Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


It has been estimated that 650,000 protein-protein interactions exist in the human interactome (Stumpf et al., 2008), a subset of all possible macromolecular partnerships that dictate life. Thus there is a continued need for the development of sensitive and user-friendly methods for cataloguing biomacromolecules in complex environments and for detecting their interactions, modifications, and cellular location. Such methods also allow for establishing differences in the interactome between a normal and diseased cellular state and for quantifying the outcome of therapeutic intervention. A promising approach for deconvoluting the role of macromolecular partnerships is split-protein reassembly, also called protein fragment complementation. This approach relies on the appropriate fragmentation of protein reporters, such as the green fluorescent protein or firefly luciferase, which when attached to possible interacting partners can reassemble and regain function, thereby confirming the partnership. Split-protein methods have been effectively utilized for detecting protein-protein interactions in cell-free systems, Escherichia coli, yeast, mammalian cells, plants, and live animals. Herein, we present recent advances in engineering split-protein systems that allow for the rapid detection of ternary protein complexes, small molecule inhibitors, as well as a variety of macromolecules including nucleic acids, poly(ADP) ribose, and iron sulfur clusters. We also present advances that combine split-protein systems with chemical inducers of dimerization strategies that allow for regulating the activity of orthogonal split-proteases as well as aid in identifying enzyme inhibitors. Finally, we discuss autoinhibition strategies leading to turn-on sensors as well as future directions in split-protein methodology including possible therapeutic approaches.

Free full text 


Logo of nihpaLink to Publisher's site
Curr Opin Chem Biol. Author manuscript; available in PMC 2012 Dec 1.
Published in final edited form as:
PMCID: PMC3237955
NIHMSID: NIHMS333671
PMID: 22070901

Split-Protein Systems: Beyond Binary Protein-Protein Interactions

Abstract

It has been estimated that 650,000 protein-protein interactions exist in the human interactome [1], a subset of all possible macromolecular partnerships that dictate life. Thus there is a continued need for the development of sensitive and user-friendly methods for cataloguing biomacromolecules in complex environments and for detecting their interactions, modifications, and cellular location. Such methods also allow for establishing differences in the interactome between a normal and diseased cellular state and for quantifying the outcome of therapeutic intervention. A promising approach for deconvoluting the role of macromolecular partnerships is split-protein reassembly, also called protein fragment complementation. This approach relies on the appropriate fragmentation of protein reporters, such as the green fluorescent protein or firefly luciferase, which when attached to possible interacting partners can reassemble and regain function, thereby confirming the partnership. Split-protein methods have been effectively utilized for detecting protein-protein interactions in cell-free systems, E. coli, yeast, mammalian cells, plants, and live animals. Herein, we present recent advances in engineering split-protein systems that allow for the rapid detection of ternary protein complexes, small molecule inhibitors, as well as a variety of macromolecules including nucleic acids, poly(ADP) ribose, and iron sulfur clusters. We also present advances that combine split-protein systems with chemical inducers of dimerization strategies that allow for regulating the activity of orthogonal split-proteases as well as aid in identifying enzyme inhibitors. Finally, we discuss autoinhibition strategies leading to turn-on sensors as well as future directions in split-protein methodology including possible therapeutic approaches.

Introduction

Methods for discovering as well as investigating macromolecular interactions in a complex milieu have seen tremendous progress. Two and three-hybrid approaches [2, 3] as well as biomolecular fluorescence resonance energy transfer (FRET)[4] are widely used to study protein-protein interactions, both qualitatively and quantitatively. More recently split-protein reassembly methods have emerged as a potentially simple and general solution for studying protein-protein interactions as well as a host of other macromolecules. The observation that fragments of proteins can reassemble into functional complexes was observed over 60 years ago in the context of ribonuclease and subsequently beta-galactosidase [5,6]. However, conditional split-protein reassembly was put on the map in 1994 by the seminal discovery by Johnsson and Varshavsky that appropriate fragments of a monomeric protein, ubiquitin, could be conditionally reassembled when attached to interacting protein pairs that raise the local concentration of the protein fragments [7]. Split-ubiquitin was a major technological leap as it provided a direct method to link the non-covalent interaction of two proteins to the function of the split-reporter protein. This method obviates the requirement for nuclear localization of the interacting proteins or the host translational machinery as in the widely used yeast two-hybrid systems. For the successful creation of a split-reporter protein, a few criteria must be met. Each protein fragment by itself should not exhibit any activity, the affinity of the fragments in the absence of attached interacting proteins should be negligible, and ideally the reassembled split-protein must provide an easily measurable read out (Figure 1). In principle, most proteins can be dissected into such fragments, however despite its deceptive simplicity the identification of appropriate dissection sites within a protein that satisfy the criterion for use in split-protein assays is limited. A case in point is the dissection of firefly luciferase utilizing incremental truncation [8], wherein, the optimal protein fragments possess an 18 amino acid overlap, which would be difficult to anticipate by rational design. Split-protein pairs that are now widely used include those derived from ubiquitin [7], GFP and its variants [9, 10], dihydofolate reductase (DHFR) [11], ß-lactamase [12], firefly [13] and other luciferases [14, 15]. More recently, the tobacco etch virus (TEV) protease [16], thymidine kinase [17] and chorismate mutase[18] have also been co-opted. Currently split-reporter proteins offer a variety of readouts and can detect transient as well as irreversible protein-protein assemblies. Split protein reassembly methods have also been used in directed evolution experiments to discover new as well as improving existing protein partnerships [19-23]. Herein, we will focus primarily upon recent advances that go beyond binary protein-protein interactions.

An external file that holds a picture, illustration, etc.
Object name is nihms-333671-f0001.jpg

Illustration of conditional split-protein reassembly. A generic split-protein system is shown where a functional protein is dissected into two inactive fragments, purple and yellow. The attachment of two interacting proteins or protein domains brings the inactive fragments into close proximity and overcomes the entropic cost of fragmentation. This leads to the reassembly or complementation of the fragments thus providing a direct readout for the partnership between the interacting domains. Crystal structures of representative proteins which have been shown to be amenable to interaction dependent reassembly, where the N-terminal and C-terminal fragments are shown as purple and yellow respectively.

Imaging Protein-Protein Interactions

Spit-protein reassembly has established itself as a valuable tool to image and probe dynamic cellular processes. Most existing fluorescent and luminescent proteins have been co-opted to create split-reporters and have been extensively applied for imaging a variety of interactions, which has been recently reviewed [24,25]. It is worth noting that the irreversible nature of the reassembled GFP-like fluorescent proteins [26] limits their ability to report on temporal information, although they are useful for trapping low affinity interactions [27]. The irreversibility of the split-GFP system has been cleverly used for improving the thermodynamic stability of a variant of the B1 domain of protein G by directed evolution [28]. Many of the split-luciferase reporters on the other hand have been shown to be reversible [29] and offer advantages such as low background, as no exogenous light is required. However, increasing light output and substrate availability inside cells or tissues is an ongoing challenge. In order to expand the scope of split-luminescent reporters much like the formidable palette of fluorescent proteins, Ozawa and co-workers have described multicolor luciferases [30] with improved sensitivity. To go beyond absorbance, fluorescence, and luminescence based modalities, Gambhir and co-workers have recently utilized Positron Emission Tomography (PET) based imaging as their readout for protein-protein interactions [17]. A new split-thymidine kinase was discovered that when activated, phosphorylates (9-(4-[18F]-fluoro-3-hydroxymethylbutyl)-guanine) and localizes it to only those cells expressing reassembled split-thymidine kinase. This approach couples a sensitive radioactivity based imaging technique, such as PET, with split protein reassembly and can potentially provide a new approach for clinical imaging of protein-protein interactions in live animals. As a proof of principle, split-protein dependent PET was used to monitor the complexation of hypoxia-inducible factor-1α with the von Hippel-Lindau tumor suppressor protein.

Recently, split-protein approaches have been paired with resonance energy methods, wherein a reconstituted split fluorescent protein can serve as an acceptor for resonance energy transfer from a fluorescent (FRET) or bioluminescent protein (BRET). Indeed, such a strategy was utilized to demonstrate the association of four different proteins as part of a G protein-coupled receptor signaling complex [31]. Taking advantage of this strategy, Bouvier and co-workers discovered an unexpected asymmetric organization of the calcitonin gene-related peptide (CGRP) receptor [32]. In a FRET-based strategy, Hu and co-workers visualized ternary complexes implicated in transcription, wherein the fluorescent protein cerulean fused to NFAT1, NF-kB and p65 allowed energy transfer to a fos-jun mediated reassembled split-venus complex [33].

Interrogating Small Molecule Inhibitors

Split-protein reassembly naturally lends itself for discovering and evaluating small molecule inhibitors, especially those that directly disrupt protein-protein interactions. Recently a cell-free split-luciferase enabled approach was established (Figure 2a) which allowed for the rapid interrogation of a large number of macromolecular interactions [29]. This methodology was used to test the small molecule specificity for a set of therapeutically relevant helix/receptor pairs. Eighteen helix/receptor pairs were first examined for specificity, which recapitulated the reported affinity of these helix/receptor pairs [34]. Further investigation of inhibition profiles of some well known inhibitors of these helix/receptor interactions, led to some surprising results. For example, significant inhibition of the p53/hDM2 interaction was also observed for a previously identified Bcl-2 family inhibitor, BH3I-3. In the near future, we expect that large protein-protein interaction (PPI) arrays using split-protein reporters will be available for interrogating new classes of inhibitors. Such methods would aid in rapidly establish the desired selectivity or promiscuity for PPI inhibitors as has been the case with kinase inhibitors. Besides probing the direct effect of small molecules on a specific protein-protein interaction, split-protein reassembly has also been used to interrogate the effect of small molecules on specific biochemical pathways, allowing for the delineation of potential off-target effects of drug candidates. In a detailed study, a cell based split-Venus system was used to carry out pharmacological profiling of a large array of downstream protein-protein interactions, that were known to report upon specific cellular pathways ranging from cell-cycle control to apoptosis [35]. In this screen, 107 known drugs representing many therapeutic areas were interrogated in tandem. Strikingly, four existing drugs were found to possess previously unknown anti-tumor activity. Thus the ease of implementation of the split-protein approach has the potential to rapidly uncover potentially promiscuous molecules, whether desired or undesired, as well as identify the off-target effects of drug targets in a pathway specific manner.

An external file that holds a picture, illustration, etc.
Object name is nihms-333671-f0002.jpg

Detection of macromolecules using split-protein reassembly via ternary complexation. a) Schematics of split-protein translation in cell-free translation systems such as rabbit reticulocyte lysate or wheat germ extract, where interacting domains of interest can be rapidly produced and interrogated. b) Detection of unmodified native proteins via ternary complexation using receptor or single-chain antibody fragments attached to split-luciferase. c) DNA and DNA modifications, such as methylation, can be detected using appropriate fusions of split luciferase and user defined DNA binding domains. The genome can be scanned at all CpG methylated sites for specific types of modifications resulting from DNA damage. d) A quaternary strategy for detection of RNA utilizing RNA detection domains such as argonaute and pumilio domains. The pentameric assembly allows targeting any ssRNA or ssDNA utilizing a zinc finger binding hairpin guide sequence. e) Co-factors such as iron-sulfur clusters detected via split-venus and apoglutaredoxin fusion pairs that dimerize upon binding 2Fe2S. e) Monitoring the dynamics of poly-ADP Ribose (PAR) levels subsequent to DNA damage through attached PAR targeted APLF domains.

Ternary Complexation: Native Protein and Nucleic Acid Dependent Split-protein Reassembly

In split-protein systems, the proteins pairs being examined are attached to the fragmented reporters. Thus a question arises as to how may one detect an unmodified native target, whether protein, DNA, or RNA. A simple solution is to identify two protein domains that interact with the desired target, resulting in a ternary complex that drives the reassembly of the split-protein reporter (Figure 2b). Towards this goal, the split halves of firefly luciferase were fused to receptor fragments or single-chain antibodies [36]. The conditional reassembly of luciferase halves was induced in presence of several biologically relevant targets such as vascular endothelial growth factor (VEGF), and the human epidermal growth factor receptor-2 (HER2). Significantly, this ternary complexation method allows for rapid determination of the protein levels of cell-surface markers such as HER2 without any additional purification or handling steps.

The ternary split-protein reassembly strategy naturally lends itself to a convenient method for the sequence-specific detection of nucleic acids, particularly DNA. For example, designed and natural Cys2-His2 Zinc finger DNA binding domains have been fused to split fragments of a reporter protein such as GFP, ß-lactamase or firefly luciferase (Figure 2c). In the presence of target DNA, harboring specific adjacent DNA sites recognized by the zinc finger domains, the reporter protein activity is reconstituted providing an easily measurable “turn-on” fluorescent [37] colorimetric [38] or luminescent [29] signal. The modular nature of this system, allows for the detection of virtually any nucleic acid sequence. This can be achieved by replacing the zinc finger binding domains with alternate detection domains that target specific nucleic acid modifications. For instance, by utilizing a methyl binding domain (MBD) and Zif268, a natural zinc finger, DNA methylation can be detected in a sequence-specific manner [39]. More recently, turn-on sensors for the detection of UV or oxidation-dependent DNA damage has also been reported [40]. In this approach, the MBD domain can scan a large fraction of the genome (50-80% of the genome is methylated at CpG sites) while a damage specific protein domain can readout specific DNA modifications, such as 8-oxoguanine or pyrimidine dimer photoproducts (Figure 2c). Similarly, bipartite MBD domains have been used for measuring the activity of DNA methyltransferase inhibitors in live cells [41].

Unlike zinc fingers and the recently discovered TAL-enhancers that recognize DNA, there are fewer avenues for co-opting sequence specific RNA recognition domains. Rackham and Brown utilized known RNA binding proteins tethered to a split-venus sensor to visualize ternary RNA-protein complexes in live mammalian cells [42]. More recently, several studies have now shown that natural and designed pumilio domains can be utilized for targeting specific RNA sequences, which may ultimately provide an inroad to targeting any RNA sequence of interest [43]. Towards the long-term goal of developing a general approach, for targeting RNA, a quaternary assembly strategy was examined, wherein RNA-binding Argonaut domains, which bind 2 nucleotide (nt) 3′ overhangs of short double stranded RNA, were fused to split-firefly luciferase [44]. The ternary assembly of the split-luciferase was then induced by Argonaut mediated recognition of nucleotide overhangs, facilitated by complementary ssRNA guide sequences. A pentameric assembly was also investigated, where designed zinc finger domains were appended to split-luciferase halves, such that a zinc finger binding hairpin containing guide sequence could target any ssRNA or ssDNA target (Figure 2d). In a similar approach, split-GFP has been directly conjugated to guide ssDNA oligonucleotides either covalently [45] or through biotin/streptavidin interactions resulting in DNA hybridization dependent GFP reassembly [46].

More recently, ternary split-protein reassembly has also been applied for the detection of other biologically relevant molecules. For example, in a clever ternary complexation strategy, Hoff and co-workers [47] developed a split-GFP based turn-on sensor for the detection of iron-sulfur clusters (2Fe-2S). Each of the fragment of split-Venus was appended to glutaredoxin 2 (GRX2), a glutathione dependent oxidoreductase. ApoGRX2 is monomeric, however, in the presence of 2Fe2S clusters, GRX2 dimerizes, bringing Venus fragments into close proximity, leading to complementation. Thus this approach allowed for imaging the biogenesis of a 2Fe2S cluster in live cells (Figure 2e). In another example, a poly-ADP ribose (PAR) binding protein was fused to both halves of split-luciferase and thus allowed for the detection of PAR [48]. This turn-on PAR sensor (Figure 2f) allowed for monitoring the dynamic change in PAR levels in several mammalian cell lines exposed to DNA-damaging agents.

Chemical Inducers of Dimerization (CID)

A desired goal of chemical biology is the conditional control of protein function with small molecules from both a therapeutic stand point as well as for delineating complex biological pathways. Since the seminal discovery of the small molecule rapamycin-dependent heterodimerization of FKBP12 (FK506 binding protein) with FRB (FKBP12 rapamycin binding protein), numerous applications have exploited the system for conditional control of cellular events [49, 50]. Not surprisingly, many split-protein reporters were initially validated using the FKBP/FRB CID system including ß-lactamase [12], firefly luciferase [13] and gaussia luciferase [14]. Small molecule-dependent protein fragment complementation is a particularly attractive technique, as it allows dose-dependent control of a functional protein inside a cell. For example, building upon the split-ubiqutin system [7], Muir and co-workers have recently described a clever strategy for controlling protein degradation in a CID-dependent fashion [51]. In this approach, a protein of interest is fused to one half of split-ubiquitin and FRB followed by a degron which tags the protein for destruction by the proteasome. The other half of split-ubiquitin is fused to FRBP, and upon addition of rapamycin, reconstituted ubiquitin rescues the protein of interest from degradation and thereby allows small molecule control of protein half-lives.

A split-TEV system was developed to monitor protein-protein interactions both at the membrane and in the cytosol of mammalian cells [16, 52]. More recently, Wells and co-workers engineered a conditional FKBP/FRB CID system using split-TEV [53] with decreased affinity of split fragments to identify and characterize substrates of specific caspases in a cellular context. In an elegant design, the authors generated TEV cleavage dependent caspase alleles, which could be turned on by activating CID dependent split-TEV (Figure 3a). Importantly, this approach led to the discovery of synergy between caspase activation and proteasome inhibition, suggesting negative regulation of proteasome activity by caspases during apoptosis, an observation which has therapeutic implications for the treatment of cancer.

An external file that holds a picture, illustration, etc.
Object name is nihms-333671-f0003.jpg

New strategies for split-protein systems. a) Selective activation of caspases incorporating TEV cleavage sites using a small molecule rapamycin triggered reassembly of split-TEV protease. b) Screening kinase inhibitors utilizing a three-hybrid strategy using a new peptide-small molecule chemical inducer of dimerization wherein a potential kinase inhibitor can be identified by split-luciferase disassembly. c) An autoinhibited coiled-coil split-luciferase turn-on protease sensor wherein the autoinhibition is relieved in presence of any protease of interest leading to split-luciferase reassembly. d) An autoinhibited strategy for detection of small molecules using supramolecular building blocks.

Beyond the classic FKBP/FRB CID system, a three-hybrid system has been reported wherein split-ubiquitin fragments were fused to glucocorticoid receptor (GR) and dihydrofolate reductase (DHFR) [54]. The reassembly of split-ubiquitin was conditionally induced in live cells by a bifunctional molecule, comprised of a classic dexamethasone and methotrexate, which bind GR and DHFR respectively. In a recent report a three-hybrid strategy for high-throughput profiling of protein kinase inhibitors was also reported using a new CID [55]. In this strategy the C-terminal half of split-firefly luciferase is fused to a protein kinase of interest while the N-terminal half of luciferase is attached to coiled coil Fos. The reassembly of split-firefly luciferase is then induced by the new CID, the peptide Jun covalently attached to staurosporine, a pan-kinase inhibitor (Figure 3b). Kinase inhibitors were rapidly identified by disrupting the staurosporine-kinase interaction with concomitant loss of firefly luciferase activity. The cell-free method was shown to be general across the kinome and also allowed for facile discrimination between competitive and allosteric kinase inhibitors.

Autoinhibited Turn-On Sensor Systems

The autoinhibition of protein activity is widely used in nature and also affords a design template for engineering split-protein sensors. A turn-on protease sensor has been designed (Figure 3c), where the reassembly of split reporter is auto-inhibited as the coiled-coils interact intramolecularly due to favorable entropy, however, upon protease mediated cleavage of the linker the two fragments can reassemble [56]. This method is tunable by modifying either the coiled-coils or protease cleavage site to monitor any protease of interest and afforded a complete panel of caspase sensors for determining caspase crosstalk [57]. This approach was also shown to serve as a platform for the construction of protein logic gates [56]. In another example of an autoinhibited system, host-guest interactions between ß-cyclodextrin and coumarin units were utilized to exercise supramolecular control over reassembly of split-GFP (Figure 3d). By incorporating both the ß-cyclodextrin and coumarin units in the same C-terminal GFP peptide fragment, the resulting intramolecular complex was autoinhibited and unable to bind the N-terminal GFP fragment [58]. The autoinhibition could be relieved by addition of an exogenous hydrophobic guest molecule with concomitant reassembly of the GFP fragments, which in effect creates a turn-on sensor for small molecules. Another self assembling autoinhibited split-GFP has also been described, wherein the autoinhibition is relieved by the action of a protease of interest instead of a small molecule [58]. In this approach, the C-terminal GFP fragment was incorporated as a loop in another protein with a protease cleavable linker, where the self assembly of split-GFP is inhibited until cleavage by a protease of interest. These autoinhibitory approaches marry biological as well as supramolecular building blocks to split-protein reporter outputs.

Outlook and Conclusion

The range of biological activities and processes that can be monitored with a variety of readouts using split-protein reassembly both in an in vitro and in vivo setting is unprecedented. There has been a veritable explosion of applications utilizing split-protein reassembly and the long-term potential of this methodology is only beginning to be realized, as the existing split-protein outputs are being combined in a variety of ways to user-defined inputs under examination. We have already seen the ability to probe binary as well as ternary partnerships and the rapid interrogation of small molecules that perturb them. Certain technical milestones would help advance the field, such as the discovery of new split-fluorescent proteins that reversibly assembles and dissembles, or the discovery of split-luminescent proteins and substrates which allow for imaging in deep tissue. We have also witnessed that the ever expanding repertoire of split-proteins at the disposal of the chemical biologist provides a means for the temporal control over the outcome of various biomolecular processes. This has already enabled the discovery of new biology, such as the synergy between caspase activation and proteasome inhibition. In the not too distant future, we can envision the explicit control over the outcome of specific signal transduction pathways by utilizing fragmented protein kinases, phosphatases, acetylases or ligases engineered to be temporally controlled by a small molecule CID. Finally, we anticipate that the general split-protein ternary complexation approach may allow for the conditional reassembly of a split toxic protein inside a particular cell type, that responds to specific over-expressed proteins, nucleic acids, and other macromolecules and thereby provide new therapeutic and imaging modalities.

Highlights

  • Advances in split -proteins using PET, FRET and BRET strategies

  • Interrogating small molecule inhibitors

  • Detection of native proteins, DNA, RNA, Fe-S clusters, PAR and small molecules

  • Chemical inducers of dimerization strategies to control split-protein activity

  • Auto-inhibited and supramolecular architectures for split-protein sensors

Acknowledgements

We thank past and present members of the Ghosh laboratory particularly Ben Jester and Jenny Furman for their thoughtful contributions. I.G. acknowledges support from the NSF (CHE-0548264) and NIH (CA141974 and CA143661).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

*of special interest

**of outstanding interest

1. Stumpf MP, Thorne T, de Silva E, Stewart R, An HJ, Lappe M, Wiuf C. Estimating the size of the human interactome. Proc Natl Acad Sci U S A. 2008;105:6959–6964. [Europe PMC free article] [Abstract] [Google Scholar]
2. Fields S, Song O. A novel genetic system to detect protein-protein interactions. Nature. 1989;340:245–246. [Abstract] [Google Scholar]
3. Licitra EJ, Liu JO. A three-hybrid system for detecting small ligand-protein receptor interactions. Proc Nat Acad Sci USA. 93(23):12817–12821. [Europe PMC free article] [Abstract] [Google Scholar]
4. Zhang J, Campbell RE, Ting A, Tsien RY. Creating new fluorescent probes for cell biology. Nat. Rev. Mol. Bio. 2002;3:906–918. [Abstract] [Google Scholar]
5. Richards FM. On the enzymic activity of subtilisin-modified ribonuclease. Proc Nat Acad Sci USA. 1958;44:162–166. [Europe PMC free article] [Abstract] [Google Scholar]
6. Ullmann A, Jacob F, Monod J. Characterization by in vitro complementation of a peptide corresponding to an operator-proximal segment of beta-galactosidase structural gene of Escherichia coli. J. Mol. Bio. 1967;24(2):339–343. [Abstract] [Google Scholar]
7. Johnsson N, Varshavsky A. Split ubiquitin as a sensor of protein interactions in vivo. Proc Natl Acad Sci U S A. 1994;91:10340–13044. [Europe PMC free article] [Abstract] [Google Scholar]
8. Ostermeier M, Nixon AE, Shim JH, Benkovic SJ. Combinatorial protein engineering by incremental truncation. Proc Nat Acad Sci USA. 1999;96:3562–3567. [Europe PMC free article] [Abstract] [Google Scholar]
9. Ghosh I, Hamilton AD, Regan L. Antiparallel leucine zipper-directed protein reassembly: Application to the green fluorescent protein. J. Am. Chem. Soc. 2000;122:5658–5659. [Google Scholar]
10. Hu CD, Kerppola TK. Simultaneous visualization of multiple protein interactions in living cells using multicolor fluorescence complementation analysis. Nat. Biotechnol. 2003;21:539–1545. [Europe PMC free article] [Abstract] [Google Scholar]
11. Pelletier JN, Campbell-Valois FX, Michnick S. Oligomerization domain-directed reassembly of active dihydrofolate reductase from rationally designed fragments. Proc Natl Acad Sci U S A. 1998;95:12141–12146. [Europe PMC free article] [Abstract] [Google Scholar]
12. Galarneau A, Primeau M, Trudeau LE, Michnick SW. beta-Lactamase protein fragment complementation assays as in vivo and in vitro sensors of protein-protein interactions. Nat. Biotech. 2002;20(6):619–622. [Abstract] [Google Scholar]
13. Luker KE, Smith MC, Luker GD, Gammon ST, Piwnica-Worms H, Piwnica-Worms D. Kinetics of regulated protein-protein interactions revealed with firefly luciferase complementation imaging in cells and living animals. Proc Natl Acad Sci U S A. 2004;101:12288–122893. [Europe PMC free article] [Abstract] [Google Scholar]
14. Remy I, Michnick SW. A highly sensitive protein-protein interaction assay based on Gaussia luciferase. Nat. Meth. 2006;3(12):977–979. [Abstract] [Google Scholar]
15. Paulmurugan R, Gambhir SS. Monitoring protein-protein interactions using split synthetic renilla luciferase protein-fragment-assisted complementation. Ana.Chem. 2003;759(7):1584–1589. [Europe PMC free article] [Abstract] [Google Scholar]
16. Wehr MC, Laage R, Bolz U, Fischer TM, Grunewald S, Scheek S, Bach A, Nave KA, Rossner MJ. Monitoring regulated protein-protein interactions using split TEV. Nat. Meth. 2006;3(12):985–993. [Abstract] [Google Scholar]
17. ** Massoud TF, Paulmurugan R, Gambhir SS. A molecularly engineered split reporter for imaging protein-protein interactions with positron emission tomography. Nat. Med. 2010;16(8):921–927. [Europe PMC free article] [Abstract] [Google Scholar] This study discloses the design of a split-thymidine kinase, which provides the first example of a strategy to detect protein-protein interactions using Positron Emission Tomography. This approach provides a new method for visualizing protein partnerships in live animals.
18. Muller MM, Kries H, Csuhai E, Kast P, Hilvert D. Design, selection, and characterization of a split chorismate mutase. Prot. Sci. 2010;19(5):1000–1010. [Europe PMC free article] [Abstract] [Google Scholar]
19. Pelletier JN, Arndt KM, Pluckthun A, Michnick SW. An in vivo library-versus-library selection of optimized protein-protein interactions. Nat. Biotech. 1999;17(7):683–690. [Abstract] [Google Scholar]
20. Amstutz P, Koch H, Binz HK, Deuber SA, Pluckthun A. Rapid selection of specific MAP kinase-binders from designed ankyrin repeat protein libraries. Prot. Eng. Des. Sel. 2006;19(5):219–229. [Abstract] [Google Scholar]
21. Mossner E, Koch H, Pluckthun A. Fast selection of antibodies without antigen purification: Adaptation of the protein fragment complementation assay to select antigen-antibody pairs. J. Mol. Biol. 2001;308(2):115–122. [Abstract] [Google Scholar]
22. Campbell-Valois FX, Tarassov K, Michnick SW. Massive sequence perturbation of a small protein. Proc Natl Acad Sci U S A. 2005;102(42):14988–14993. [Europe PMC free article] [Abstract] [Google Scholar]
23. * Jackrel ME, Cortajarena AL, Liu TY, Regan L. Screening Libraries To Identify Proteins with Desired Binding Activities Using a Split-GFP Reassembly Assay. ACS Chem. Biol. 2010;5(6):553–562. [Abstract] [Google Scholar] This study successfully uses the split-GFP screen for discovering new protein-protein partnerships.
24. Michnick SW, Ear PH, Manderson EN, Remy I, Stefan E. Universal strategies in research and drug discovery based on protein-fragment complementation assays. Nat. Rev. Drug Disc. 2007;6(7):569–582. [Abstract] [Google Scholar]
25. Muller J, Johnsson N. Split-ubiquitin and the split-protein sensors: Chessman for the endgame. Chembiochem. 2008;9(13):2029–2038. [Abstract] [Google Scholar]
26. Magliery TJ, Wilson CGM, Pan WL, Mishler D, Ghosh I, Hamilton AD, Regan L. Detecting protein-protein interactions with a green fluorescent protein fragment reassembly trap: Scope and mechanism. J. Am. Chem. Soc. 2005;127(1):146–157. [Abstract] [Google Scholar]
27. Nyfeler B, Michnick SW, Hauri HP. Capturing protein interactions in the secretory pathway of living cells. Proc Natl Acad Sci U S A. 2005;102(18):6350–6355. [Abstract] [Google Scholar]
28. * Lindman S, Hernandez-Garcia A, Szczepankiewicz O, Frohm B, Linse S. In vivo protein stabilization based on fragment complementation and a split GFP system. Proc Natl Acad Sci U S A. 2010;107(46):19826–19831. [Europe PMC free article] [Abstract] [Google Scholar] This study provides a clever approach for using the split-GFP system to specifically interrogate interactions with weak affinities but to utilize the weak interactions for identifying small thermostable proteins.
29. ** Porter JR, Stains CI, Jester BW, Ghosh I. A general and rapid cell-free approach for the interrogation of protein-protein, protein-DNA, and protein-RNA interactions and their antagonists utilizing split-protein reporters. J. Am. Chem. Soc. 2008;130(20):6488–6497. [Abstract] [Google Scholar] This study provides a cell-free method for rapidly using a variety of split-protein systems to test a large number of macromolecular interactions that include protein-protein, protein-DNA, protein-RNA, as well as small molecule dimerizers. This study also demonstrates that inhibitors of almost any macromolecular interaction can be rapidly probed and it also provides proof of principle experiments for using split-proteins in artificial cell systems comprising water-in-oil emulsions.
30. Hida N, Awais M, Takeuchi M, Ueno N, Tashiro M, Takagi C, Singh T, Hayashi M, Ohmiya Y, Ozawa T. High-Sensitivity Real-Time Imaging of Dual Protein-Protein Interactions in Living Subjects Using Multicolor Luciferases. Plos One. 2009;4(6):e5868. [Europe PMC free article] [Abstract] [Google Scholar]
31. * Dupre DJ, Robitaille M, Ethier N, Villeneuve LR, Mamarbachi AM, Hebert TE. Seven Transmembrane Receptor Core Signaling Complexes Are Assembled Prior to Plasma Membrane Trafficking. J. Bio. Chem. 2006;281(45):34561–34573. [Abstract] [Google Scholar] This study allows for interrogating ternary complexes by combining split-protein reassembly that detects a pair of proteins with bioluminescence resonance energy transfer (BRET) where a third protein is attached to luciferase. This allows for reporting upon ternary partnerships.
32. Heroux M, Hogue M, Lemieux S, Bouvier M. Functional Calcitonin Gene-related Peptide Receptors Are Formed by the Asymmetric Assembly of a Calcitonin Receptor-like Receptor Homo-oligomer and a Monomer of Receptor Activity-modifying Protein-1. J. Bio. Chem. 2007;282(43):31610–31620. [Abstract] [Google Scholar]
33. * Shyu YJ, Suarez CD, Hu CD. Visualization of AP-1-NF-kappa B ternary complexes in living cells by using a BiFC-based FRET. Proc Natl Acad Sci U S A. 2008;105(1):151–156. [Europe PMC free article] [Abstract] [Google Scholar] This study allows for interrogating ternary complexes by combining split-protein reassembly that detects two proteins with fluorescence resonance energy transfer (FRET) through a third protein that allows for reporting upon a ternary partnership.
34. Porter JR, Helmers MR, Wang P, Furman JL, Joy ST, Arora PS, Ghosh I. Profiling small molecule inhibitors against helix-receptor interactions: the Bcl-2 family inhibitor BH3I-1 potently inhibits p53/hDM2. Chem. Comm. 2010;46(42):8020–8022. [Europe PMC free article] [Abstract] [Google Scholar]
35. MacDonald ML, Lamerdin J, Owens S, Keon BH, Bilter GK, Shang Z, Huang Z, Yu H, Dias J, Minami T, Michnick SW, Westwick JK. Identifying off-target effects and hidden phenotypes of drugs in human cells. Nat. Chem. Biol. 2006;2:329–337. [Abstract] [Google Scholar]
36. * Stains CI, Furman JL, Porter JR, Rajagopal S, Li YX, Wyatt RT, Ghosh I. A General Approach for Receptor and Antibody-Targeted Detection of Native Proteins Utilizing Split-Luciferase Reassembly. ACS Chem. Biol. 2010;5(10):943–952. [Europe PMC free article] [Abstract] [Google Scholar] This study provides several strategies for detecting any native protein of interest using ternary complexation and lays the groundwork for native protein expression level dependent activation of split-proteins.
37. Stains CI, Porter JR, Ooi A, Segal DJ, Ghosh I. DNA sequence-enabled reassembly of the green fluorescent protein. J. Am. Chem. Soc. 2005;127(31):10782–10783. [Abstract] [Google Scholar]
38. Porter JR, Stains CI, Segal DJ, Ghosh I. Split beta-lactamase sensor for the sequence-specific detection of DNA methylation. Ana. Chem. 2007;79(17):6702–6708. [Abstract] [Google Scholar]
39. Stains CI, Furman JL, Segal DJ, Ghosh I. Site-specific detection of DNA methylation utilizing mCpG-SEER. J. Am. Chem. Soc. 2006;128(30):9761–9765. [Abstract] [Google Scholar]
40. * Furman JL, Mok PW, Shen S, Badran AH, Ghosh I. Turn-On DNA Damage Sensors for the Direct Detection of 8-Oxoguanine and Photoproducts in Native DNA. J. Am. Chem. Soc. 2011;133(32):12518–12527. [Europe PMC free article] [Abstract] [Google Scholar] This study provides a general method for scanning the genome for DNA modifications. In this approach methyl binding domains serve as a genome targeting domain and when paired with DNA modification sensitive proteins, this approach allows for reading out global levels of DNA damage.
41. Badran AH, Furman JL, Ma AS, Comi TJ, Porter JR, Ghosh I. Evaluating the Global CpG Methylation Status of Native DNA Utilizing a Bipartite Split-Luciferase Sensor. Anal. Chem. 2011;83(18):7151–7157. [Europe PMC free article] [Abstract] [Google Scholar]
42. Rackham O, Brown CM. Visualization of RNA-protein interactions in living cells: FMRP and IMP1 interact on mRNAs. EMBO J. 2004;23:3346–55. [Europe PMC free article] [Abstract] [Google Scholar]
43. ** Ozawa T, Natori Y, Sato M, Umezawa Y. Imaging dynamics of endogenous mitochondrial RNA in single living cell. Nat. Methods. 2007;4(5):413–419. [Abstract] [Google Scholar] This study provides the first example of pumilio domain mediated detection of endogenous RNA in live cells. This is significant as the pumilio domain is one of the few modular RNA binding domains where systematic protein engineering raises the possibility for redesigning this domain to target any RNA sequence of interest.
44. Furman JL, Badran AH, Ajulo O, Porter JR, Stains CI, Segal DJ, Ghosh I. Toward a General Approach for RNA-Templated Hierarchical Assembly of Split-Proteins. J. Am. Chem. Soc. 2010;132(33):11692–11701. [Europe PMC free article] [Abstract] [Google Scholar]
45. Takeda S, Tsukiji S, Ueda H, Nagamune T. Covalent split protein fragment-DNA hybrids generated through N-terminus-specific modification of proteins by oligonucleotides. Org. Biomol. Chem. 2008;6(12):2187–2194. [Abstract] [Google Scholar]
46. Demidov VV, Dokholyan NV, Witte-Hoffmann C, Chalasani P, Yiu HW, Ding F, Yu Y, Cantor CR, Broude NE. Fast complementation of split fluorescent protein triggered by DNA hybridization. Proc Natl Acad Sci U S A. 2006;103(7):2052–2056. [Europe PMC free article] [Abstract] [Google Scholar]
47. * Hoff KG, Culler SJ, Nguyen PQ, McGuire RM, Silberg JJ, Smolke CD. In Vivo Fluorescent Detection of Fe-S Clusters Coordinated by Human GRX2. Chemistry & Biology. 2009;16(12):1299–1308. [Abstract] [Google Scholar] This study establishes proof of principle methods for using split-protein assembly for visualizing the biogenesis of an Fe-S cluster in live cells using apo glutaredoxin 2.
48. Furman JL, Mok PW, Shen S, Stains CI, Ghosh I. A turn-on split-luciferase sensor for the direct detection of poly(ADP-ribose) as a marker for DNA repair and cell death. Chem. Comm. 2011;47:397–399. [Abstract] [Google Scholar]
49. Spencer DM, Wandless TJ, Schreiber SL, Crabtree GR. Controlling signal-transduction with synthetic ligands. Science. 1993;262(5136):1019–1024. [Abstract] [Google Scholar]
50. Fegan A, White B, Carlson JCT, Wagner CT. Chemically controlled protein assembly: Techniques and Applications. Chem. Rev. 2010;110:3315–3336. [Abstract] [Google Scholar]
51. * Pratt MR, Schwartz EC, Muir TW. Small molecule-mediated rescue of protein function by an inducible proteolytic shunt. Proc Natl Acad Sci U S A. 2007;104(27):11209–11214. [Europe PMC free article] [Abstract] [Google Scholar] This study provides a redesigned split-ubiquitin and FKBP/FRB system that responds to an enginered degron sequences and thus provides a potentail method for controlling the protein half-life of any protein using small molecules.
52. Williams DJ, Puhl HL, Ikeda SR. Rapid Modification of Proteins Using a Rapamycin-Inducible Tobacco Etch Virus Protease System. PLOS One. 2009;4(10):14. [Europe PMC free article] [Abstract] [Google Scholar]
53. ** Gray DC, Mahrus S, Wells JA. Activation of Specific Apoptotic Caspases with an Engineered Small-Molecule-Activated Protease. Cell. 2010;142(4):637–646. [Europe PMC free article] [Abstract] [Google Scholar] This study describes a FKBP/FRB dependent split-TEV protease that allows for small molecule dependent activation of engineered caspases in living cells. Importantly, this approach led to the discovery of a surprising synergy between caspase activation and proteasome inhibition.
54. Dirnberger D, Unsin G, Schlenker S, Reichel C. A small-molecule-protein interaction system with split-ubiquitin as sensor. Chembiochem. 2006;7(6):936–942. [Abstract] [Google Scholar]
55. ** Jester BW, Cox KJ, Gaj A, Shomin CD, Porter JR, Ghosh I. A Coiled-Coil Enabled Split-Luciferase Three-Hybrid System: Applied Toward Profiling Inhibitors of Protein Kinases. J. Am. Chem. Soc. 2010;132(33):11727–11735. [Europe PMC free article] [Abstract] [Google Scholar] This study describes a new chemical inducer of dimerization linking a small molecule to a coiled-coil peptide that allows for the rapid interrogation of protein kinase-small molecule interactions using split-protein systems. Moreover it allows for identifying small molecule kinase inhibitors as well as true allosteric inhibitors.
56. * Shekhawat SS, Porter JR, Sriprasad A, Ghosh I. An Autoinhibited Coiled-Coil Design Strategy for Split-Protein Protease Sensors. J. Am. Chem. Soc. 2009;131(42):15284–15290. [Europe PMC free article] [Abstract] [Google Scholar] This study provides a tunable autoinhibited coiled-coil design integrated with several split-protein systems that can be turned on upon cleavage of a protease sensitive linker. This method allows for probing the specificity of proteases but also allows for building logic gates.
57. Shekhawat SS, Campbell ST, Ghosh I. A Comprehensive Panel of Turn-On Caspase Biosensors for Investigating Caspase Specificity and Caspase Activation Pathways. ChemBioChem. 10.1002/cbic.201100372. [Abstract] [Google Scholar]
58. * Sakamoto S, Kudo K. Supramolecular control of split-GFP reassembly by conjugation of beta-cyclodextrin and coumarin units. J. Am. Chem. Soc. 2008;130(29):9574–9582. [Abstract] [Google Scholar] This study integrates split-protein systems with classic supramolecular systems to develop a autoinhibited system that can directly respond to small molecules that bind β-cyclodextrin.
59. Callahan BP, Stanger MJ, Belfort M. Protease activation of split green fluorescent protein. ChemBioChem. 2010;11:2259–2263. [Europe PMC free article] [Abstract] [Google Scholar]

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/447441
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/447441

Smart citations by scite.ai
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
Explore citation contexts and check if this article has been supported or disputed.
https://scite.ai/reports/10.1016/j.cbpa.2011.10.014

Supporting
Mentioning
Contrasting
0
159
0

Article citations


Go to all (96) article citations

Funding 


Funders who supported this work.

NCI NIH HHS (8)