information on enzyme activity that is not available by means of genetic manipulation.
Ultimately, the ABP approach enabled us to identify a C. difficile cysteine protease (Cwp84) de novo by its catalytic activity, define its localisation in the S-layer and verify conservation of its activity across multiple C. difficile strains. Using an S-layer processing system reconstituted in E. coli we demonstrated on-target processing of SlpA by Cwp84 and showed that specificity is dictated largely by the P2 residue of the substrate, which is also likely to be the binding site for the ABPs. Furthermore, this project led to the discovery and optimisation of the first inhibitors of S-layer biogenesis, with EC50 values in the low-μM range. Finally, labour-intensive genetic manipulation of C. difficile has led to a Cwp84 knockout strain that proves unequivocally that Cwp84 is the only protease responsible for the cleavage of SlpA into the major S-layer components.39
ABPs for hydrolases involved in the virulence of pathogenic microbes also continue to be the subject of intensive study. In addition to the example from C. difficile described above,13 recent reports from the Sieber and Vocadlo labs describe the first such probes for a range of lactamases in MRSA49 and glucosaminidases in Vibrio cholera,50 respectively. The Bogyo and Boothroyd labs have recently reported a remarkably simple ABP based on an irreversible phospholipase inhibitor, 4-bromo phenacyl bromide, which in combination with knockout strains has been used to highlight a large number of enzymes involved in invasion by the apicomplexan pathogen Toxoplasma gondii.51 The interested reader is referred to an excellent recent review by Puri and Bogyo covering ABP developments in the microbial pathogenesis field.52
A very unusual example of a ‘structure-based’ probe was recently disclosed by Kelly et al.62 In this instance, a stilbene analogue was reported to modify a single lysine side chain (amongst eight) of a single protein, transthyretin, against a background of >4000 other proteins present in blood plasma. Although this labelling does not appear to rely on enzymatic activity, exploiting the unique chemistry of a single residue in a protein has strong parallels to ABP and AfBP technologies.
The coming years will see the extension of ABP technology to a growing range of enzyme classes, and the multiplexing of multiple probe types to analyse several activities in one experiment. In particular, due to their importance in signalling and gene regulation, we are likely to see many more ABPs directed against kinases67 and transferases, as well as against their corresponding hydrolases (e.g. phosphatases44 glycosidases,68 and thioesterases69). For example, the elegant bioorthogonal tools recently developed for the identification of transferase substrates would be well-complemented by ABPs directed against the corresponding enzymes.70,71 A range of powerful recombinant and small molecule probes exist for the specific and general detection of enzyme activity (without enzyme labelling),72 and their use in concert with ABPs may prove to be a powerful combination. There are also opportunities for the tighter coupling of ABPs with substrate identification, and recent reports have described the first steps towards this goal.73 Addressing the challenge of enzyme activity associated with sub-cellular localisation or specific complex formation is a frontier in catalomics that will demand greater refinement of current technology, probably in step with the development of novel allosteric inhibitors and in vivo labelling strategies.
The future of catalomics will depend in large part on the capacity of bioorganic chemists to innovate in probe design and the optimisation and application of probes in living systems. Whether this is achieved by interdisciplinary collaboration or by multidisciplinary labs, organic chemistry is set to make a decisive contribution to the exploration of biocatalysis, and to our ability to comprehend and manipulate living systems.
This journal is © The Royal Society of Chemistry 2011 |