NanoBRET—A Novel BRET Platform for the Analysis of Protein–Protein Interactions
- Thomas Machleidt
- ,
- Carolyn C. Woodroofe
- ,
- Marie K. Schwinn
- ,
- Jacqui Méndez
- ,
- Matthew B. Robers
- ,
- Kris Zimmerman
- ,
- Paul Otto
- ,
- Danette L. Daniels
- ,
- Thomas A. Kirkland
- , and
- Keith V. Wood
Abstract

Dynamic interactions between proteins comprise a key mechanism for temporal control of cellular function and thus hold promise for development of novel drug therapies. It remains technically challenging, however, to quantitatively characterize these interactions within the biologically relevant context of living cells. Although, bioluminescence resonance energy transfer (BRET) has often been used for this purpose, its general applicability has been hindered by limited sensitivity and dynamic range. We have addressed this by combining an extremely bright luciferase (Nanoluc) with a means for tagging intracellular proteins with a long-wavelength fluorophore (HaloTag). The small size (19 kDa), high emission intensity, and relatively narrow spectrum (460 nm peak intensity) make Nanoluc luciferase well suited as an energy donor. By selecting an efficient red-emitting fluorophore (635 nm peak intensity) for attachment onto the HaloTag, an overall spectral separation exceeding 175 nm was achieved. This combination of greater light intensity with improved spectral resolution results in substantially increased detection sensitivity and dynamic range over current BRET technologies. Enhanced performance is demonstrated using several established model systems, as well as the ability to image BRET in individual cells. The capabilities are further exhibited in a novel assay developed for analyzing the interactions of bromodomain proteins with chromatin in living cells.
Results and Discussion
Optimization of the Fluorescent Acceptor
Figure 1

Figure 1. Comparison of different acceptors for NanoBRET. (a) BRET spectra for Nluc-HT (black) and Nluc-HT conjugated to HaloTag Oregon Green (green), HaloTag TMR (red), or HaloTag NCT (blue) ligands. (b) Correlation of BRET with fractional occupancy. Differing degrees of occupancy were created by combining Nluc-HT-NCT (fully acceptor occupied) with Nluc-HT-PEG-biotin (unoccupied donor) in defined ratios at a constant donor concentration. Fractional occupancy (%) indicates the amount of acceptor-occupied Nluc-HT relative to the total amount of Nluc-HT in the sample. Shown are the means ± SD (n = 7) of a representative experiment. The results were plotted as mBRET units.
Z′-factora | |||||
---|---|---|---|---|---|
acceptor fluorophore | |||||
fractional occupancy (%) | NCT | TMR | OG | YFP | RFP |
0.05 | 0.46 | <0 | <0 | <0 | <0 |
0.10 | 0.65 | <0 | <0 | <0 | <0 |
0.25 | 0.78 | 0.57 | <0 | <0 | <0 |
0.50 | 0.85 | 0.71 | <0 | <0 | <0 |
1.0 | 0.92 | 0.81 | 0.046 | <0 | <0 |
2.5 | 0.95 | 0.90 | 0.78 | <0 | <0 |
5.0 | 0.96 | 0.93 | 0.88 | 0.23 | 0.21 |
10 | 0.98 | 0.95 | 0.90 | 0.75 | 0.65 |
25 | 0.98 | 0.97 | 0.96 | 0.89 | 0.82 |
50 | 0.98 | 0.98 | 0.98 | 0.92 | 0.91 |
100 | 0.99 | 0.99 | 0.97 | 0.97 | 0.93 |
The Z′-factor was determined for each value of % occupancy for Nluc-HT-OG, Nluc-HT-TMR, and Nluc-HT-NCT and for Nluc-turboYFP and Nluc-TagRFP (using Nluc-HT-PEG-biotin as the unoccupied donor).
Performance of NanoBRET for Quantifying Protein Interactions
Figure 2

Figure 2. Schematic of NanoBRET for detecting protein–protein interactions.
Figure 3

Figure 3. Comparative analysis of BRET1 and NanoBRET using the Frb/FKBP model system. (a) Optimization of donor and acceptor orientation for the Frb/FKBP interaction. BRET ratios were determined on HEK293 cells transiently transfected with the indicated Frb and FKBP fusion proteins at a donor/acceptor ratio of 1:4, each measured using untreated and rapamycin treated (1 μM, 15 min) samples. The optimal configuration for either BRET1 or NanoBRET is indicated by the hatch pattern. (b) HEK293 cells were transiently transfected with either Frb-Nluc/FKBP-HT or Frb-RLuc8/FKBP-turboYFP at a donor/acceptor ratio of 1:4. The cells were plated in a 384-well plate and left untreated (n = 3) or treated with the indicated concentration of rapamycin (n = 3) for 15 min before determining the BRET ratio.
Figure 4

Figure 4. Effect of expression level on assay sensitivity for detecting the interaction of FKBP with Frb. HeLa cells were transfected with a serial dilution of expression constructs for (a) Frb-Nluc and FKBP-HT or (b) Frb-Rluc8 and FKBP-TurboYFP (1:1 ratio) and plated in thre 96-well plates. BRET ratios were determined for untreated or rapamycin treated samples (1 μM rapamycin, 15 min). Shown is a representative experiment with all individual data points (n = 12) as well as mean ± SD plotted against DNA dilution used for transfection.
Analysis of Bromodomain Interactions
Figure 5

Figure 5. NanoBRET assays for bromodomain protein interactions with chromatin. NanoBRET assays were developed to measure the binding of Nluc-BRD4 (a) or Nluc-CBP (b) to Histone H3.3-HT. HEK293 cells were transfected with the indicated combination of constructs. The BRET ratio for each sample was determined following treatment for 18 h with the indicated concentrations of I-BET151. Shown are the means ± SEM of three independent experiments performed in quadruplicate.
NanoBRET for Cellular Imaging
Figure 6

Figure 6. Real time imaging of ligand induced interaction of AVPR2-HT with Nluc-β-arrestin2 interaction. HeLa cells were transfected with expression constructs for AVPR2-HT and Nluc-β-arrestin2. Sequential acquisition of images was initiated under unstimulated conditions. After establishing a baseline for seven acquisition cycles, 1 μM arginine vasopressin was added to the sample by injection followed by acquisition cycles (13.4 s per cycle) covering approximately 20 min. (a) The panel shows pseudocolored images of the donor (cyan) and acceptor (yellow) channel at indicated time points following treatment with AVP. The images are representative of the results obtained in five independent experiments (scale bar = 10 μm). (b) Kinetic of Nluc-β-arrestin2 recruitment following stimulation with AVP measured by imaging or in plate reader. The imaging data represent five individual regions of interest (ROI), each representing a single cell. The data shown for the plate based assay format are the results of a representative experiment performed in triplicate. In order to compare results obtained from different analytical platforms, the data were plotted as relative change in BRET normalized to the maximum BRET ratio obtained for each data set.
Conclusions
Material and Methods
Fractional Occupancy Assay
Live Cell Labeling with HaloTag NCT Ligand
Frb/FKBP Interaction Assays
Supporting Information
Detailed experimental protocols, supplementary figures, and tables. This material is available free of charge at The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acschembio.5b00143.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
References
This article references 25 other publications.
- 1Ciruela, F. (2008) Fluorescence-based methods in the study of protein-protein interactions in living cells Curr. Opin. Biotechnol. 19, 338– 343Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtValsL7K&md5=1e7c77ca817344a77800b02aa7052791Fluorescence-based methods in the study of protein-protein interactions in living cellsCiruela, FranciscoCurrent Opinion in Biotechnology (2008), 19 (4), 338-343CODEN: CUOBE3; ISSN:0958-1669. (Elsevier B.V.)A review. Multiprotein complexes partake in nearly all cell functions, thus the characterization and visualization of protein-protein interactions in living cells constitute an important step in the study of a large array of cellular mechanisms. Recently, noninvasive fluorescence-based methods using resonance energy transfer (RET), namely bioluminescence-RET (BRET) and fluorescence-RET (FRET), and those centered on protein fragment complementation, such as bimol. fluorescence complementation (BiFC), have been successfully used in the study of protein interactions. These new technologies are nowadays the most powerful approaches for visualizing the interactions occurring within protein complexes in living cells, thus enabling the investigation of protein behavior in their normal milieu. Here we address the individual strengths and weaknesses of these methods when applied to the study of protein-protein interactions.
- 2Angers, S., Salahpour, A., Joly, E., Hilairet, S., Chelsky, D., Dennis, M., and Bouvier, M. (2000) Detection of beta 2-adrenergic receptor dimerization in living cells using bioluminescence resonance energy transfer (BRET) Proc. Natl. Acad. Sci. U.S.A. 97, 3684– 3689Google ScholarThere is no corresponding record for this reference.
- 3Xu, Y., Piston, D. W., and Johnson, C. H. (1999) A bioluminescence resonance energy transfer (BRET) system: application to interacting circadian clock proteins Proc. Natl. Acad. Sci. U.S.A. 96, 151– 156Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXjvV2itw%253D%253D&md5=eb87cabdad3ba1ee84b52692e4ef8f20A bioluminescence resonance energy transfer (BRET) system: application to interacting circadian clock proteinsXu, Yao; Piston, David W.; Johnson, Carl HirschieProceedings of the National Academy of Sciences of the United States of America (1999), 96 (1), 151-156CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)We describe a method for assaying protein interactions that offers some attractive advantages over previous assays. This method, called bioluminescence resonance energy transfer (BRET), uses a bioluminescent luciferase that is genetically fused to one candidate protein, and a green fluorescent protein mutant fused to another protein of interest. Interactions between the two fusion proteins can bring the luciferase and green fluorescent protein close enough for resonance energy transfer to occur, thus changing the color of the bioluminescent emission. By using proteins encoded by circadian (daily) clock genes from cyanobacteria, we use the BRET technique to demonstrate that the clock protein KaiB interacts to form homodimers. BRET should be particularly useful for testing protein interactions within native cells, esp. with integral membrane proteins or proteins targeted to specific organelles.
- 4Wampler, J. E., Hori, K., Lee, J. W., and Cormier, M. J. (1971) Structured bioluminescence. Two emitters during both the in vitro and the in vivo bioluminescence of the sea pansy, Renilla Biochemistry 10, 2903– 2909Google ScholarThere is no corresponding record for this reference.
- 5Pfleger, K. D. and Eidne, K. A. (2006) Illuminating insights into protein-protein interactions using bioluminescence resonance energy transfer (BRET) Nat. Methods 3, 165– 174Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhsFOru7k%253D&md5=bc83e4f90f5ab10ed12286eb20849cccIlluminating insights into protein-protein interactions using bioluminescence resonance energy transfer (BRET)Pfleger, Kevin D. G.; Eidne, Karin A.Nature Methods (2006), 3 (3), 165-174CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)A review. Bioluminescence resonance energy transfer (BRET) is a straightforward biophys. technique for studying protein-protein interactions. It requires: (1) that proteins of interest and suitable controls be labeled with either a donor or acceptor mol., (2) placement of these labeled proteins in the desired environment for assessing their potential interaction, and (3) use of suitable detection instrumentation to monitor resultant energy transfer. There are now several possible applications, combinations of donor and acceptor mols., potential assay environments and detection system perturbations. Therefore, this review aims to demystify and clarify the important aspects of the BRET methodol. that should be considered when using this technique.
- 6Bertrand, L., Parent, S., Caron, M., Legault, M., Joly, E., Angers, S., Bouvier, M., Brown, M., Houle, B., and Menard, L. (2002) The BRET2/arrestin assay in stable recombinant cells: a platform to screen for compounds that interact with G protein-coupled receptors (GPCRS) J. Recept. Signal Transduction Res. 22, 533– 541Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38Xps1ejsbY%253D&md5=a2b21a28d1737f18c064989299e33086The BRET2/arrestin assay in stable recombinant cells: a platform to screen for compounds that interact with G protein-coupled receptors (GPCRS)Bertrand, Lucie; Parent, Stephane; Caron, Mireille; Legault, Mireille; Joly, Erik; Angers, Stephane; Bouvier, Michel; Brown, Mike; Houle, Benoit; Menard, LucJournal of Receptors and Signal Transduction (2002), 22 (1-4), 533-541CODEN: JRSTCT ISSN:. (Marcel Dekker, Inc.)In BRET2 (Bioluminescence Resonance Energy Transfer), a Renilla luciferase (RLuc) is used as the donor protein, while a Green Fluorescent Protein (GFP2) is used as the acceptor protein. In the presence of the cell permeable substrate DeepBlueC, RLuc emits blue light at 395 nm. If the GFP2 is brought into close proximity to RLuc via a specific biomol. interaction, the GFP2 will absorb the blue light energy and reemit green light at 510 nm. BRET2 signals are therefore easily detd. by measuring the ratio of green over blue light (510/395 nm) using appropriate dual channel luminometry instruments (e.g., Fusion Universal Microplate Analyzer, Packard BioScience). Since no light source is required for BRET2 assays, the technol. does not suffer from high fluorescent background or photobleaching, the common problems assocd. with std. FRET-based assays. Using BRET2, the authors developed a generic G Protein-Coupled Receptor (GPCR) assay based on the observation that activation of the majority of GPCRs by agonists leads to the interaction of β-arrestin (a protein that is involved in receptor desensitization and sequestration) with the receptor. The authors established a cell line stably expressing the GFP2:β-arrestin 2 fusion protein, and showed that it can be used to monitor the activation of various transiently expressed GPCRs, in BRET2/arrestin assays. In addn., using the HEK 293/GFP2:β-arrestin 2 cell line as a recipient, the authors generated a double-stable line co-expressing the vasopressin 2 receptor (V2R) fused to RLuc (V2R:RLuc) and used it for the pharmacol. characterization of compds. in BRET2/arrestin assays. This approach yields genuine pharmacol. and supports the BRET2/arrestin assay as a tool that can be used with recombinant cell lines to characterize ligand-GPCR interactions which can be applied to ligand identification for orphan receptors.
- 7Hamdan, F. F., Audet, M., Garneau, P., Pelletier, J., and Bouvier, M. (2005) High-throughput screening of G protein-coupled receptor antagonists using a bioluminescence resonance energy transfer 1-based beta-arrestin2 recruitment assay J. Biomol. Screening 10, 463– 475Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhtVers77J&md5=40aadd9c704f1eee984a2795c374e60aHigh-throughput screening of G protein-coupled receptor antagonists using a bioluminescence resonance energy transfer 1-based β-arrestin2 recruitment assayHamdan, Fadi F.; Audet, Martin; Garneau, Philippe; Pelletier, Jerry; Bouvier, MichelJournal of Biomolecular Screening (2005), 10 (5), 463-475CODEN: JBISF3; ISSN:1087-0571. (Sage Publications)In this study, the authors developed HEK293 cell lines that stably coexpressed optimal amts. of β-arrestin2-Rluc and VENUS fusions of G protein-coupled receptors (GPCRs) belonging to both class A and class B receptors, which include receptors that interact transiently or stably with β-arrestins. This allowed the use of a bioluminescence resonance energy transfer (BRET) 1-β-arrestin2 translocation assay to quantify receptor activation or inhibition. One of the developed cell lines coexpressing CCR5-VENUS and β-arrestin2-/Renilla luciferase was then used for high-throughput screening (HTS) for antagonists of the chemokine receptor CCR5, the primary co-receptor for HIV. A total of 26,000 compds. were screened for inhibition of the agonist-promoted β-arrestin2 recruitment to CCR5, and 12 compds. were found to specifically inhibit the agonist-induced β-arrestin2 recruitment to CCR5. Three of the potential hits were further tested using other functional assays, and their abilities to inhibit CCR5 agonist-promoted signaling were confirmed. This is the 1st study describing a BRET1-β-arrestin recruitment assay in stable mammalian cells and its successful application in HTS for GPCRs antagonists.
- 8Loening, A. M., Fenn, T. D., Wu, A. M., and Gambhir, S. S. (2006) Consensus guided mutagenesis of Renilla luciferase yields enhanced stability and light output Protein Eng., Des. Sel. 19, 391– 400Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xotl2ksro%253D&md5=c006a4ff7a42c04c1ccf02b80fb1c9ffConsensus guided mutagenesis of Renilla luciferase yields enhanced stability and light outputLoening, Andreas Markus; Fenn, Timothy David; Wu, Anna M.; Gambhir, Sanjiv SamProtein Engineering, Design & Selection (2006), 19 (9), 391-400CODEN: PEDSBR; ISSN:1741-0126. (Oxford University Press)Luciferases, which have seen expansive employment as reporter genes in biol. research, could also be used in applications where the protein itself is conjugated to ligands to create probes that are appropriate for use in small animal imaging. As the bioluminescence activity of commonly used luciferases is too labile in serum to permit this application, specific mutations of Renilla luciferase, selected using a consensus sequence driven strategy, were screened for their ability to confer stability of activity in serum as well as their light output. Using this information, a total of eight favorable mutations were combined to generate a mutant Renilla luciferase (RLuc8) that, compared with the parental enzyme, is 200-fold more resistant to inactivation in murine serum and exhibits a 4-fold improvement in light output. Results of the mutational anal. were also used to generate a double mutant optimized for use as a reporter gene. The double mutant had half the resistance to inactivation in serum of the native enzyme while yielding a 5-fold improvement in light output.These variants of Renilla luciferase, which exhibit significantly improved properties compared with the native enzyme, will allow enhanced sensitivity in existing luciferase-based assays as well as enable the development of novel probes labeled with the luciferase protein.
- 9Kocan, M., See, H. B., Seeber, R. M., Eidne, K. A., and Pfleger, K. D. (2008) Demonstration of improvements to the bioluminescence resonance energy transfer (BRET) technology for the monitoring of G protein-coupled receptors in live cells J. Biomol. Screening 13, 888– 898Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjtVymtr0%253D&md5=52bd4ecaa2dd5f265e7a02deeb407556Demonstration of improvements to the bioluminescence resonance energy transfer (BRET) technology for the monitoring of G protein-coupled receptors in live cellsKocan, Martina; See, Heng B.; Seeber, Ruth M.; Eidne, Karin A.; Pfleger, Kevin D. G.Journal of Biomolecular Screening (2008), 13 (9), 888-898CODEN: JBISF3; ISSN:1087-0571. (Sage Publications)The bioluminescence resonance energy transfer (BRET) technique has become extremely popular for studying protein-protein interactions in living cells and real time. Of particular interest is the ability to monitor interactions between G protein-coupled receptors, such as the TSH-releasing hormone receptor (TRHR), and proteins crit. for regulating their function, such as β-arrestin. Using TRHR/β-arrestin interactions, we have demonstrated improvements to all 3 generations of BRET (BRET, BRET, and eBRET) by using the novel forms of luciferase, Rluc2 and Rluc8, developed by the Gambhir lab. Furthermore, for the 1st time it was possible to use the BRET2 system to detect ligand-induced G protein-coupled receptor/β-arrestin interactions over prolonged periods (on the scale of hours rather than seconds) with a very stable signal. As demonstrated by our Z'-factor data, these luciferases increase the sensitivity of BRET to such an extent that they substantially increase the potential applicability of this technol. for effective drug discovery high-throughput screening.
- 10Hall, M. P., Unch, J., Binkowski, B. F., Valley, M. P., Butler, B. L., Wood, M. G., Otto, P., Zimmerman, K., Vidugiris, G., Machleidt, T., Robers, M. B., Benink, H. A., Eggers, C. T., Slater, M. R., Meisenheimer, P. L., Klaubert, D. H., Fan, F., Encell, L. P., and Wood, K. V. (2012) Engineered luciferase reporter from a deep sea shrimp utilizing a novel imidazopyrazinone substrate ACS Chem. Biol. 7, 1848– 1857Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xht1Smu7%252FN&md5=0581a7e44935671471577ade1ff538ebEngineered Luciferase Reporter from a Deep Sea Shrimp Utilizing a Novel Imidazopyrazinone SubstrateHall, Mary P.; Unch, James; Binkowski, Brock F.; Valley, Michael P.; Butler, Braeden L.; Wood, Monika G.; Otto, Paul; Zimmerman, Kristopher; Vidugiris, Gediminas; Machleidt, Thomas; Robers, Matthew B.; Benink, Helene A.; Eggers, Christopher T.; Slater, Michael R.; Meisenheimer, Poncho L.; Klaubert, Dieter H.; Fan, Frank; Encell, Lance P.; Wood, Keith V.ACS Chemical Biology (2012), 7 (11), 1848-1857CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Bioluminescence methodologies have been extraordinarily useful due to their high sensitivity, broad dynamic range, and operational simplicity. These capabilities have been realized largely through incremental adaptations of native enzymes and substrates, originating from luminous organisms of diverse evolutionary lineages. We engineered both an enzyme and substrate in combination to create a novel bioluminescence system capable of more efficient light emission with superior biochem. and phys. characteristics. Using a small luciferase subunit (19 kDa) from the deep sea shrimp Oplophorus gracilirostris, we have improved luminescence expression in mammalian cells ∼2.5 million-fold by merging optimization of protein structure with development of a novel imidazopyrazinone substrate (furimazine). The new luciferase, NanoLuc, produces glow-type luminescence (signal half-life >2 h) with a specific activity ∼150-fold greater than that of either firefly (Photinus pyralis) or Renilla luciferases similarly configured for glow-type assays. In mammalian cells, NanoLuc shows no evidence of post-translational modifications or subcellular partitioning. The enzyme exhibits high phys. stability, retaining activity with incubation up to 55 °C or in culture medium for >15 h at 37 °C. As a genetic reporter, NanoLuc may be configured for high sensitivity or for response dynamics by appending a degrdn. sequence to reduce intracellular accumulation. Appending a signal sequence allows NanoLuc to be exported to the culture medium, where reporter expression can be measured without cell lysis. Fusion onto other proteins allows luminescent assays of their metab. or localization within cells. Reporter quantitation is achievable even at very low expression levels to facilitate more reliable coupling with endogenous cellular processes.
- 11Los, G. V., Encell, L. P., McDougall, M. G., Hartzell, D. D., Karassina, N., Zimprich, C., Wood, M. G., Learish, R., Ohana, R. F., Urh, M., Simpson, D., Mendez, J., Zimmerman, K., Otto, P., Vidugiris, G., Zhu, J., Darzins, A., Klaubert, D. H., Bulleit, R. F., and Wood, K. V. (2008) HaloTag: a novel protein labeling technology for cell imaging and protein analysis ACS Chem. Biol. 3, 373– 382Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXmvVGru7k%253D&md5=66e11d2da0992e634150392ea9575f75HaloTag: A Novel Protein Labeling Technology for Cell Imaging and Protein AnalysisLos, Georgyi V.; Encell, Lance P.; McDougall, Mark G.; Hartzell, Danette D.; Karassina, Natasha; Zimprich, Chad; Wood, Monika G.; Learish, Randy; Ohana, Rachel Friedman; Urh, Marjeta; Simpson, Dan; Mendez, Jacqui; Zimmerman, Kris; Otto, Paul; Vidugiris, Gediminas; Zhu, Ji; Darzins, Aldis; Klaubert, Dieter H.; Bulleit, Robert F.; Wood, Keith V.ACS Chemical Biology (2008), 3 (6), 373-382CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)We have designed a modular protein tagging system that allows different functionalities to be linked onto a single genetic fusion, either in soln., in living cells, or in chem. fixed cells. The protein tag (HaloTag) is a modified haloalkane dehalogenase designed to covalently bind to synthetic ligands (HaloTag ligands). The synthetic ligands comprise a chloroalkane linker attached to a variety of useful mols., such as fluorescent dyes, affinity handles, or solid surfaces. Covalent bond formation between the protein tag and the chloroalkane linker is highly specific, occurs rapidly under physiol. conditions, and is essentially irreversible. We demonstrate the utility of this system for cellular imaging and protein immobilization by analyzing multiple mol. processes assocd. with NF-κB-mediated cellular physiol., including imaging of subcellular protein translocation and capture of protein-protein and protein-DNA complexes.
- 12Shagin, D. A., Barsova, E. V., Yanushevich, Y. G., Fradkov, A. F., Lukyanov, K. A., Labas, Y. A., Semenova, T. N., Ugalde, J. A., Meyers, A., Nunez, J. M., Widder, E. A., Lukyanov, S. A., and Matz, M. V. (2004) GFP-like proteins as ubiquitous metazoan superfamily: evolution of functional features and structural complexity Mol. Biol. Evol. 21, 841– 850Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjsFyqsrs%253D&md5=3b2ea95fc034e2485d9c5b76e32b71e1GFP-like proteins as ubiquitous metazoan superfamily: evolution of functional features and structural complexityShagin, Dmitry A.; Barsova, Ekaterina V.; Yanushevich, Yurii G.; Fradkov, Arkady F.; Lukyanov, Konstantin A.; Labas, Yulii A.; Semenova, Tatiana N.; Ugalde, Juan A.; Meyers, Ann; Nunez, Jose M.; Widder, Edith A.; Lukyanov, Sergey A.; Matz, Mikhail V.Molecular Biology and Evolution (2004), 21 (5), 841-850CODEN: MBEVEO; ISSN:0737-4038. (Oxford University Press)Homologs of the green fluorescent protein (GFP), including the recently described GFP-like domains of certain extracellular matrix proteins in Bilaterian organisms, are remarkably similar at the protein structure level, yet they often perform totally unrelated functions, thereby warranting recognition as a superfamily. Here we describe diverse GFP-like proteins from previously undersampled and completely new sources, including hydromedusae and planktonic Copepoda. In hydromedusae, yellow and nonfluorescent purple proteins were found in addn. to greens. Notably, the new yellow protein seems to follow exactly the same structural soln. to achieving the yellow color of fluorescence as YFP, an engineered yellow-emitting mutant variant of GFP. The addn. of these new sequences made it possible to resolve deep-level phylogenetic relationships within the superfamily. Fluorescence (most likely green) must have already existed in the common ancestor of Cnidaria and Bilateria, and therefore GFP-like proteins may be responsible for fluorescence and/or coloration in virtually any animal. At least 15 color diversification events can be inferred following the max. parsimony principle in Cnidaria. Origination of red fluorescence and nonfluorescent purple-blue colors on several independent occasions provides a remarkable example of convergent evolution of complex features at the mol. level.
- 13Merzlyak, E. M., Goedhart, J., Shcherbo, D., Bulina, M. E., Shcheglov, A. S., Fradkov, A. F., Gaintzeva, A., Lukyanov, K. A., Lukyanov, S., Gadella, T. W., and Chudakov, D. M. (2007) Bright monomeric red fluorescent protein with an extended fluorescence lifetime Nat. Methods 4, 555– 557Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXntVCitLk%253D&md5=3351e2581f2ff447cc1f114b7c91cc46Bright monomeric red fluorescent protein with an extended fluorescence lifetimeMerzlyak, Ekaterina M.; Goedhart, Joachim; Shcherbo, Dmitry; Bulina, Mariya E.; Shcheglov, Aleksandr S.; Fradkov, Arkady F.; Gaintzeva, Anna; Lukyanov, Konstantin A.; Lukyanov, Sergey; Gadella, Theodorus W. J.; Chudakov, Dmitriy M.Nature Methods (2007), 4 (7), 555-557CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Fluorescent proteins have become extremely popular tools for in vivo imaging and esp. for the study of localization, motility and interaction of proteins in living cells. Here the authors report TagRFP, a monomeric red fluorescent protein, which is characterized by high brightness, complete chromophore maturation, prolonged fluorescence lifetime and high pH-stability. These properties make TagRFP an excellent tag for protein localization studies and fluorescence resonance energy transfer (FRET) applications.
- 14Nagai, T., Ibata, K., Park, E. S., Kubota, M., Mikoshiba, K., and Miyawaki, A. (2002) A variant of yellow fluorescent protein with fast and efficient maturation for cell-biological applications Nat. Biotechnol. 20, 87– 90Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XislSrsQ%253D%253D&md5=1eae1970c042f58236113702a2f660d0A variant of yellow fluorescent protein with fast and efficient maturation for cell-biological applicationsNagai, Takeharu; Ibata, Keiji; Park, Eun Sun; Kubota, Mie; Mikoshiba, Katsuhiko; Miyawaki, AtsushiNature Biotechnology (2002), 20 (1), 87-90CODEN: NABIF9; ISSN:1087-0156. (Nature America Inc.)The green fluorescent protein (GFP) from the jellyfish Aequorea victoria has provided a myriad of applications for biol. systems. Over the last several years, mutagenesis studies have improved folding properties of GFP (refs. 1,2). However, slow maturation is still a big obstacle to the use of GFP variants for visualization. These problems are exacerbated when GFP variants are expressed at 37° and/or targeted to certain organelles. Thus, obtaining GFP variants that mature more efficiently is crucial for the development of expanded research applications. Among Aequorea GFP variants, yellow fluorescent proteins (YFPs) are relatively acid-sensitive, and uniquely quenched by chloride ion (Cl-). For YFP to be fully and stably fluorescent, mutations that decrease the sensitivity to both pH and Cl- are desired. Here we describe the development of an improved version of YFP named "Venus". Venus contains a novel mutation, F46L, which at 37° greatly accelerates oxidn. of the chromophore, the rate-limiting step of maturation. As a result of other mutations, F64L/M153T/V163A/S175G, Venus folds well and is relatively tolerant of exposure to acidosis and Cl-. We succeeded in efficiently targeting a neuropeptide Y-Venus fusion protein to the dense-core granules of PC12 cells. Its secretion was readily monitored by measuring release of fluorescence into the medium. The use of Venus as an acceptor allowed early detection of reliable signals of fluorescence resonance energy transfer (FRET) for Ca2+ measurements in brain slices. With the improved speed and efficiency of maturation and the increased resistance to environment, Venus will enable fluorescent labeling that were not possible before.
- 15Shaner, N. C., Steinbach, P. A., and Tsien, R. Y. (2005) A guide to choosing fluorescent proteins Nat. Methods 2, 905– 909Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXht1aksr3F&md5=2aefee1269ed0f92888a47cedd53dcabA guide to choosing fluorescent proteinsShaner, Nathan C.; Steinbach, Paul A.; Tsien, Roger Y.Nature Methods (2005), 2 (12), 905-909CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)The recent explosion in the diversity of available fluorescent proteins (FPs) promises a wide variety of new tools for biol. imaging. With no unified std. for assessing these tools, however, a researcher is faced with difficult questions. Which FPs are best for general use. Which are the brightest. What addnl. factors det. which are best for a given expt.. Although in many cases, a trial-and-error approach may still be necessary in detg. the answers to these questions, a unified characterization of the best available FPs provides a useful guide in narrowing down the options.
- 16Galarneau, A., Primeau, M., Trudeau, L. E., and Michnick, S. W. (2002) Beta-lactamase protein fragment complementation assays as in vivo and in vitro sensors of protein protein interactions Nat. Biotechnol. 20, 619– 622Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XktFOhsrY%253D&md5=284ef04b2969d864811bab12a6de3e4bβ-Lactamase protein fragment complementation assays as in vivo and in vitro sensors of protein-protein interactionsGalarneau, Andre; Primeau, Martin; Trudeau, Louis-Eric; Michnick, Stephen W.Nature Biotechnology (2002), 20 (6), 619-622CODEN: NABIF9; ISSN:1087-0156. (Nature America Inc.)We have previously described a strategy for detecting protein-protein interactions based on protein interaction-assisted folding of rationally designed fragments of enzymes. We call this strategy the protein fragment complementation assay (PCA). Here we describe PCAs based on the enzyme TEM-1 β-lactamase (EC: 3.5.2.6), which include simple colorimetric in vitro assays using the cephalosporin nitrocefin and assays in intact cells using the fluorescent substrate CCF2/AM (ref. 6). Constitutive protein-protein interactions of the GCN4 leucine zippers and of apoptotic proteins Bcl2 and Bad, and the homodimerization of Smad3, were tested in an in vitro assay using cell lysates. With the same in vitro assay, we also demonstrate interactions of protein kinase PKB with substrate Bad. The in vitro assay is facile and amenable to high-throughput modes of screening with signal-to-background ratios in the range of 10:1 to 250:1, which is superior to other PCAs developed to date. Furthermore, we show that the in vitro assay can be used for quant. anal. of a small mol.-induced protein interaction, the rapamycin-induced interaction of FKBP and yeast FRB (the FKBP-rapamycin binding domain of TOR (target of rapamycin)). The assay reproduces the known dissocn. const. and no. of sites for this interaction. The combination of in vitro colorimetric and in vivo fluorescence assays of β-lactamase in mammalian cells suggests a wide variety of sensitive and high-throughput large-scale applications, including in vitro protein array anal. of protein-protein or enzyme-protein interactions and in vivo applications such as clonal selection for cells expressing interacting protein partners.
- 17Zhang, J. H., Chung, T. D., and Oldenburg, K. R. (1999) A Simple Statistical Parameter for Use in Evaluation and Validation of High Throughput Screening Assays J. Biomol. Screening 4, 67– 73Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2sbit12juw%253D%253D&md5=0ec4ed2ccdd3b4a16521567a0975d31fA Simple Statistical Parameter for Use in Evaluation and Validation of High Throughput Screening AssaysZhang; Chung; OldenburgJournal of biomolecular screening (1999), 4 (2), 67-73 ISSN:.The ability to identify active compounds (3hits2) from large chemical libraries accurately and rapidly has been the ultimate goal in developing high-throughput screening (HTS) assays. The ability to identify hits from a particular HTS assay depends largely on the suitability or quality of the assay used in the screening. The criteria or parameters for evaluating the 3suitability2 of an HTS assay for hit identification are not well defined and hence it still remains difficult to compare the quality of assays directly. In this report, a screening window coefficient, called 3Z-factor,2 is defined. This coefficient is reflective of both the assay signal dynamic range and the data variation associated with the signal measurements, and therefore is suitable for assay quality assessment. The Z-factor is a dimensionless, simple statistical characteristic for each HTS assay. The Z-factor provides a useful tool for comparison and evaluation of the quality of assays, and can be utilized in assay optimization and validation.
- 18Wells, J. A. and McClendon, C. L. (2007) Reaching for high-hanging fruit in drug discovery at protein-protein interfaces Nature 450, 1001– 1009Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhsVaqtr7F&md5=e7ed69fd8c362a71c4b99f029c16a6fcReaching for high-hanging fruit in drug discovery at protein-protein interfacesWells, James A.; McClendon, Christopher L.Nature (London, United Kingdom) (2007), 450 (7172), 1001-1009CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)A review. Targeting the interfaces between proteins has huge therapeutic potential, but discovering small-mol. drugs that disrupt protein-protein interactions is an enormous challenge. Several recent success stories, however, indicate that protein-protein interfaces might be more tractable than has been thought. These studies discovered small mols. that bind with drug-like potencies to 'hotspots' on the contact surfaces involved in protein-protein interactions. Remarkably, these small mols. bind deeper within the contact surface of the target protein, and bind with much higher efficiencies, than do the contact atoms of the natural protein partner. Some of these small mols. are now making their way through clin. trials, so this high-hanging fruit might not be far out of reach.
- 19Sanchez, R., Meslamani, J., and Zhou, M. M. (2014) The bromodomain: from epigenome reader to druggable target Biochim. Biophys. Acta 1839, 676– 685Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmtFagtrw%253D&md5=95ec21cf13231d9e63bce9b3ccdd6d6fThe bromodomain: From epigenome reader to druggable targetSanchez, Roberto; Meslamani, Jamel; Zhou, Ming-MingBiochimica et Biophysica Acta, Gene Regulatory Mechanisms (2014), 1839 (8), 676-685CODEN: BBAGC6; ISSN:1874-9399. (Elsevier B.V.)A review. Lysine acetylation is a fundamental post-translational modification that plays an important role in the control of gene transcription in chromatin in an ordered fashion. The bromodomain, the conserved structural module present in transcription-assocd. proteins, functions exclusively to recognize acetyl-lysine on histones and non-histone proteins. The structural analyses of bromodomains' recognition of lysine-acetylated peptides derived from histones and cellular proteins provide detailed insights into the differences and unifying features of biol. ligand binding selectivity by the bromodomains. Newly developed small-mol. inhibitors targeting bromodomain proteins further highlight the functional importance of bromodomain/acetyl-lysine binding as a key mechanism in orchestrating mol. interactions and regulation in chromatin biol. and gene transcription. These new studies argue that modulating bromodomain/acetyl-lysine interactions with small-mol. chems. offer new opportunities to control gene expression in a wide array of human diseases including cancer and inflammation. This article is part of a Special Issue entitled: Mol. mechanisms of histone modification function.
- 20Dawson, M. A., Kouzarides, T., and Huntly, B. J. (2012) Targeting epigenetic readers in cancer N. Engl. J. Med. 367, 647– 657Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xht1Chu77F&md5=f5e1e82c8a1105f798b1f7719138721cTargeting epigenetic readers in cancerDawson, Mark A.; Kouzarides, Tony; Huntly, Brian J. P.New England Journal of Medicine (2012), 367 (7), 647-657CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)A review on assocn. between epigenetics and chromatin biol., BET bromodomain inhibitors, and role of BET inhibitors in cancer.
- 21Filippakopoulos, P., Qi, J., Picaud, S., Shen, Y., Smith, W. B., Fedorov, O., Morse, E. M., Keates, T., Hickman, T. T., Felletar, I., Philpott, M., Munro, S., McKeown, M. R., Wang, Y., Christie, A. L., West, N., Cameron, M. J., Schwartz, B., Heightman, T. D., La Thangue, N., French, C. A., Wiest, O., Kung, A. L., Knapp, S., and Bradner, J. E. (2010) Selective inhibition of BET bromodomains Nature 468, 1067– 1073Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXotlGmtA%253D%253D&md5=bd9cadac86124d2c6c3cd5b2a92d68c2Selective inhibition of BET bromodomainsFilippakopoulos, Panagis; Qi, Jun; Picaud, Sarah; Shen, Yao; Smith, William B.; Fedorov, Oleg; Morse, Elizabeth M.; Keates, Tracey; Hickman, Tyler T.; Felletar, Ildiko; Philpott, Martin; Munro, Shongah; McKeown, Michael R.; Wang, Yuchuan; Christie, Amanda L.; West, Nathan; Cameron, Michael J.; Schwartz, Brian; Heightman, Tom D.; La Thangue, Nicholas; French, Christopher; Wiest, Olaf; Kung, Andrew L.; Knapp, Stefan; Bradner, James E.Nature (London, United Kingdom) (2010), 468 (7327), 1067-1073CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Epigenetic proteins are intently pursued targets in ligand discovery. So far, successful efforts have been limited to chromatin modifying enzymes, or so-called epigenetic 'writers' and 'erasers'. Potent inhibitors of histone binding modules have not yet been described. Here the authors report a cell-permeable small mol. (I,JQ1) that binds competitively to acetyl-lysine recognition motifs, or bromodomains. High potency and specificity towards a subset of human bromodomains is explained by co-crystal structures with bromodomain and extra-terminal (BET) family member BRD4, revealing excellent shape complementarity with the acetyl-lysine binding cavity. Recurrent translocation of BRD4 is obsd. in a genetically-defined, incurable subtype of human squamous carcinoma. Competitive binding by JQ1 displaces the BRD4 fusion oncoprotein from chromatin, prompting squamous differentiation and specific antiproliferative effects in BRD4-dependent cell lines and patient-derived xenograft models. These data establish proof-of-concept for targeting protein-protein interactions of epigenetic 'readers', and provide a versatile chem. scaffold for the development of chem. probes more broadly throughout the bromodomain family.
- 22Dawson, M. A., Prinjha, R. K., Dittmann, A., Giotopoulos, G., Bantscheff, M., Chan, W. I., Robson, S. C., Chung, C. W., Hopf, C., Savitski, M. M., Huthmacher, C., Gudgin, E., Lugo, D., Beinke, S., Chapman, T. D., Roberts, E. J., Soden, P. E., Auger, K. R., Mirguet, O., Doehner, K., Delwel, R., Burnett, A. K., Jeffrey, P., Drewes, G., Lee, K., Huntly, B. J., and Kouzarides, T. (2011) Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia Nature 478, 529– 533Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXht1GqsL%252FO&md5=dffb4b81da4ca8291c27139d0df86324Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemiaDawson, Mark A.; Prinjha, Rab K.; Dittman, Antje; Giotopoulos, George; Bantscheff, Marcus; Chan, Wai-In; Robson, Samuel C.; Chung, Chun-wa; Hopf, Carsten; Savitski, Mikhail M.; Huthmacher, Carola; Gudgin, Emma; Lugo, Dave; Beinke, Soren; Chapman, Trevor D.; Roberts, Emma J.; Soden, Peter E.; Auger, Kurt R.; Mirguet, Olivier; Doehner, Konstanze; Delwel, Ruud; Burnett, Alan K.; Jeffrey, Phillip; Drewes, Gerard; Lee, Kevin; Huntly, Brian J. P.; Kouzarides, TonyNature (London, United Kingdom) (2011), 478 (7370), 529-533CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Recurrent chromosomal translocations involving the mixed lineage leukemia (MLL) gene initiate aggressive forms of leukemia, which are often refractory to conventional therapies. Many MLL-fusion partners are members of the super elongation complex (SEC), a crit. regulator of transcriptional elongation, suggesting that aberrant control of this process has an important role in leukemia induction. Here we use a global proteomic strategy to demonstrate that MLL fusions, as part of SEC and the polymerase-assocd. factor complex (PAFc), are assocd. with the BET family of acetyl-lysine recognizing, chromatin adaptor' proteins. These data provided the basis for therapeutic intervention in MLL-fusion leukemia, via the displacement of the BET family of proteins from chromatin. We show that a novel small mol. inhibitor of the BET family, GSK1210151A (I-BET151), has profound efficacy against human and murine MLL-fusion leukemic cell lines, through the induction of early cell cycle arrest and apoptosis. I-BET151 treatment in two human leukemia cell lines with different MLL fusions alters the expression of a common set of genes whose function may account for these phenotypic changes. The mode of action of I-BET151 is, at least in part, due to the inhibition of transcription at key genes (BCL2, C-MYC and CDK6) through the displacement of BRD3/4, PAFc and SEC components from chromatin. In vivo studies indicate that I-BET151 has significant therapeutic value, providing survival benefit in two distinct mouse models of murine MLL-AF9 and human MLL-AF4 leukemia. Finally, the efficacy of I-BET151 against human leukemia stem cells is demonstrated, providing further evidence of its potent therapeutic potential. These findings establish the displacement of BET proteins from chromatin as a promising epigenetic therapy for these aggressive leukemias.
- 23Oakley, R. H., Laporte, S. A., Holt, J. A., Barak, L. S., and Caron, M. G. (1999) Association of beta-arrestin with G protein-coupled receptors during clathrin-mediated endocytosis dictates the profile of receptor resensitization J. Biol. Chem. 274, 32248– 32257Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXnt12jtbg%253D&md5=6a884b71028ff86e53f664fec39a66cdAssociation of β-arrestin with G protein-coupled receptors during clathrin-mediated endocytosis dictates the profile of receptor resensitizationOakley, Robert H.; Laporte, Stephane A.; Holt, Jason A.; Barak, Larry S.; Caron, Marc G.Journal of Biological Chemistry (1999), 274 (45), 32248-32257CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Resensitization of G protein-coupled receptors (GPCRs) following agonist-mediated desensitization is a necessary step for maintaining physiol. responsiveness. However, the mol. mechanisms governing the nature of GPCR resensitization are poorly understood. Here, we examine the role of β-arrestin in the resensitization of the β2 adrenergic receptor (β2AR), known to recycle and resensitize rapidly, and the vasopressin V2 receptor (V2R), known to recycle and resensitize slowly. Upon agonist activation, both receptors recruit β-arrestin to the plasma membrane and internalize in a β-arrestin- and clathrin-dependent manner. However, whereas β-arrestin dissocs. from the β2AR at the plasma membrane, it internalizes with the V2R into endosomes. The differential trafficking of β-arrestin and the ability of these two receptors to dephosphorylate, recycle, and resensitize is completely reversed when the carboxyl-terminal tails of these two receptors are switched. Moreover, the ability of β-arrestin to remain assocd. with desensitized GPCRs during clathrin-mediated endocytosis is mediated by a specific cluster of phosphorylated serine residues in the receptor carboxyl-terminal tail. These results demonstrate that the interaction of β-arrestin with a specific motif in the GPCR carboxyl-terminal tail dictates the rate of receptor dephosphorylation, recycling, and resensitization, and thus provide direct evidence for a novel mechanism by which β-arrestins regulate the reestablishment of GPCR responsiveness.
- 24Laporte, S. A., Oakley, R. H., Zhang, J., Holt, J. A., Ferguson, S. S., Caron, M. G., and Barak, L. S. (1999) The beta2-adrenergic receptor/betaarrestin complex recruits the clathrin adaptor AP-2 during endocytosis Proc. Natl. Acad. Sci. U.S.A. 96, 3712– 3717Google ScholarThere is no corresponding record for this reference.
- 25Mahen, R., Koch, B., Wachsmuth, M., Politi, A. Z., Perez-Gonzalez, A., Mergenthaler, J., Cai, Y., and Ellenberg, J. (2014) Comparative assessment of fluorescent transgene methods for quantitative imaging in human cells Mol. Biol. Cell 25, 3610Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitFOlsLbF&md5=b41ac0f052dfa7bf26ce2bdcbf9bf093Comparative assessment of fluorescent transgene methods for quantitative imaging in human cellsMahen, Robert; Koch, Birgit; Wachsmuth, Malte; Politi, Antonio Z.; Perez-Gonzalez, Alexis; Mergenthaler, Julia; Cai, Yin; Ellenberg, JanMolecular Biology of the Cell (2014), 25 (22), 3610-3618, 9 pp.CODEN: MBCEEV; ISSN:1939-4586. (American Society for Cell Biology)Fluorescence tagging of proteins is a widely used tool to study protein function and dynamics in live cells. However, the extent to which different mammalian transgene methods faithfully report on the properties of endogenous proteins has not been studied comparatively. Here we use quant. live-cell imaging and single-mol. spectroscopy to analyze how different transgene systems affect imaging of the functional properties of the mitotic kinase Aurora B. We show that the transgene method fundamentally influences level and variability of expression and can severely compromise the ability to report on endogenous binding and localization parameters, providing a guide for quant. imaging studies in mammalian cells.
Cited By
This article is cited by 300 publications.
- Roman C. Sarott, Inchul You, Yen-Der Li, Sean T. Toenjes, Katherine A. Donovan, Pooreum Seo, Martha Ordonez, Woong Sub Byun, Muhammad Murtaza Hassan, Franziska Wachter, Edward T. Chouchani, Mikołaj Słabicki, Eric S. Fischer, Benjamin L. Ebert, Stephen M. Hinshaw, Nathanael S. Gray. Chemical Specification of E3 Ubiquitin Ligase Engagement by Cysteine-Reactive Chemistry. Journal of the American Chemical Society 2023, 145
(40)
, 21937-21944. https://doi.org/10.1021/jacs.3c06622
- Anirban Mukherjee, Farzad Zamani, Takayoshi Suzuki. Evolution of Slow-Binding Inhibitors Targeting Histone Deacetylase Isoforms. Journal of Medicinal Chemistry 2023, 66
(17)
, 11672-11700. https://doi.org/10.1021/acs.jmedchem.3c01160
- Huda Zahid, Jeff P. Costello, Yao Li, Jennifer R. Kimbrough, Marisa Actis, Zoran Rankovic, Qin Yan, William C. K. Pomerantz. Design of Class I/IV Bromodomain-Targeting Degraders for Chromatin Remodeling Complexes. ACS Chemical Biology 2023, 18
(6)
, 1278-1293. https://doi.org/10.1021/acschembio.2c00902
- Wenchao Lu, Yao Liu, Yang Gao, Qixiang Geng, Deepak Gurbani, Lianbo Li, Scott B. Ficarro, Cynthia J. Meyer, Dhiraj Sinha, Inchul You, Jason Tse, Zhixiang He, Wenzhi Ji, Jianwei Che, Audrey Y. Kim, Tengteng Yu, Kenneth Wen, Kenneth C. Anderson, Jarrod A. Marto, Kenneth D. Westover, Tinghu Zhang, Nathanael S. Gray. Development of a Covalent Inhibitor of c-Jun N-Terminal Protein Kinase (JNK) 2/3 with Selectivity over JNK1. Journal of Medicinal Chemistry 2023, 66
(5)
, 3356-3371. https://doi.org/10.1021/acs.jmedchem.2c01834
- Anton DelwigNoriko IshisokoRobert A. Blake. CELLULAR TARGET ENGAGEMENT ASSAYS FOR SMALL-MOLECULE DRUG DISCOVERY. , 419-440. https://doi.org/10.1021/mc-2022-vol57.ch17
- Dimitra Apostolidou, Pan Zhang, Weitao Yang, Piotr E. Marszalek. Mechanical Unfolding and Refolding of NanoLuc via Single-Molecule Force Spectroscopy and Computer Simulations. Biomacromolecules 2022, 23
(12)
, 5164-5178. https://doi.org/10.1021/acs.biomac.2c00997
- Kevin K. Ng, Jennifer A. Prescher. Generalized Bioluminescent Platform To Observe and Track Cellular Interactions. Bioconjugate Chemistry 2022, 33
(10)
, 1876-1884. https://doi.org/10.1021/acs.bioconjchem.2c00348
- Thomas Hanke, Sebastian Mathea, Julia Woortman, Eidarus Salah, Benedict-Tilman Berger, Anthony Tumber, Risa Kashima, Akiko Hata, Bernhard Kuster, Susanne Müller, Stefan Knapp. Development and Characterization of Type I, Type II, and Type III LIM-Kinase Chemical Probes. Journal of Medicinal Chemistry 2022, 65
(19)
, 13264-13287. https://doi.org/10.1021/acs.jmedchem.2c01106
- Rebeka C. Fanti, Stanley N. S. Vasconcelos, Carolina M. C. Catta-Preta, Jaryd R. Sullivan, Gustavo P. Riboldi, Caio V. dos Reis, Priscila Z. Ramos, Aled M. Edwards, Marcel A. Behr, Rafael M. Couñago. A Target Engagement Assay to Assess Uptake, Potency, and Retention of Antibiotics in Living Bacteria. ACS Infectious Diseases 2022, 8
(8)
, 1449-1467. https://doi.org/10.1021/acsinfecdis.2c00073
- Florence Gbahou, Sergiy Levin, Irina G. Tikhonova, Gloria Somalo Barranco, Charlotte Izabelle, Rachel Friedman Ohana, Ralf Jockers. Luminogenic HiBiT Peptide-Based NanoBRET Ligand Binding Assays for Melatonin Receptors. ACS Pharmacology & Translational Science 2022, 5
(8)
, 668-678. https://doi.org/10.1021/acsptsci.2c00096
- Laetitia Ganier, Stephane Betzi, Carine Derviaux, Philippe Roche, Charlotte Dessaux, Christophe Muller, Laurent Hoffer, Xavier Morelli, Jean-Paul Borg. Discovery of Small-Molecule Inhibitors of the PTK7/β-Catenin Interaction Targeting the Wnt Signaling Pathway in Colorectal Cancer. ACS Chemical Biology 2022, 17
(5)
, 1061-1072. https://doi.org/10.1021/acschembio.1c00826
- Yi Wan, Chengli Zong, Xiangpeng Li, Aimin Wang, Yan Li, Tao Yang, Qing Bao, Michael Dubow, Mingying Yang, Ledesma-Amaro Rodrigo, Chuanbin Mao. New Insights for Biosensing: Lessons from Microbial Defense Systems. Chemical Reviews 2022, 122
(9)
, 8126-8180. https://doi.org/10.1021/acs.chemrev.1c01063
- Mariko Orioka, Masatoshi Eguchi, Yuki Mizui, Yuma Ikeda, Akihiro Sakama, Qiaojing Li, Hideaki Yoshimura, Takeaki Ozawa, Daniel Citterio, Yuki Hiruta. A Series of Furimazine Derivatives for Sustained Live-Cell Bioluminescence Imaging and Application to the Monitoring of Myogenesis at the Single-Cell Level. Bioconjugate Chemistry 2022, 33
(3)
, 496-504. https://doi.org/10.1021/acs.bioconjchem.2c00035
- Wallace H. Liu, Robert E. Miner III, Brittany N. Albaugh, Gene E. Ananiev, Scott A. Wildman, John M. Denu. Discovery and Mechanism of Small Molecule Inhibitors Selective for the Chromatin-Binding Domains of Oncogenic UHRF1. Biochemistry 2022, 61
(5)
, 354-366. https://doi.org/10.1021/acs.biochem.1c00698
- Nabanita Nawar, Shazreh Bukhari, Ashley A. Adile, Yujin Suk, Pimyupa Manaswiyoungkul, Krimo Toutah, Olasunkanmi O. Olaoye, Yasir S. Raouf, Abootaleb Sedighi, Harsimran Kaur Garcha, Muhammad Murtaza Hassan, William Gwynne, Johan Israelian, Tudor B. Radu, Mulu Geletu, Ayah Abdeldayem, Justyna M. Gawel, Aaron D. Cabral, Chitra Venugopal, Elvin D. de Araujo, Sheila K. Singh, Patrick T. Gunning. Discovery of HDAC6-Selective Inhibitor NN-390 with in Vitro Efficacy in Group 3 Medulloblastoma. Journal of Medicinal Chemistry 2022, 65
(4)
, 3193-3217. https://doi.org/10.1021/acs.jmedchem.1c01585
- Jon J. Winter-Holt, Catherine Bardelle, Elisabetta Chiarparin, Ian L. Dale, Paul R. J. Davey, Nichola L. Davies, Christopher Denz, Shaun M. Fillery, Carine M. Guérot, Fujin Han, Samantha J. Hughes, Meghana Kulkarni, Zhaoqun Liu, Alexander Milbradt, Thomas A. Moss, Huijun Niu, Joe Patel, Alfred A. Rabow, Marianne Schimpl, Junjie Shi, Dongqing Sun, Dejian Yang, Sylvie Guichard. Discovery of a Potent and Selective ATAD2 Bromodomain Inhibitor with Antiproliferative Activity in Breast Cancer Models. Journal of Medicinal Chemistry 2022, 65
(4)
, 3306-3331. https://doi.org/10.1021/acs.jmedchem.1c01871
- Matthew H. Daniels, Goran Malojcic, Susan L. Clugston, Brett Williams, Marie Coeffet-Le Gal, Xin-Ru Pan-Zhou, Srinivasan Venkatachalan, Jean-Christophe Harmange, Mark Ledeboer. Discovery and Optimization of Highly Selective Inhibitors of CDK5. Journal of Medicinal Chemistry 2022, 65
(4)
, 3575-3596. https://doi.org/10.1021/acs.jmedchem.1c02069
- Mengying Deng, Jing Yuan, Haibin Yang, Xuli Wu, Xiaoyuan Wei, Yang Du, Garry Wong, Yuyong Tao, Gang Liu, Zongwen Jin, Jun Chu. A Genetically Encoded Bioluminescent System for Fast and Highly Sensitive Detection of Antibodies with a Bright Green Fluorescent Protein. ACS Nano 2021, 15
(11)
, 17602-17612. https://doi.org/10.1021/acsnano.1c05164
- Muyang Guan, Mikael V. Garabedian, Marcel Leutenegger, Benjamin S. Schuster, Matthew C. Good, Daniel A. Hammer. Incorporation and Assembly of a Light-Emitting Enzymatic Reaction into Model Protein Condensates. Biochemistry 2021, 60
(42)
, 3137-3151. https://doi.org/10.1021/acs.biochem.1c00373
- Sandra Röhm, Benedict-Tilman Berger, Martin Schröder, Deep Chatterjee, Sebastian Mathea, Andreas C. Joerger, Daniel M. Pinkas, Joshua C. Bufton, Amelie Tjaden, Lohitesh Kovooru, Mark Kudolo, Christian Pohl, Alex N. Bullock, Susanne Müller, Stefan Laufer, Stefan Knapp. Development of a Selective Dual Discoidin Domain Receptor (DDR)/p38 Kinase Chemical Probe. Journal of Medicinal Chemistry 2021, 64
(18)
, 13451-13474. https://doi.org/10.1021/acs.jmedchem.1c00868
- Michael A. Clegg, Natalie H. Theodoulou, Paul Bamborough, Chun-wa Chung, Peter D. Craggs, Emmanuel H. Demont, Laurie J. Gordon, Gemma M. Liwicki, Alex Phillipou, Nicholas C. O. Tomkinson, Rab K. Prinjha, Philip G. Humphreys. Optimization of Naphthyridones into Selective TATA-Binding Protein Associated Factor 1 (TAF1) Bromodomain Inhibitors. ACS Medicinal Chemistry Letters 2021, 12
(8)
, 1308-1317. https://doi.org/10.1021/acsmedchemlett.1c00294
- Riho Takahashi, Takanobu Yasuda, Yuki Ohmuro-Matsuyama, Hiroshi Ueda. BRET Q-Body: A Ratiometric Quench-based Bioluminescent Immunosensor Made of Luciferase–Dye–Antibody Fusion with Enhanced Response. Analytical Chemistry 2021, 93
(21)
, 7571-7578. https://doi.org/10.1021/acs.analchem.0c05217
- Brittany R. Benlian, Pavel E. Z. Klier, Kayli N. Martinez, Marie K. Schwinn, Thomas A. Kirkland, Evan W. Miller. Small Molecule–Protein Hybrid for Voltage Imaging via Quenching of Bioluminescence. ACS Sensors 2021, 6
(5)
, 1857-1863. https://doi.org/10.1021/acssensors.1c00058
- Andrea Peier, Lan Ge, Nicolas Boyer, John Frost, Ruchia Duggal, Kaustav Biswas, Scott Edmondson, Jeffrey D. Hermes, Lin Yan, Chad Zimprich, Ahmad Sadruddin, Hung Yi Kristal Kaan, Arun Chandramohan, Christopher J. Brown, Dawn Thean, Xue Er Lee, Tsz Ying Yuen, Fernando J. Ferrer-Gago, Charles W. Johannes, David P. Lane, Brad Sherborne, Cesear Corona, Matthew B. Robers, Tomi K. Sawyer, Anthony W. Partridge. NanoClick: A High Throughput, Target-Agnostic Peptide Cell Permeability Assay. ACS Chemical Biology 2021, 16
(2)
, 293-309. https://doi.org/10.1021/acschembio.0c00804
- Rachel Friedman
Ohana, Sergiy Levin, Robin Hurst, Michael M. Rosenblatt, Kristopher Zimmerman, Thomas Machleidt, Keith V. Wood, Thomas A. Kirkland. Streamlined Target Deconvolution Approach Utilizing a Single Photoreactive Chloroalkane Capture Tag. ACS Chemical Biology 2021, 16
(2)
, 404-413. https://doi.org/10.1021/acschembio.0c00987
- Daniela Volpato, Michael Kauk, Regina Messerer, Marcel Bermudez, Gerhard Wolber, Andreas Bock, Carsten Hoffmann, Ulrike Holzgrabe. The Role of Orthosteric Building Blocks of Bitopic Ligands for Muscarinic M1 Receptors. ACS Omega 2020, 5
(49)
, 31706-31715. https://doi.org/10.1021/acsomega.0c04220
- Scott A. Busby, Seth Carbonneau, John Concannon, Christoph E. Dumelin, YounKyoung Lee, Shin Numao, Nicole Renaud, Thomas M. Smith, Douglas S. Auld. Advancements in Assay Technologies and Strategies to Enable Drug Discovery. ACS Chemical Biology 2020, 15
(10)
, 2636-2648. https://doi.org/10.1021/acschembio.0c00495
- Travis J. Nelson, Shuo Liang, Cliff I. Stains. A Luminescence-Based System for Identification of Genetically Encodable Inhibitors of Protein Aggregation. ACS Omega 2020, 5
(22)
, 12974-12978. https://doi.org/10.1021/acsomega.0c00779
- Simon C. C. Lucas, Stephen J. Atkinson, Paul Bamborough, Heather Barnett, Chun-wa Chung, Laurie Gordon, Darren J. Mitchell, Alexander Phillipou, Rab K. Prinjha, Robert J. Sheppard, Nicholas C. O. Tomkinson, Robert J. Watson, Emmanuel H. Demont. Optimization of Potent ATAD2 and CECR2 Bromodomain Inhibitors with an Atypical Binding Mode. Journal of Medicinal Chemistry 2020, 63
(10)
, 5212-5241. https://doi.org/10.1021/acs.jmedchem.0c00021
- George S. Sheppard, Le Wang, Steven D. Fidanze, Lisa A. Hasvold, Dachun Liu, John K. Pratt, Chang H. Park, Kenton Longenecker, Wei Qiu, Maricel Torrent, Peter J. Kovar, Mai Bui, Emily Faivre, Xiaoli Huang, Xiaoyu Lin, Denise Wilcox, Lu Zhang, Yu Shen, Daniel H. Albert, Terrance J. Magoc, Ganesh Rajaraman, Warren M. Kati, Keith F. McDaniel. Discovery of N-Ethyl-4-[2-(4-fluoro-2,6-dimethyl-phenoxy)-5-(1-hydroxy-1-methyl-ethyl)phenyl]-6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridine-2-carboxamide (ABBV-744), a BET Bromodomain Inhibitor with Selectivity for the Second Bromodomain. Journal of Medicinal Chemistry 2020, 63
(10)
, 5585-5623. https://doi.org/10.1021/acs.jmedchem.0c00628
- Ivan Pavlinov, Maryna Salkovski, Leslie N. Aldrich. Beclin 1–ATG14L Protein–Protein Interaction Inhibitor Selectively Inhibits Autophagy through Disruption of VPS34 Complex I. Journal of the American Chemical Society 2020, 142
(18)
, 8174-8182. https://doi.org/10.1021/jacs.9b12705
- Eleonora Comeo, Nicholas D. Kindon, Mark Soave, Leigh A. Stoddart, Laura E. Kilpatrick, Peter J. Scammells, Stephen J. Hill, Barrie Kellam. Subtype-Selective Fluorescent Ligands as Pharmacological Research Tools for the Human Adenosine A2A Receptor. Journal of Medicinal Chemistry 2020, 63
(5)
, 2656-2672. https://doi.org/10.1021/acs.jmedchem.9b01856
- Sandra Röhm, Benedict-Tilman Berger, Martin Schröder, Apirat Chaikuad, Rob Winkel, Koen F. W. Hekking, Jorg J. C. Benningshof, Gerhard Müller, Roberta Tesch, Mark Kudolo, Michael Forster, Stefan Laufer, Stefan Knapp. Fast Iterative Synthetic Approach toward Identification of Novel Highly Selective p38 MAP Kinase Inhibitors. Journal of Medicinal Chemistry 2019, 62
(23)
, 10757-10782. https://doi.org/10.1021/acs.jmedchem.9b01227
- Yan Xiong, Holger Greschik, Catrine Johansson, Ludwig Seifert, Johannes Bacher, Kwang-su Park, Nicolas Babault, Michael Martini, Vincent Fagan, Fengling Li, Irene Chau, Thomas Christott, David Dilworth, Dalia Barsyte-Lovejoy, Masoud Vedadi, Cheryl H. Arrowsmith, Paul Brennan, Oleg Fedorov, Manfred Jung, Gillian Farnie, Jing Liu, Udo Oppermann, Roland Schüle, Jian Jin. Discovery of a Potent and Selective Fragment-like Inhibitor of Methyllysine Reader Protein Spindlin 1 (SPIN1). Journal of Medicinal Chemistry 2019, 62
(20)
, 8996-9007. https://doi.org/10.1021/acs.jmedchem.9b00522
- Vincent Fagan, Catrine Johansson, Carina Gileadi, Octovia Monteiro, James E. Dunford, Reshma Nibhani, Martin Philpott, Jessica Malzahn, Graham Wells, Ruth Faram, Adam P. Cribbs, Nadia Halidi, Fengling Li, Irene Chau, Holger Greschik, Srikannathasan Velupillai, Abdellah Allali-Hassani, James Bennett, Thomas Christott, Charline Giroud, Andrew M. Lewis, Kilian V. M. Huber, Nick Athanasou, Chas Bountra, Manfred Jung, Roland Schüle, Masoud Vedadi, Cheryl Arrowsmith, Yan Xiong, Jian Jin, Oleg Fedorov, Gillian Farnie, Paul E. Brennan, Udo Oppermann. A Chemical Probe for Tudor Domain Protein Spindlin1 to Investigate Chromatin Function. Journal of Medicinal Chemistry 2019, 62
(20)
, 9008-9025. https://doi.org/10.1021/acs.jmedchem.9b00562
- Xin Li, Yizhe Wu, Gaofei Tian, Yixiang Jiang, Zheng Liu, Xianbin Meng, Xiucong Bao, Ling Feng, Hongyan Sun, Haiteng Deng, Xiang David Li. Chemical Proteomic Profiling of Bromodomains Enables the Wide-Spectrum Evaluation of Bromodomain Inhibitors in Living Cells. Journal of the American Chemical Society 2019, 141
(29)
, 11497-11505. https://doi.org/10.1021/jacs.9b02738
- Maxwell
M. Sakyiamah, Wataru Nomura, Takuya Kobayakawa, Hirokazu Tamamura. Development of a NanoBRET-Based Sensitive Screening Method for CXCR4 Ligands. Bioconjugate Chemistry 2019, 30
(5)
, 1442-1450. https://doi.org/10.1021/acs.bioconjchem.9b00182
- Luyi Huang, Hui Li, Linli Li, Lu Niu, Raina Seupel, Chengyong Wu, Wei Cheng, Chong Chen, Bisen Ding, Paul E. Brennan, Shengyong Yang. Discovery of Pyrrolo[3,2-d]pyrimidin-4-one Derivatives as a New Class of Potent and Cell-Active Inhibitors of P300/CBP-Associated Factor Bromodomain. Journal of Medicinal Chemistry 2019, 62
(9)
, 4526-4542. https://doi.org/10.1021/acs.jmedchem.9b00096
- Ellen Watts, David Heidenreich, Elizabeth Tucker, Monika Raab, Klaus Strebhardt, Louis Chesler, Stefan Knapp, Benjamin Bellenie, Swen Hoelder. Designing Dual Inhibitors of Anaplastic Lymphoma Kinase (ALK) and Bromodomain-4 (BRD4) by Tuning Kinase Selectivity. Journal of Medicinal Chemistry 2019, 62
(5)
, 2618-2637. https://doi.org/10.1021/acs.jmedchem.8b01947
- Kenneth M. Comess, Shaun M. McLoughlin, Jon A. Oyer, Paul L. Richardson, Henning Stöckmann, Anil Vasudevan, Scott E. Warder. Emerging Approaches for the Identification of Protein Targets of Small Molecules - A Practitioners’ Perspective. Journal of Medicinal Chemistry 2018, 61
(19)
, 8504-8535. https://doi.org/10.1021/acs.jmedchem.7b01921
- Yong Li, Panchun Yang, Ning Lei, Yihan Ma, Yaoting Ji, Chunnan Zhu, Yunhua Wu. Assembly of DNA-Templated Bioluminescent Modules for Amplified Detection of Protein Biomarkers. Analytical Chemistry 2018, 90
(19)
, 11495-11502. https://doi.org/10.1021/acs.analchem.8b02734
- Kristin
M. Riching, Sarah Mahan, Cesear R. Corona, Mark McDougall, James D. Vasta, Matthew B. Robers, Marjeta Urh, Danette L. Daniels. Quantitative Live-Cell Kinetic Degradation and Mechanistic Profiling of PROTAC Mode of Action. ACS Chemical Biology 2018, 13
(9)
, 2758-2770. https://doi.org/10.1021/acschembio.8b00692
- Sean Conroy, Nicholas D. Kindon, Jacqueline Glenn, Leigh A. Stoddart, Richard J. Lewis, Stephen J. Hill, Barrie Kellam, Michael J. Stocks. Synthesis and Evaluation of the First Fluorescent Antagonists of the Human P2Y2 Receptor Based on AR-C118925. Journal of Medicinal Chemistry 2018, 61
(7)
, 3089-3113. https://doi.org/10.1021/acs.jmedchem.8b00139
- Marie K. Schwinn, Thomas Machleidt, Kris Zimmerman, Christopher T. Eggers, Andrew S. Dixon, Robin Hurst, Mary P. Hall, Lance P. Encell, Brock F. Binkowski, and Keith V. Wood . CRISPR-Mediated Tagging of Endogenous Proteins with a Luminescent Peptide. ACS Chemical Biology 2018, 13
(2)
, 467-474. https://doi.org/10.1021/acschembio.7b00549
- Peter J. Tonge . Drug–Target Kinetics in Drug Discovery. ACS Chemical Neuroscience 2018, 9
(1)
, 29-39. https://doi.org/10.1021/acschemneuro.7b00185
- Léa Bouché, Clara D. Christ, Stephan Siegel, Amaury E. Fernández-Montalván, Simon J. Holton, Oleg Fedorov, Antonius ter Laak, Tatsuo Sugawara, Detlef Stöckigt, Cynthia Tallant, James Bennett, Octovia Monteiro, Laura Díaz-Sáez, Paulina Siejka, Julia Meier, Vera Pütter, Jörg Weiske, Susanne Müller, Kilian V. M. Huber, Ingo V. Hartung, and Bernard Haendler . Benzoisoquinolinediones as Potent and Selective Inhibitors of BRPF2 and TAF1/TAF1L Bromodomains. Journal of Medicinal Chemistry 2017, 60
(9)
, 4002-4022. https://doi.org/10.1021/acs.jmedchem.7b00306
- Joel R. Walker, Mary P. Hall, Chad A. Zimprich, Matthew B. Robers, Sarah J. Duellman, Thomas Machleidt, Jacquelynn Rodriguez, and Wenhui Zhou . Highly Potent Cell-Permeable and Impermeable NanoLuc Luciferase Inhibitors. ACS Chemical Biology 2017, 12
(4)
, 1028-1037. https://doi.org/10.1021/acschembio.6b01129
- Philip G. Humphreys, Paul Bamborough, Chun-wa Chung, Peter D. Craggs, Laurie Gordon, Paola Grandi, Thomas G. Hayhow, Jameed Hussain, Katherine L. Jones, Matthew Lindon, Anne-Marie Michon, Jessica F. Renaux, Colin J. Suckling, David F. Tough, and Rab K. Prinjha . Discovery of a Potent, Cell Penetrant, and Selective p300/CBP-Associated Factor (PCAF)/General Control Nonderepressible 5 (GCN5) Bromodomain Chemical Probe. Journal of Medicinal Chemistry 2017, 60
(2)
, 695-709. https://doi.org/10.1021/acs.jmedchem.6b01566
- Stephen J. Dwight and Sergiy Levin . Scalable Regioselective Synthesis of Rhodamine Dyes. Organic Letters 2016, 18
(20)
, 5316-5319. https://doi.org/10.1021/acs.orglett.6b02635
- Stijn J. A. Aper, Pieterjan Dierickx, and Maarten Merkx . Dual Readout BRET/FRET Sensors for Measuring Intracellular Zinc. ACS Chemical Biology 2016, 11
(10)
, 2854-2864. https://doi.org/10.1021/acschembio.6b00453
- Christopher G. England, Emily B. Ehlerding, and Weibo Cai . NanoLuc: A Small Luciferase Is Brightening Up the Field of Bioluminescence. Bioconjugate Chemistry 2016, 27
(5)
, 1175-1187. https://doi.org/10.1021/acs.bioconjchem.6b00112
- Alexander M. Taylor, Alexandre Côté, Michael C. Hewitt, Richard Pastor, Yves Leblanc, Christopher G. Nasveschuk, F. Anthony Romero, Terry D. Crawford, Nico Cantone, Hariharan Jayaram, Jeremy Setser, Jeremy Murray, Maureen H. Beresini, Gladys de Leon Boenig, Zhongguo Chen, Andrew R. Conery, Richard T. Cummings, Leslie A. Dakin, E. Megan Flynn, Oscar W. Huang, Susan Kaufman, Patricia J. Keller, James R. Kiefer, Tommy Lai, Yingjie Li, Jiangpeng Liao, Wenfeng Liu, Henry Lu, Eneida Pardo, Vickie Tsui, Jian Wang, Yongyun Wang, Zhaowu Xu, Fen Yan, Dong Yu, Laura Zawadzke, Xiaoqin Zhu, Xiaoyu Zhu, Robert J. Sims, III, Andrea G. Cochran, Steve Bellon, James E. Audia, Steven Magnuson, and Brian K. Albrecht . Fragment-Based Discovery of a Selective and Cell-Active Benzodiazepinone CBP/EP300 Bromodomain Inhibitor (CPI-637). ACS Medicinal Chemistry Letters 2016, 7
(5)
, 531-536. https://doi.org/10.1021/acsmedchemlett.6b00075
- Remco Arts, Ilona den Hartog, Stefan E. Zijlema, Vito Thijssen, Stan H. E. van der Beelen, and Maarten Merkx . Detection of Antibodies in Blood Plasma Using Bioluminescent Sensor Proteins and a Smartphone. Analytical Chemistry 2016, 88
(8)
, 4525-4532. https://doi.org/10.1021/acs.analchem.6b00534
- F. Anthony Romero, Alexander M. Taylor, Terry D. Crawford, Vickie Tsui, Alexandre Côté, and Steven Magnuson . Disrupting Acetyl-Lysine Recognition: Progress in the Development of Bromodomain Inhibitors. Journal of Medicinal Chemistry 2016, 59
(4)
, 1271-1298. https://doi.org/10.1021/acs.jmedchem.5b01514
- Jia Zhao, Travis J. Nelson, Quyen Vu, Tiffany Truong, and Cliff I. Stains . Self-Assembling NanoLuc Luciferase Fragments as Probes for Protein Aggregation in Living Cells. ACS Chemical Biology 2016, 11
(1)
, 132-138. https://doi.org/10.1021/acschembio.5b00758
- Rachel Friedman Ohana, Thomas A. Kirkland, Carolyn C. Woodroofe, Sergiy Levin, H. Tetsuo Uyeda, Paul Otto, Robin Hurst, Matthew B. Robers, Kris Zimmerman, Lance P. Encell, and Keith V. Wood . Deciphering the Cellular Targets of Bioactive Compounds Using a Chloroalkane Capture Tag. ACS Chemical Biology 2015, 10
(10)
, 2316-2324. https://doi.org/10.1021/acschembio.5b00351
- Jordan M. Mattheisen, Victoria A. Rasmussen, Emilie Ceraudo, Arielle Kolodzinski, Mizuho Horioka-Duplix, Thomas P. Sakmar, Thomas Huber. Application of bioluminescence resonance energy transfer to quantitate cell-surface expression of membrane proteins. Analytical Biochemistry 2024, 684 , 115361. https://doi.org/10.1016/j.ab.2023.115361
- Alexander H. Swan, Roland F. R. Schindler, Marco Savarese, Isabelle Mayer, Susanne Rinné, Felix Bleser, Anne Schänzer, Andreas Hahn, Mario Sabatelli, Francesco Perna, Kathryn Chapman, Mark Pfuhl, Alan C. Spivey, Niels Decher, Bjarne Udd, Giorgio Tasca, Thomas Brand. Differential effects of mutations of POPDC proteins on heteromeric interaction and membrane trafficking. Acta Neuropathologica Communications 2023, 11
(1)
https://doi.org/10.1186/s40478-022-01501-w
- Nezha S. Benabdallah, Vineet Dalal, R. Wilder Scott, Fady Marcous, Afroditi Sotiriou, Felix K. F. Kommoss, Anastasija Pejkovska, Ludmila Gaspar, Lena Wagner, Francisco J. Sánchez-Rivera, Monica Ta, Shelby Thornton, Torsten O. Nielsen, T. Michael Underhill, Ana Banito. Aberrant gene activation in synovial sarcoma relies on SSX specificity and increased PRC1.1 stability. Nature Structural & Molecular Biology 2023, 30
(11)
, 1640-1652. https://doi.org/10.1038/s41594-023-01096-3
- Younghoon Kim, Pooreum Seo, Eunhye Jeon, Inchul You, Kyubin Hwang, Namkyoung Kim, Jason Tse, Juhyeon Bae, Ha-Soon Choi, Stephen M. Hinshaw, Nathanael S. Gray, Taebo Sim. Targeted kinase degradation via the KLHDC2 ubiquitin E3 ligase. Cell Chemical Biology 2023, 30
(11)
, 1414-1420.e5. https://doi.org/10.1016/j.chembiol.2023.07.008
- Natalia A. Szulc, Małgorzata Piechota, Lilla Biriczová, Pankaj Thapa, Wojciech Pokrzywa. Lysine deserts and cullin-RING ligase receptors: Navigating untrodden paths in proteostasis. iScience 2023, 26
(11)
, 108344. https://doi.org/10.1016/j.isci.2023.108344
- Michael Wichroski, Joseph Benci, Si-Qi Liu, Louis Chupak, Jie Fang, Carolyn Cao, Cindy Wang, Joelle Onorato, Hongchen Qiu, Yongli Shan, Dana Banas, Ryan Powles, Gregory Locke, Abigail Witt, Caitlyn Stromko, Huilin Qi, Xiaofan Zheng, Scott Martin, Min Ding, Robert Gentles, Nicholas Meanwell, Upender Velaparthi, Richard Olson, Susan Wee, Daniel Tenney, Christopher G. Parker, Benjamin F. Cravatt, Michael Lawrence, Robert Borzilleri, Emma Lees. DGKα/ζ inhibitors combine with PD-1 checkpoint therapy to promote T cell–mediated antitumor immunity. Science Translational Medicine 2023, 15
(719)
https://doi.org/10.1126/scitranslmed.adh1892
- Maria Maddalena Calabretta, Elisa Michelini. Current advances in the use of bioluminescence assays for drug discovery: an update of the last ten years. Expert Opinion on Drug Discovery 2023, , 1-11. https://doi.org/10.1080/17460441.2023.2266989
- Stephen H. Kasper, Stephanie Otten, Brian Squadroni, Cionna Orr‐Terry, Yi Kuang, Lily Mussallem, Lan Ge, Lin Yan, Srinivasaraghavan Kannan, Chandra S. Verma, Christopher J. Brown, Charles W. Johannes, David P. Lane, Arun Chandramohan, Anthony W. Partridge, Lee R. Roberts, Hubert Josien, Alex G. Therien, Erik C. Hett, Bonnie J. Howell, Andrea Peier, Xi Ai, Jason Cassaday. A high‐throughput microfluidic mechanoporation platform to enable intracellular delivery of cyclic peptides in cell‐based assays. Bioengineering & Translational Medicine 2023, 8
(5)
https://doi.org/10.1002/btm2.10542
- Russell Spencer-Smith, Deborah K. Morrison. Protocol for measuring BRAF autoinhibition in live cells using a proximity-based NanoBRET assay. STAR Protocols 2023, 4
(3)
, 102461. https://doi.org/10.1016/j.xpro.2023.102461
- Mina Oliayi, Rahman Emamzadeh, Mojgan Rastegar, Mahboobeh Nazari. Tri-part NanoLuc as a new split technology with potential applications in chemical biology: a mini-review. Analytical Methods 2023, 15
(32)
, 3924-3931. https://doi.org/10.1039/D3AY00512G
- Gage O. Leighton, Shengzhe Shang, Sean Hageman, Gordon D. Ginder, David C. Williams. Analysis of the complex between MBD2 and the histone deacetylase core of NuRD reveals key interactions critical for gene silencing. Proceedings of the National Academy of Sciences 2023, 120
(33)
https://doi.org/10.1073/pnas.2307287120
- Alice Lanne, Laura E J Usselmann, Poppy Llowarch, Iacovos N Michaelides, Martin Fillmore, Geoffrey A Holdgate. A perspective on the changing landscape of HTS. Drug Discovery Today 2023, 28
(8)
, 103670. https://doi.org/10.1016/j.drudis.2023.103670
- Kelly A. Teske, Wenji Su, Cesear R. Corona, Jing Wen, Jason Deng, Yan Ping, Zaihong Zhang, Qi Zhang, Jennifer Wilkinson, Michael T. Beck, Kendra R. Nealey, James D. Vasta, Mei Cong, Poncho L. Meisenheimer, Letian Kuai, Matthew B. Robers. DELs enable the development of BRET probes for target engagement studies in cells. Cell Chemical Biology 2023, 30
(8)
, 987-998.e24. https://doi.org/10.1016/j.chembiol.2023.06.019
- Lena Ball, Julia Bauer, Dietmar Krautwurst. Heterodimerization of Chemoreceptors TAS1R3 and mGlu2 in Human Blood Leukocytes. International Journal of Molecular Sciences 2023, 24
(16)
, 12942. https://doi.org/10.3390/ijms241612942
- Katrijn Boon, Nathan Vanalken, Eef Meyen, Dominique Schols, Tom Van Loy. REGA-SIGN: Development of a Novel Set of NanoBRET-Based G Protein Biosensors. Biosensors 2023, 13
(8)
, 767. https://doi.org/10.3390/bios13080767
- Lydia Ogrodzinski, Simon Platt, Joelle Goulding, Cameron Alexander, Tracy D. Farr, Jeanette Woolard, Stephen J. Hill, Laura E. Kilpatrick. Probing expression of E-selectin using CRISPR-Cas9-mediated tagging with HiBiT in human endothelial cells. iScience 2023, 26
(7)
, 107232. https://doi.org/10.1016/j.isci.2023.107232
- Yichao Zhao, Daniel Tabet, Diana Rubio Contreras, Linjiang Lao, Arne Nedergaard Kousholt, Jochen Weile, Henrique Melo, Lisa Hoeg, Sumin Feng, Atina G. Coté, Zhen-Yuan Lin, Dheva Setiaputra, Jos Jonkers, Anne-Claude Gingras, Fernando Gómez Herreros, Frederick P. Roth, Daniel Durocher. Genome-scale mapping of DNA damage suppressors through phenotypic CRISPR-Cas9 screens. Molecular Cell 2023, 14 https://doi.org/10.1016/j.molcel.2023.06.025
- Marieke Van Daele, Laura E Kilpatrick, Jeanette Woolard, Stephen J Hill. Characterisation of tyrosine kinase inhibitor-receptor interactions at VEGFR2 using sunitinib-red and nanoBRET. Biochemical Pharmacology 2023, 25 , 115672. https://doi.org/10.1016/j.bcp.2023.115672
- Kyle R. Christensen, Benjamin Combs, Collin Richards, Tessa Grabinski, Mohammed M. Alhadidy, Nicholas M. Kanaan. Phosphomimetics at Ser199/Ser202/Thr205 in Tau Impairs Axonal Transport in Rat Hippocampal Neurons. Molecular Neurobiology 2023, 60
(6)
, 3423-3438. https://doi.org/10.1007/s12035-023-03281-3
- Micael R. Cunha, Carolina M.C. Catta-Preta, Jéssica E. Takarada, Gabriela A. Moreira, Katlin B. Massirer, Rafael M. Couñago. A novel BRET-based assay to investigate binding and residence time of unmodified ligands to the human lysosomal ion channel TRPML1 in intact cells. Journal of Biological Chemistry 2023, 299
(6)
, 104807. https://doi.org/10.1016/j.jbc.2023.104807
- Remi Janicot, Jong‐Chan Park, Mikel Garcia‐Marcos. Detecting GPCR Signals With Optical Biosensors of Gα‐GTP in Cell Lines and Primary Cell Cultures. Current Protocols 2023, 3
(6)
https://doi.org/10.1002/cpz1.796
- Ismail Dahmani, Kezhen Qin, Youjun Zhang, Alisdair R. Fernie. The formation and function of plant metabolons. The Plant Journal 2023, 114
(5)
, 1080-1092. https://doi.org/10.1111/tpj.16179
- Charles S. Lay, Laura E. Kilpatrick, Peter D. Craggs, Stephen J. Hill. Use of NanoBiT and NanoBRET to characterise interleukin‐23 receptor dimer formation in living cells. British Journal of Pharmacology 2023, 180
(11)
, 1444-1459. https://doi.org/10.1111/bph.16018
- Gaoxian Chen, Detlef Obal. Detecting and measuring of GPCR signaling – comparison of human induced pluripotent stem cells and immortal cell lines. Frontiers in Endocrinology 2023, 14 https://doi.org/10.3389/fendo.2023.1179600
- Xuan Yang, Jeffery L. Smith, Michael T. Beck, Jennifer M. Wilkinson, Ani Michaud, James D. Vasta, Matthew B. Robers, Timothy M. Willson. Development of Cell Permeable NanoBRET Probes for the Measurement of PLK1 Target Engagement in Live Cells. Molecules 2023, 28
(7)
, 2950. https://doi.org/10.3390/molecules28072950
- Taiichiro Shirai, Akiko Nakai, Emiko Ando, Jun Fujimoto, Sarah Leach, Takao Arimori, Daisuke Higo, Floris J. van Eerden, Janyerkye Tulyeu, Yu-Chen Liu, Daisuke Okuzaki, Masanori A. Murayama, Haruhiko Miyata, Kazuto Nunomura, Bangzhong Lin, Akiyoshi Tani, Atsushi Kumanogoh, Masahito Ikawa, James B. Wing, Daron M. Standley, Junichi Takagi, Kazuhiro Suzuki. Celastrol suppresses humoral immune responses and autoimmunity by targeting the COMMD3/8 complex. Science Immunology 2023, 8
(81)
https://doi.org/10.1126/sciimmunol.adc9324
- Paola Martinelli, Otmar Schaaf, Andreas Mantoulidis, Laetitia J. Martin, Julian E. Fuchs, Gerd Bader, Andreas Gollner, Bernhard Wolkerstorfer, Catherine Rogers, Esra Balıkçı, Jesse J. Lipp, Nikolai Mischerikow, Sandra Doebel, Thomas Gerstberger, Wolfgang Sommergruber, Kilian V. M. Huber, Jark Böttcher. Discovery of a Chemical Probe to Study Implications of BPTF Bromodomain Inhibition in Cellular and
in vivo
Experiments. ChemMedChem 2023, 18
(6)
https://doi.org/10.1002/cmdc.202200686
- Jakub Ptacek, Ivan Snajdr, Jiri Schimer, Zsofia Kutil, Jana Mikesova, Petra Baranova, Barbora Havlinova, Werner Tueckmantel, Pavel Majer, Alan Kozikowski, Cyril Barinka. Selectivity of Hydroxamate- and Difluoromethyloxadiazole-Based Inhibitors of Histone Deacetylase 6 In Vitro and in Cells. International Journal of Molecular Sciences 2023, 24
(5)
, 4720. https://doi.org/10.3390/ijms24054720
- Minji Wang, Yifan Da, Yang Tian. Fluorescent proteins and genetically encoded biosensors. Chemical Society Reviews 2023, 52
(4)
, 1189-1214. https://doi.org/10.1039/D2CS00419D
- Bolormaa Baljinnyam, Michael Ronzetti, Anton Simeonov. Advances in luminescence-based technologies for drug discovery. Expert Opinion on Drug Discovery 2023, 18
(1)
, 25-35. https://doi.org/10.1080/17460441.2023.2160441
- Ilana B. Kotliar, Emily Lorenzen, Jochen M. Schwenk, Debbie L. Hay, Thomas P. Sakmar, . Elucidating the Interactome of G Protein-Coupled Receptors and Receptor Activity-Modifying Proteins. Pharmacological Reviews 2023, 75
(1)
, 1-34. https://doi.org/10.1124/pharmrev.120.000180
- Xin Yu, Jin Wang. Quantitative measurement of PROTAC intracellular accumulation. 2023, 189-214. https://doi.org/10.1016/bs.mie.2022.11.001
- Magdalena M. Szewczyk, Dominic D. G. Owens, Dalia Barsyte-Lovejoy. Measuring Protein–Protein Interactions in Cells using Nanoluciferase Bioluminescence Resonance Energy Transfer (NanoBRET) Assay. 2023, 137-148. https://doi.org/10.1007/978-1-0716-3397-7_10
- Matilda Bingham, Thomas Pesnot, Andrew D. Scott. Biophysical screening and characterisation in medicinal chemistry. 2023, 61-104. https://doi.org/10.1016/bs.pmch.2023.10.002
- Anne Sophie Czerniak, Kevin Kretschmer, Tina Weiß, Annette G. Beck‐Sickinger. The Chemerin Receptor CMKLR1 Requires Full‐Length Chemerin for High Affinity in Contrast to GPR1 as Demonstrated by a New Nanoluciferase‐Based Binding Assay. ChemMedChem 2022, 17
(23)
https://doi.org/10.1002/cmdc.202200413
- Victoria Elisabeth Groß, Miron Mikhailowitsch Gershkovich, Torsten Schöneberg, Anette Kaiser, Simone Prömel. NanoBRET in C. elegans illuminates functional receptor interactions in real time. BMC Molecular and Cell Biology 2022, 23
(1)
https://doi.org/10.1186/s12860-022-00405-w
- Anupriya M. Geethakumari, Wesam S. Ahmed, Saad Rasool, Asma Fatima, S. M. Nasir Uddin, Mustapha Aouida, Kabir H. Biswas. A genetically encoded BRET-based SARS-CoV-2 Mpro protease activity sensor. Communications Chemistry 2022, 5
(1)
https://doi.org/10.1038/s42004-022-00731-2
- Olivier A. Pierrat, Manjuan Liu, Gavin W. Collie, Kartika Shetty, Matthew J. Rodrigues, Yann-Vaï Le Bihan, Emma A. Gunnell, P. Craig McAndrew, Mark Stubbs, Martin G. Rowlands, Norhakim Yahya, Erald Shehu, Rachel Talbot, Lisa Pickard, Benjamin R. Bellenie, Kwai-Ming J. Cheung, Ludovic Drouin, Paolo Innocenti, Hannah Woodward, Owen A. Davis, Matthew G. Lloyd, Ana Varela, Rosemary Huckvale, Fabio Broccatelli, Michael Carter, David Galiwango, Angela Hayes, Florence I. Raynaud, Christopher Bryant, Steven Whittaker, Olivia W. Rossanese, Swen Hoelder, Rosemary Burke, Rob L. M. van Montfort. Discovering cell-active BCL6 inhibitors: effectively combining biochemical HTS with multiple biophysical techniques, X-ray crystallography and cell-based assays. Scientific Reports 2022, 12
(1)
https://doi.org/10.1038/s41598-022-23264-z
- Harsimran Kaur Garcha, Nabanita Nawar, Helena Sorger, Fettah Erdogan, Myint Myat Khine Aung, Abootaleb Sedighi, Pimyupa Manaswiyoungkul, Hyuk-Soo Seo, Susann Schönefeldt, Daniel Pölöske, Sirano Dhe-Paganon, Heidi A. Neubauer, Satu M. Mustjoki, Marco Herling, Elvin D. de Araujo, Richard Moriggl, Patrick T. Gunning. High Efficacy and Drug Synergy of HDAC6-Selective Inhibitor NN-429 in Natural Killer (NK)/T-Cell Lymphoma. Pharmaceuticals 2022, 15
(11)
, 1321. https://doi.org/10.3390/ph15111321
- N. Connor Payne, Ralph Mazitschek. Tiny Titans: Nanobodies as Powerful Tools for TR‐FRET Assay Development. Analysis & Sensing 2022, 2
(6)
https://doi.org/10.1002/anse.202200020
- Natasha C. Dale, Carl W. White, Elizabeth K.M. Johnstone, Kevin D.G. Pfleger. Bioluminescence Resonance Energy Transfer (
BRET
) Technologies to Study
GPCRs. 2022, 841-873. https://doi.org/10.1002/9781119564782.ch23
- Caroline K. Brennan, Zi Yao, Anastasia A. Ionkina, Colin M. Rathbun, Buvaneshwari Sathishkumar, Jennifer A. Prescher. Multiplexed bioluminescence imaging with a substrate unmixing platform. Cell Chemical Biology 2022, 29
(11)
, 1649-1660.e4. https://doi.org/10.1016/j.chembiol.2022.10.004
- Russell Spencer-Smith, Elizabeth M. Terrell, Christine Insinna, Constance Agamasu, Morgan E. Wagner, Daniel A. Ritt, Jim Stauffer, Andrew G. Stephen, Deborah K. Morrison. RASopathy mutations provide functional insight into the BRAF cysteine-rich domain and reveal the importance of autoinhibition in BRAF regulation. Molecular Cell 2022, 82
(22)
, 4262-4276.e5. https://doi.org/10.1016/j.molcel.2022.10.016
- Kelvin F. Cho, Noelle Javier, Kaylee Choi. BRET measurement on CCD camera-based microtiter plate readers. SLAS Discovery 2022, 27
(7)
, 413-417. https://doi.org/10.1016/j.slasd.2022.08.002
Abstract
Figure 1
Figure 1. Comparison of different acceptors for NanoBRET. (a) BRET spectra for Nluc-HT (black) and Nluc-HT conjugated to HaloTag Oregon Green (green), HaloTag TMR (red), or HaloTag NCT (blue) ligands. (b) Correlation of BRET with fractional occupancy. Differing degrees of occupancy were created by combining Nluc-HT-NCT (fully acceptor occupied) with Nluc-HT-PEG-biotin (unoccupied donor) in defined ratios at a constant donor concentration. Fractional occupancy (%) indicates the amount of acceptor-occupied Nluc-HT relative to the total amount of Nluc-HT in the sample. Shown are the means ± SD (n = 7) of a representative experiment. The results were plotted as mBRET units.
Figure 2
Figure 2. Schematic of NanoBRET for detecting protein–protein interactions.
Figure 3
Figure 3. Comparative analysis of BRET1 and NanoBRET using the Frb/FKBP model system. (a) Optimization of donor and acceptor orientation for the Frb/FKBP interaction. BRET ratios were determined on HEK293 cells transiently transfected with the indicated Frb and FKBP fusion proteins at a donor/acceptor ratio of 1:4, each measured using untreated and rapamycin treated (1 μM, 15 min) samples. The optimal configuration for either BRET1 or NanoBRET is indicated by the hatch pattern. (b) HEK293 cells were transiently transfected with either Frb-Nluc/FKBP-HT or Frb-RLuc8/FKBP-turboYFP at a donor/acceptor ratio of 1:4. The cells were plated in a 384-well plate and left untreated (n = 3) or treated with the indicated concentration of rapamycin (n = 3) for 15 min before determining the BRET ratio.
Figure 4
Figure 4. Effect of expression level on assay sensitivity for detecting the interaction of FKBP with Frb. HeLa cells were transfected with a serial dilution of expression constructs for (a) Frb-Nluc and FKBP-HT or (b) Frb-Rluc8 and FKBP-TurboYFP (1:1 ratio) and plated in thre 96-well plates. BRET ratios were determined for untreated or rapamycin treated samples (1 μM rapamycin, 15 min). Shown is a representative experiment with all individual data points (n = 12) as well as mean ± SD plotted against DNA dilution used for transfection.
Figure 5
Figure 5. NanoBRET assays for bromodomain protein interactions with chromatin. NanoBRET assays were developed to measure the binding of Nluc-BRD4 (a) or Nluc-CBP (b) to Histone H3.3-HT. HEK293 cells were transfected with the indicated combination of constructs. The BRET ratio for each sample was determined following treatment for 18 h with the indicated concentrations of I-BET151. Shown are the means ± SEM of three independent experiments performed in quadruplicate.
Figure 6
Figure 6. Real time imaging of ligand induced interaction of AVPR2-HT with Nluc-β-arrestin2 interaction. HeLa cells were transfected with expression constructs for AVPR2-HT and Nluc-β-arrestin2. Sequential acquisition of images was initiated under unstimulated conditions. After establishing a baseline for seven acquisition cycles, 1 μM arginine vasopressin was added to the sample by injection followed by acquisition cycles (13.4 s per cycle) covering approximately 20 min. (a) The panel shows pseudocolored images of the donor (cyan) and acceptor (yellow) channel at indicated time points following treatment with AVP. The images are representative of the results obtained in five independent experiments (scale bar = 10 μm). (b) Kinetic of Nluc-β-arrestin2 recruitment following stimulation with AVP measured by imaging or in plate reader. The imaging data represent five individual regions of interest (ROI), each representing a single cell. The data shown for the plate based assay format are the results of a representative experiment performed in triplicate. In order to compare results obtained from different analytical platforms, the data were plotted as relative change in BRET normalized to the maximum BRET ratio obtained for each data set.
References
ARTICLE SECTIONSThis article references 25 other publications.
- 1Ciruela, F. (2008) Fluorescence-based methods in the study of protein-protein interactions in living cells Curr. Opin. Biotechnol. 19, 338– 343Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtValsL7K&md5=1e7c77ca817344a77800b02aa7052791Fluorescence-based methods in the study of protein-protein interactions in living cellsCiruela, FranciscoCurrent Opinion in Biotechnology (2008), 19 (4), 338-343CODEN: CUOBE3; ISSN:0958-1669. (Elsevier B.V.)A review. Multiprotein complexes partake in nearly all cell functions, thus the characterization and visualization of protein-protein interactions in living cells constitute an important step in the study of a large array of cellular mechanisms. Recently, noninvasive fluorescence-based methods using resonance energy transfer (RET), namely bioluminescence-RET (BRET) and fluorescence-RET (FRET), and those centered on protein fragment complementation, such as bimol. fluorescence complementation (BiFC), have been successfully used in the study of protein interactions. These new technologies are nowadays the most powerful approaches for visualizing the interactions occurring within protein complexes in living cells, thus enabling the investigation of protein behavior in their normal milieu. Here we address the individual strengths and weaknesses of these methods when applied to the study of protein-protein interactions.
- 2Angers, S., Salahpour, A., Joly, E., Hilairet, S., Chelsky, D., Dennis, M., and Bouvier, M. (2000) Detection of beta 2-adrenergic receptor dimerization in living cells using bioluminescence resonance energy transfer (BRET) Proc. Natl. Acad. Sci. U.S.A. 97, 3684– 3689Google ScholarThere is no corresponding record for this reference.
- 3Xu, Y., Piston, D. W., and Johnson, C. H. (1999) A bioluminescence resonance energy transfer (BRET) system: application to interacting circadian clock proteins Proc. Natl. Acad. Sci. U.S.A. 96, 151– 156Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXjvV2itw%253D%253D&md5=eb87cabdad3ba1ee84b52692e4ef8f20A bioluminescence resonance energy transfer (BRET) system: application to interacting circadian clock proteinsXu, Yao; Piston, David W.; Johnson, Carl HirschieProceedings of the National Academy of Sciences of the United States of America (1999), 96 (1), 151-156CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)We describe a method for assaying protein interactions that offers some attractive advantages over previous assays. This method, called bioluminescence resonance energy transfer (BRET), uses a bioluminescent luciferase that is genetically fused to one candidate protein, and a green fluorescent protein mutant fused to another protein of interest. Interactions between the two fusion proteins can bring the luciferase and green fluorescent protein close enough for resonance energy transfer to occur, thus changing the color of the bioluminescent emission. By using proteins encoded by circadian (daily) clock genes from cyanobacteria, we use the BRET technique to demonstrate that the clock protein KaiB interacts to form homodimers. BRET should be particularly useful for testing protein interactions within native cells, esp. with integral membrane proteins or proteins targeted to specific organelles.
- 4Wampler, J. E., Hori, K., Lee, J. W., and Cormier, M. J. (1971) Structured bioluminescence. Two emitters during both the in vitro and the in vivo bioluminescence of the sea pansy, Renilla Biochemistry 10, 2903– 2909Google ScholarThere is no corresponding record for this reference.
- 5Pfleger, K. D. and Eidne, K. A. (2006) Illuminating insights into protein-protein interactions using bioluminescence resonance energy transfer (BRET) Nat. Methods 3, 165– 174Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhsFOru7k%253D&md5=bc83e4f90f5ab10ed12286eb20849cccIlluminating insights into protein-protein interactions using bioluminescence resonance energy transfer (BRET)Pfleger, Kevin D. G.; Eidne, Karin A.Nature Methods (2006), 3 (3), 165-174CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)A review. Bioluminescence resonance energy transfer (BRET) is a straightforward biophys. technique for studying protein-protein interactions. It requires: (1) that proteins of interest and suitable controls be labeled with either a donor or acceptor mol., (2) placement of these labeled proteins in the desired environment for assessing their potential interaction, and (3) use of suitable detection instrumentation to monitor resultant energy transfer. There are now several possible applications, combinations of donor and acceptor mols., potential assay environments and detection system perturbations. Therefore, this review aims to demystify and clarify the important aspects of the BRET methodol. that should be considered when using this technique.
- 6Bertrand, L., Parent, S., Caron, M., Legault, M., Joly, E., Angers, S., Bouvier, M., Brown, M., Houle, B., and Menard, L. (2002) The BRET2/arrestin assay in stable recombinant cells: a platform to screen for compounds that interact with G protein-coupled receptors (GPCRS) J. Recept. Signal Transduction Res. 22, 533– 541Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38Xps1ejsbY%253D&md5=a2b21a28d1737f18c064989299e33086The BRET2/arrestin assay in stable recombinant cells: a platform to screen for compounds that interact with G protein-coupled receptors (GPCRS)Bertrand, Lucie; Parent, Stephane; Caron, Mireille; Legault, Mireille; Joly, Erik; Angers, Stephane; Bouvier, Michel; Brown, Mike; Houle, Benoit; Menard, LucJournal of Receptors and Signal Transduction (2002), 22 (1-4), 533-541CODEN: JRSTCT ISSN:. (Marcel Dekker, Inc.)In BRET2 (Bioluminescence Resonance Energy Transfer), a Renilla luciferase (RLuc) is used as the donor protein, while a Green Fluorescent Protein (GFP2) is used as the acceptor protein. In the presence of the cell permeable substrate DeepBlueC, RLuc emits blue light at 395 nm. If the GFP2 is brought into close proximity to RLuc via a specific biomol. interaction, the GFP2 will absorb the blue light energy and reemit green light at 510 nm. BRET2 signals are therefore easily detd. by measuring the ratio of green over blue light (510/395 nm) using appropriate dual channel luminometry instruments (e.g., Fusion Universal Microplate Analyzer, Packard BioScience). Since no light source is required for BRET2 assays, the technol. does not suffer from high fluorescent background or photobleaching, the common problems assocd. with std. FRET-based assays. Using BRET2, the authors developed a generic G Protein-Coupled Receptor (GPCR) assay based on the observation that activation of the majority of GPCRs by agonists leads to the interaction of β-arrestin (a protein that is involved in receptor desensitization and sequestration) with the receptor. The authors established a cell line stably expressing the GFP2:β-arrestin 2 fusion protein, and showed that it can be used to monitor the activation of various transiently expressed GPCRs, in BRET2/arrestin assays. In addn., using the HEK 293/GFP2:β-arrestin 2 cell line as a recipient, the authors generated a double-stable line co-expressing the vasopressin 2 receptor (V2R) fused to RLuc (V2R:RLuc) and used it for the pharmacol. characterization of compds. in BRET2/arrestin assays. This approach yields genuine pharmacol. and supports the BRET2/arrestin assay as a tool that can be used with recombinant cell lines to characterize ligand-GPCR interactions which can be applied to ligand identification for orphan receptors.
- 7Hamdan, F. F., Audet, M., Garneau, P., Pelletier, J., and Bouvier, M. (2005) High-throughput screening of G protein-coupled receptor antagonists using a bioluminescence resonance energy transfer 1-based beta-arrestin2 recruitment assay J. Biomol. Screening 10, 463– 475Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhtVers77J&md5=40aadd9c704f1eee984a2795c374e60aHigh-throughput screening of G protein-coupled receptor antagonists using a bioluminescence resonance energy transfer 1-based β-arrestin2 recruitment assayHamdan, Fadi F.; Audet, Martin; Garneau, Philippe; Pelletier, Jerry; Bouvier, MichelJournal of Biomolecular Screening (2005), 10 (5), 463-475CODEN: JBISF3; ISSN:1087-0571. (Sage Publications)In this study, the authors developed HEK293 cell lines that stably coexpressed optimal amts. of β-arrestin2-Rluc and VENUS fusions of G protein-coupled receptors (GPCRs) belonging to both class A and class B receptors, which include receptors that interact transiently or stably with β-arrestins. This allowed the use of a bioluminescence resonance energy transfer (BRET) 1-β-arrestin2 translocation assay to quantify receptor activation or inhibition. One of the developed cell lines coexpressing CCR5-VENUS and β-arrestin2-/Renilla luciferase was then used for high-throughput screening (HTS) for antagonists of the chemokine receptor CCR5, the primary co-receptor for HIV. A total of 26,000 compds. were screened for inhibition of the agonist-promoted β-arrestin2 recruitment to CCR5, and 12 compds. were found to specifically inhibit the agonist-induced β-arrestin2 recruitment to CCR5. Three of the potential hits were further tested using other functional assays, and their abilities to inhibit CCR5 agonist-promoted signaling were confirmed. This is the 1st study describing a BRET1-β-arrestin recruitment assay in stable mammalian cells and its successful application in HTS for GPCRs antagonists.
- 8Loening, A. M., Fenn, T. D., Wu, A. M., and Gambhir, S. S. (2006) Consensus guided mutagenesis of Renilla luciferase yields enhanced stability and light output Protein Eng., Des. Sel. 19, 391– 400Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xotl2ksro%253D&md5=c006a4ff7a42c04c1ccf02b80fb1c9ffConsensus guided mutagenesis of Renilla luciferase yields enhanced stability and light outputLoening, Andreas Markus; Fenn, Timothy David; Wu, Anna M.; Gambhir, Sanjiv SamProtein Engineering, Design & Selection (2006), 19 (9), 391-400CODEN: PEDSBR; ISSN:1741-0126. (Oxford University Press)Luciferases, which have seen expansive employment as reporter genes in biol. research, could also be used in applications where the protein itself is conjugated to ligands to create probes that are appropriate for use in small animal imaging. As the bioluminescence activity of commonly used luciferases is too labile in serum to permit this application, specific mutations of Renilla luciferase, selected using a consensus sequence driven strategy, were screened for their ability to confer stability of activity in serum as well as their light output. Using this information, a total of eight favorable mutations were combined to generate a mutant Renilla luciferase (RLuc8) that, compared with the parental enzyme, is 200-fold more resistant to inactivation in murine serum and exhibits a 4-fold improvement in light output. Results of the mutational anal. were also used to generate a double mutant optimized for use as a reporter gene. The double mutant had half the resistance to inactivation in serum of the native enzyme while yielding a 5-fold improvement in light output.These variants of Renilla luciferase, which exhibit significantly improved properties compared with the native enzyme, will allow enhanced sensitivity in existing luciferase-based assays as well as enable the development of novel probes labeled with the luciferase protein.
- 9Kocan, M., See, H. B., Seeber, R. M., Eidne, K. A., and Pfleger, K. D. (2008) Demonstration of improvements to the bioluminescence resonance energy transfer (BRET) technology for the monitoring of G protein-coupled receptors in live cells J. Biomol. Screening 13, 888– 898Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjtVymtr0%253D&md5=52bd4ecaa2dd5f265e7a02deeb407556Demonstration of improvements to the bioluminescence resonance energy transfer (BRET) technology for the monitoring of G protein-coupled receptors in live cellsKocan, Martina; See, Heng B.; Seeber, Ruth M.; Eidne, Karin A.; Pfleger, Kevin D. G.Journal of Biomolecular Screening (2008), 13 (9), 888-898CODEN: JBISF3; ISSN:1087-0571. (Sage Publications)The bioluminescence resonance energy transfer (BRET) technique has become extremely popular for studying protein-protein interactions in living cells and real time. Of particular interest is the ability to monitor interactions between G protein-coupled receptors, such as the TSH-releasing hormone receptor (TRHR), and proteins crit. for regulating their function, such as β-arrestin. Using TRHR/β-arrestin interactions, we have demonstrated improvements to all 3 generations of BRET (BRET, BRET, and eBRET) by using the novel forms of luciferase, Rluc2 and Rluc8, developed by the Gambhir lab. Furthermore, for the 1st time it was possible to use the BRET2 system to detect ligand-induced G protein-coupled receptor/β-arrestin interactions over prolonged periods (on the scale of hours rather than seconds) with a very stable signal. As demonstrated by our Z'-factor data, these luciferases increase the sensitivity of BRET to such an extent that they substantially increase the potential applicability of this technol. for effective drug discovery high-throughput screening.
- 10Hall, M. P., Unch, J., Binkowski, B. F., Valley, M. P., Butler, B. L., Wood, M. G., Otto, P., Zimmerman, K., Vidugiris, G., Machleidt, T., Robers, M. B., Benink, H. A., Eggers, C. T., Slater, M. R., Meisenheimer, P. L., Klaubert, D. H., Fan, F., Encell, L. P., and Wood, K. V. (2012) Engineered luciferase reporter from a deep sea shrimp utilizing a novel imidazopyrazinone substrate ACS Chem. Biol. 7, 1848– 1857Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xht1Smu7%252FN&md5=0581a7e44935671471577ade1ff538ebEngineered Luciferase Reporter from a Deep Sea Shrimp Utilizing a Novel Imidazopyrazinone SubstrateHall, Mary P.; Unch, James; Binkowski, Brock F.; Valley, Michael P.; Butler, Braeden L.; Wood, Monika G.; Otto, Paul; Zimmerman, Kristopher; Vidugiris, Gediminas; Machleidt, Thomas; Robers, Matthew B.; Benink, Helene A.; Eggers, Christopher T.; Slater, Michael R.; Meisenheimer, Poncho L.; Klaubert, Dieter H.; Fan, Frank; Encell, Lance P.; Wood, Keith V.ACS Chemical Biology (2012), 7 (11), 1848-1857CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Bioluminescence methodologies have been extraordinarily useful due to their high sensitivity, broad dynamic range, and operational simplicity. These capabilities have been realized largely through incremental adaptations of native enzymes and substrates, originating from luminous organisms of diverse evolutionary lineages. We engineered both an enzyme and substrate in combination to create a novel bioluminescence system capable of more efficient light emission with superior biochem. and phys. characteristics. Using a small luciferase subunit (19 kDa) from the deep sea shrimp Oplophorus gracilirostris, we have improved luminescence expression in mammalian cells ∼2.5 million-fold by merging optimization of protein structure with development of a novel imidazopyrazinone substrate (furimazine). The new luciferase, NanoLuc, produces glow-type luminescence (signal half-life >2 h) with a specific activity ∼150-fold greater than that of either firefly (Photinus pyralis) or Renilla luciferases similarly configured for glow-type assays. In mammalian cells, NanoLuc shows no evidence of post-translational modifications or subcellular partitioning. The enzyme exhibits high phys. stability, retaining activity with incubation up to 55 °C or in culture medium for >15 h at 37 °C. As a genetic reporter, NanoLuc may be configured for high sensitivity or for response dynamics by appending a degrdn. sequence to reduce intracellular accumulation. Appending a signal sequence allows NanoLuc to be exported to the culture medium, where reporter expression can be measured without cell lysis. Fusion onto other proteins allows luminescent assays of their metab. or localization within cells. Reporter quantitation is achievable even at very low expression levels to facilitate more reliable coupling with endogenous cellular processes.
- 11Los, G. V., Encell, L. P., McDougall, M. G., Hartzell, D. D., Karassina, N., Zimprich, C., Wood, M. G., Learish, R., Ohana, R. F., Urh, M., Simpson, D., Mendez, J., Zimmerman, K., Otto, P., Vidugiris, G., Zhu, J., Darzins, A., Klaubert, D. H., Bulleit, R. F., and Wood, K. V. (2008) HaloTag: a novel protein labeling technology for cell imaging and protein analysis ACS Chem. Biol. 3, 373– 382Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXmvVGru7k%253D&md5=66e11d2da0992e634150392ea9575f75HaloTag: A Novel Protein Labeling Technology for Cell Imaging and Protein AnalysisLos, Georgyi V.; Encell, Lance P.; McDougall, Mark G.; Hartzell, Danette D.; Karassina, Natasha; Zimprich, Chad; Wood, Monika G.; Learish, Randy; Ohana, Rachel Friedman; Urh, Marjeta; Simpson, Dan; Mendez, Jacqui; Zimmerman, Kris; Otto, Paul; Vidugiris, Gediminas; Zhu, Ji; Darzins, Aldis; Klaubert, Dieter H.; Bulleit, Robert F.; Wood, Keith V.ACS Chemical Biology (2008), 3 (6), 373-382CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)We have designed a modular protein tagging system that allows different functionalities to be linked onto a single genetic fusion, either in soln., in living cells, or in chem. fixed cells. The protein tag (HaloTag) is a modified haloalkane dehalogenase designed to covalently bind to synthetic ligands (HaloTag ligands). The synthetic ligands comprise a chloroalkane linker attached to a variety of useful mols., such as fluorescent dyes, affinity handles, or solid surfaces. Covalent bond formation between the protein tag and the chloroalkane linker is highly specific, occurs rapidly under physiol. conditions, and is essentially irreversible. We demonstrate the utility of this system for cellular imaging and protein immobilization by analyzing multiple mol. processes assocd. with NF-κB-mediated cellular physiol., including imaging of subcellular protein translocation and capture of protein-protein and protein-DNA complexes.
- 12Shagin, D. A., Barsova, E. V., Yanushevich, Y. G., Fradkov, A. F., Lukyanov, K. A., Labas, Y. A., Semenova, T. N., Ugalde, J. A., Meyers, A., Nunez, J. M., Widder, E. A., Lukyanov, S. A., and Matz, M. V. (2004) GFP-like proteins as ubiquitous metazoan superfamily: evolution of functional features and structural complexity Mol. Biol. Evol. 21, 841– 850Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjsFyqsrs%253D&md5=3b2ea95fc034e2485d9c5b76e32b71e1GFP-like proteins as ubiquitous metazoan superfamily: evolution of functional features and structural complexityShagin, Dmitry A.; Barsova, Ekaterina V.; Yanushevich, Yurii G.; Fradkov, Arkady F.; Lukyanov, Konstantin A.; Labas, Yulii A.; Semenova, Tatiana N.; Ugalde, Juan A.; Meyers, Ann; Nunez, Jose M.; Widder, Edith A.; Lukyanov, Sergey A.; Matz, Mikhail V.Molecular Biology and Evolution (2004), 21 (5), 841-850CODEN: MBEVEO; ISSN:0737-4038. (Oxford University Press)Homologs of the green fluorescent protein (GFP), including the recently described GFP-like domains of certain extracellular matrix proteins in Bilaterian organisms, are remarkably similar at the protein structure level, yet they often perform totally unrelated functions, thereby warranting recognition as a superfamily. Here we describe diverse GFP-like proteins from previously undersampled and completely new sources, including hydromedusae and planktonic Copepoda. In hydromedusae, yellow and nonfluorescent purple proteins were found in addn. to greens. Notably, the new yellow protein seems to follow exactly the same structural soln. to achieving the yellow color of fluorescence as YFP, an engineered yellow-emitting mutant variant of GFP. The addn. of these new sequences made it possible to resolve deep-level phylogenetic relationships within the superfamily. Fluorescence (most likely green) must have already existed in the common ancestor of Cnidaria and Bilateria, and therefore GFP-like proteins may be responsible for fluorescence and/or coloration in virtually any animal. At least 15 color diversification events can be inferred following the max. parsimony principle in Cnidaria. Origination of red fluorescence and nonfluorescent purple-blue colors on several independent occasions provides a remarkable example of convergent evolution of complex features at the mol. level.
- 13Merzlyak, E. M., Goedhart, J., Shcherbo, D., Bulina, M. E., Shcheglov, A. S., Fradkov, A. F., Gaintzeva, A., Lukyanov, K. A., Lukyanov, S., Gadella, T. W., and Chudakov, D. M. (2007) Bright monomeric red fluorescent protein with an extended fluorescence lifetime Nat. Methods 4, 555– 557Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXntVCitLk%253D&md5=3351e2581f2ff447cc1f114b7c91cc46Bright monomeric red fluorescent protein with an extended fluorescence lifetimeMerzlyak, Ekaterina M.; Goedhart, Joachim; Shcherbo, Dmitry; Bulina, Mariya E.; Shcheglov, Aleksandr S.; Fradkov, Arkady F.; Gaintzeva, Anna; Lukyanov, Konstantin A.; Lukyanov, Sergey; Gadella, Theodorus W. J.; Chudakov, Dmitriy M.Nature Methods (2007), 4 (7), 555-557CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Fluorescent proteins have become extremely popular tools for in vivo imaging and esp. for the study of localization, motility and interaction of proteins in living cells. Here the authors report TagRFP, a monomeric red fluorescent protein, which is characterized by high brightness, complete chromophore maturation, prolonged fluorescence lifetime and high pH-stability. These properties make TagRFP an excellent tag for protein localization studies and fluorescence resonance energy transfer (FRET) applications.
- 14Nagai, T., Ibata, K., Park, E. S., Kubota, M., Mikoshiba, K., and Miyawaki, A. (2002) A variant of yellow fluorescent protein with fast and efficient maturation for cell-biological applications Nat. Biotechnol. 20, 87– 90Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XislSrsQ%253D%253D&md5=1eae1970c042f58236113702a2f660d0A variant of yellow fluorescent protein with fast and efficient maturation for cell-biological applicationsNagai, Takeharu; Ibata, Keiji; Park, Eun Sun; Kubota, Mie; Mikoshiba, Katsuhiko; Miyawaki, AtsushiNature Biotechnology (2002), 20 (1), 87-90CODEN: NABIF9; ISSN:1087-0156. (Nature America Inc.)The green fluorescent protein (GFP) from the jellyfish Aequorea victoria has provided a myriad of applications for biol. systems. Over the last several years, mutagenesis studies have improved folding properties of GFP (refs. 1,2). However, slow maturation is still a big obstacle to the use of GFP variants for visualization. These problems are exacerbated when GFP variants are expressed at 37° and/or targeted to certain organelles. Thus, obtaining GFP variants that mature more efficiently is crucial for the development of expanded research applications. Among Aequorea GFP variants, yellow fluorescent proteins (YFPs) are relatively acid-sensitive, and uniquely quenched by chloride ion (Cl-). For YFP to be fully and stably fluorescent, mutations that decrease the sensitivity to both pH and Cl- are desired. Here we describe the development of an improved version of YFP named "Venus". Venus contains a novel mutation, F46L, which at 37° greatly accelerates oxidn. of the chromophore, the rate-limiting step of maturation. As a result of other mutations, F64L/M153T/V163A/S175G, Venus folds well and is relatively tolerant of exposure to acidosis and Cl-. We succeeded in efficiently targeting a neuropeptide Y-Venus fusion protein to the dense-core granules of PC12 cells. Its secretion was readily monitored by measuring release of fluorescence into the medium. The use of Venus as an acceptor allowed early detection of reliable signals of fluorescence resonance energy transfer (FRET) for Ca2+ measurements in brain slices. With the improved speed and efficiency of maturation and the increased resistance to environment, Venus will enable fluorescent labeling that were not possible before.
- 15Shaner, N. C., Steinbach, P. A., and Tsien, R. Y. (2005) A guide to choosing fluorescent proteins Nat. Methods 2, 905– 909Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXht1aksr3F&md5=2aefee1269ed0f92888a47cedd53dcabA guide to choosing fluorescent proteinsShaner, Nathan C.; Steinbach, Paul A.; Tsien, Roger Y.Nature Methods (2005), 2 (12), 905-909CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)The recent explosion in the diversity of available fluorescent proteins (FPs) promises a wide variety of new tools for biol. imaging. With no unified std. for assessing these tools, however, a researcher is faced with difficult questions. Which FPs are best for general use. Which are the brightest. What addnl. factors det. which are best for a given expt.. Although in many cases, a trial-and-error approach may still be necessary in detg. the answers to these questions, a unified characterization of the best available FPs provides a useful guide in narrowing down the options.
- 16Galarneau, A., Primeau, M., Trudeau, L. E., and Michnick, S. W. (2002) Beta-lactamase protein fragment complementation assays as in vivo and in vitro sensors of protein protein interactions Nat. Biotechnol. 20, 619– 622Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XktFOhsrY%253D&md5=284ef04b2969d864811bab12a6de3e4bβ-Lactamase protein fragment complementation assays as in vivo and in vitro sensors of protein-protein interactionsGalarneau, Andre; Primeau, Martin; Trudeau, Louis-Eric; Michnick, Stephen W.Nature Biotechnology (2002), 20 (6), 619-622CODEN: NABIF9; ISSN:1087-0156. (Nature America Inc.)We have previously described a strategy for detecting protein-protein interactions based on protein interaction-assisted folding of rationally designed fragments of enzymes. We call this strategy the protein fragment complementation assay (PCA). Here we describe PCAs based on the enzyme TEM-1 β-lactamase (EC: 3.5.2.6), which include simple colorimetric in vitro assays using the cephalosporin nitrocefin and assays in intact cells using the fluorescent substrate CCF2/AM (ref. 6). Constitutive protein-protein interactions of the GCN4 leucine zippers and of apoptotic proteins Bcl2 and Bad, and the homodimerization of Smad3, were tested in an in vitro assay using cell lysates. With the same in vitro assay, we also demonstrate interactions of protein kinase PKB with substrate Bad. The in vitro assay is facile and amenable to high-throughput modes of screening with signal-to-background ratios in the range of 10:1 to 250:1, which is superior to other PCAs developed to date. Furthermore, we show that the in vitro assay can be used for quant. anal. of a small mol.-induced protein interaction, the rapamycin-induced interaction of FKBP and yeast FRB (the FKBP-rapamycin binding domain of TOR (target of rapamycin)). The assay reproduces the known dissocn. const. and no. of sites for this interaction. The combination of in vitro colorimetric and in vivo fluorescence assays of β-lactamase in mammalian cells suggests a wide variety of sensitive and high-throughput large-scale applications, including in vitro protein array anal. of protein-protein or enzyme-protein interactions and in vivo applications such as clonal selection for cells expressing interacting protein partners.
- 17Zhang, J. H., Chung, T. D., and Oldenburg, K. R. (1999) A Simple Statistical Parameter for Use in Evaluation and Validation of High Throughput Screening Assays J. Biomol. Screening 4, 67– 73Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2sbit12juw%253D%253D&md5=0ec4ed2ccdd3b4a16521567a0975d31fA Simple Statistical Parameter for Use in Evaluation and Validation of High Throughput Screening AssaysZhang; Chung; OldenburgJournal of biomolecular screening (1999), 4 (2), 67-73 ISSN:.The ability to identify active compounds (3hits2) from large chemical libraries accurately and rapidly has been the ultimate goal in developing high-throughput screening (HTS) assays. The ability to identify hits from a particular HTS assay depends largely on the suitability or quality of the assay used in the screening. The criteria or parameters for evaluating the 3suitability2 of an HTS assay for hit identification are not well defined and hence it still remains difficult to compare the quality of assays directly. In this report, a screening window coefficient, called 3Z-factor,2 is defined. This coefficient is reflective of both the assay signal dynamic range and the data variation associated with the signal measurements, and therefore is suitable for assay quality assessment. The Z-factor is a dimensionless, simple statistical characteristic for each HTS assay. The Z-factor provides a useful tool for comparison and evaluation of the quality of assays, and can be utilized in assay optimization and validation.
- 18Wells, J. A. and McClendon, C. L. (2007) Reaching for high-hanging fruit in drug discovery at protein-protein interfaces Nature 450, 1001– 1009Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhsVaqtr7F&md5=e7ed69fd8c362a71c4b99f029c16a6fcReaching for high-hanging fruit in drug discovery at protein-protein interfacesWells, James A.; McClendon, Christopher L.Nature (London, United Kingdom) (2007), 450 (7172), 1001-1009CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)A review. Targeting the interfaces between proteins has huge therapeutic potential, but discovering small-mol. drugs that disrupt protein-protein interactions is an enormous challenge. Several recent success stories, however, indicate that protein-protein interfaces might be more tractable than has been thought. These studies discovered small mols. that bind with drug-like potencies to 'hotspots' on the contact surfaces involved in protein-protein interactions. Remarkably, these small mols. bind deeper within the contact surface of the target protein, and bind with much higher efficiencies, than do the contact atoms of the natural protein partner. Some of these small mols. are now making their way through clin. trials, so this high-hanging fruit might not be far out of reach.
- 19Sanchez, R., Meslamani, J., and Zhou, M. M. (2014) The bromodomain: from epigenome reader to druggable target Biochim. Biophys. Acta 1839, 676– 685Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmtFagtrw%253D&md5=95ec21cf13231d9e63bce9b3ccdd6d6fThe bromodomain: From epigenome reader to druggable targetSanchez, Roberto; Meslamani, Jamel; Zhou, Ming-MingBiochimica et Biophysica Acta, Gene Regulatory Mechanisms (2014), 1839 (8), 676-685CODEN: BBAGC6; ISSN:1874-9399. (Elsevier B.V.)A review. Lysine acetylation is a fundamental post-translational modification that plays an important role in the control of gene transcription in chromatin in an ordered fashion. The bromodomain, the conserved structural module present in transcription-assocd. proteins, functions exclusively to recognize acetyl-lysine on histones and non-histone proteins. The structural analyses of bromodomains' recognition of lysine-acetylated peptides derived from histones and cellular proteins provide detailed insights into the differences and unifying features of biol. ligand binding selectivity by the bromodomains. Newly developed small-mol. inhibitors targeting bromodomain proteins further highlight the functional importance of bromodomain/acetyl-lysine binding as a key mechanism in orchestrating mol. interactions and regulation in chromatin biol. and gene transcription. These new studies argue that modulating bromodomain/acetyl-lysine interactions with small-mol. chems. offer new opportunities to control gene expression in a wide array of human diseases including cancer and inflammation. This article is part of a Special Issue entitled: Mol. mechanisms of histone modification function.
- 20Dawson, M. A., Kouzarides, T., and Huntly, B. J. (2012) Targeting epigenetic readers in cancer N. Engl. J. Med. 367, 647– 657Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xht1Chu77F&md5=f5e1e82c8a1105f798b1f7719138721cTargeting epigenetic readers in cancerDawson, Mark A.; Kouzarides, Tony; Huntly, Brian J. P.New England Journal of Medicine (2012), 367 (7), 647-657CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)A review on assocn. between epigenetics and chromatin biol., BET bromodomain inhibitors, and role of BET inhibitors in cancer.
- 21Filippakopoulos, P., Qi, J., Picaud, S., Shen, Y., Smith, W. B., Fedorov, O., Morse, E. M., Keates, T., Hickman, T. T., Felletar, I., Philpott, M., Munro, S., McKeown, M. R., Wang, Y., Christie, A. L., West, N., Cameron, M. J., Schwartz, B., Heightman, T. D., La Thangue, N., French, C. A., Wiest, O., Kung, A. L., Knapp, S., and Bradner, J. E. (2010) Selective inhibition of BET bromodomains Nature 468, 1067– 1073Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXotlGmtA%253D%253D&md5=bd9cadac86124d2c6c3cd5b2a92d68c2Selective inhibition of BET bromodomainsFilippakopoulos, Panagis; Qi, Jun; Picaud, Sarah; Shen, Yao; Smith, William B.; Fedorov, Oleg; Morse, Elizabeth M.; Keates, Tracey; Hickman, Tyler T.; Felletar, Ildiko; Philpott, Martin; Munro, Shongah; McKeown, Michael R.; Wang, Yuchuan; Christie, Amanda L.; West, Nathan; Cameron, Michael J.; Schwartz, Brian; Heightman, Tom D.; La Thangue, Nicholas; French, Christopher; Wiest, Olaf; Kung, Andrew L.; Knapp, Stefan; Bradner, James E.Nature (London, United Kingdom) (2010), 468 (7327), 1067-1073CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Epigenetic proteins are intently pursued targets in ligand discovery. So far, successful efforts have been limited to chromatin modifying enzymes, or so-called epigenetic 'writers' and 'erasers'. Potent inhibitors of histone binding modules have not yet been described. Here the authors report a cell-permeable small mol. (I,JQ1) that binds competitively to acetyl-lysine recognition motifs, or bromodomains. High potency and specificity towards a subset of human bromodomains is explained by co-crystal structures with bromodomain and extra-terminal (BET) family member BRD4, revealing excellent shape complementarity with the acetyl-lysine binding cavity. Recurrent translocation of BRD4 is obsd. in a genetically-defined, incurable subtype of human squamous carcinoma. Competitive binding by JQ1 displaces the BRD4 fusion oncoprotein from chromatin, prompting squamous differentiation and specific antiproliferative effects in BRD4-dependent cell lines and patient-derived xenograft models. These data establish proof-of-concept for targeting protein-protein interactions of epigenetic 'readers', and provide a versatile chem. scaffold for the development of chem. probes more broadly throughout the bromodomain family.
- 22Dawson, M. A., Prinjha, R. K., Dittmann, A., Giotopoulos, G., Bantscheff, M., Chan, W. I., Robson, S. C., Chung, C. W., Hopf, C., Savitski, M. M., Huthmacher, C., Gudgin, E., Lugo, D., Beinke, S., Chapman, T. D., Roberts, E. J., Soden, P. E., Auger, K. R., Mirguet, O., Doehner, K., Delwel, R., Burnett, A. K., Jeffrey, P., Drewes, G., Lee, K., Huntly, B. J., and Kouzarides, T. (2011) Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia Nature 478, 529– 533Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXht1GqsL%252FO&md5=dffb4b81da4ca8291c27139d0df86324Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemiaDawson, Mark A.; Prinjha, Rab K.; Dittman, Antje; Giotopoulos, George; Bantscheff, Marcus; Chan, Wai-In; Robson, Samuel C.; Chung, Chun-wa; Hopf, Carsten; Savitski, Mikhail M.; Huthmacher, Carola; Gudgin, Emma; Lugo, Dave; Beinke, Soren; Chapman, Trevor D.; Roberts, Emma J.; Soden, Peter E.; Auger, Kurt R.; Mirguet, Olivier; Doehner, Konstanze; Delwel, Ruud; Burnett, Alan K.; Jeffrey, Phillip; Drewes, Gerard; Lee, Kevin; Huntly, Brian J. P.; Kouzarides, TonyNature (London, United Kingdom) (2011), 478 (7370), 529-533CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Recurrent chromosomal translocations involving the mixed lineage leukemia (MLL) gene initiate aggressive forms of leukemia, which are often refractory to conventional therapies. Many MLL-fusion partners are members of the super elongation complex (SEC), a crit. regulator of transcriptional elongation, suggesting that aberrant control of this process has an important role in leukemia induction. Here we use a global proteomic strategy to demonstrate that MLL fusions, as part of SEC and the polymerase-assocd. factor complex (PAFc), are assocd. with the BET family of acetyl-lysine recognizing, chromatin adaptor' proteins. These data provided the basis for therapeutic intervention in MLL-fusion leukemia, via the displacement of the BET family of proteins from chromatin. We show that a novel small mol. inhibitor of the BET family, GSK1210151A (I-BET151), has profound efficacy against human and murine MLL-fusion leukemic cell lines, through the induction of early cell cycle arrest and apoptosis. I-BET151 treatment in two human leukemia cell lines with different MLL fusions alters the expression of a common set of genes whose function may account for these phenotypic changes. The mode of action of I-BET151 is, at least in part, due to the inhibition of transcription at key genes (BCL2, C-MYC and CDK6) through the displacement of BRD3/4, PAFc and SEC components from chromatin. In vivo studies indicate that I-BET151 has significant therapeutic value, providing survival benefit in two distinct mouse models of murine MLL-AF9 and human MLL-AF4 leukemia. Finally, the efficacy of I-BET151 against human leukemia stem cells is demonstrated, providing further evidence of its potent therapeutic potential. These findings establish the displacement of BET proteins from chromatin as a promising epigenetic therapy for these aggressive leukemias.
- 23Oakley, R. H., Laporte, S. A., Holt, J. A., Barak, L. S., and Caron, M. G. (1999) Association of beta-arrestin with G protein-coupled receptors during clathrin-mediated endocytosis dictates the profile of receptor resensitization J. Biol. Chem. 274, 32248– 32257Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXnt12jtbg%253D&md5=6a884b71028ff86e53f664fec39a66cdAssociation of β-arrestin with G protein-coupled receptors during clathrin-mediated endocytosis dictates the profile of receptor resensitizationOakley, Robert H.; Laporte, Stephane A.; Holt, Jason A.; Barak, Larry S.; Caron, Marc G.Journal of Biological Chemistry (1999), 274 (45), 32248-32257CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Resensitization of G protein-coupled receptors (GPCRs) following agonist-mediated desensitization is a necessary step for maintaining physiol. responsiveness. However, the mol. mechanisms governing the nature of GPCR resensitization are poorly understood. Here, we examine the role of β-arrestin in the resensitization of the β2 adrenergic receptor (β2AR), known to recycle and resensitize rapidly, and the vasopressin V2 receptor (V2R), known to recycle and resensitize slowly. Upon agonist activation, both receptors recruit β-arrestin to the plasma membrane and internalize in a β-arrestin- and clathrin-dependent manner. However, whereas β-arrestin dissocs. from the β2AR at the plasma membrane, it internalizes with the V2R into endosomes. The differential trafficking of β-arrestin and the ability of these two receptors to dephosphorylate, recycle, and resensitize is completely reversed when the carboxyl-terminal tails of these two receptors are switched. Moreover, the ability of β-arrestin to remain assocd. with desensitized GPCRs during clathrin-mediated endocytosis is mediated by a specific cluster of phosphorylated serine residues in the receptor carboxyl-terminal tail. These results demonstrate that the interaction of β-arrestin with a specific motif in the GPCR carboxyl-terminal tail dictates the rate of receptor dephosphorylation, recycling, and resensitization, and thus provide direct evidence for a novel mechanism by which β-arrestins regulate the reestablishment of GPCR responsiveness.
- 24Laporte, S. A., Oakley, R. H., Zhang, J., Holt, J. A., Ferguson, S. S., Caron, M. G., and Barak, L. S. (1999) The beta2-adrenergic receptor/betaarrestin complex recruits the clathrin adaptor AP-2 during endocytosis Proc. Natl. Acad. Sci. U.S.A. 96, 3712– 3717Google ScholarThere is no corresponding record for this reference.
- 25Mahen, R., Koch, B., Wachsmuth, M., Politi, A. Z., Perez-Gonzalez, A., Mergenthaler, J., Cai, Y., and Ellenberg, J. (2014) Comparative assessment of fluorescent transgene methods for quantitative imaging in human cells Mol. Biol. Cell 25, 3610Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitFOlsLbF&md5=b41ac0f052dfa7bf26ce2bdcbf9bf093Comparative assessment of fluorescent transgene methods for quantitative imaging in human cellsMahen, Robert; Koch, Birgit; Wachsmuth, Malte; Politi, Antonio Z.; Perez-Gonzalez, Alexis; Mergenthaler, Julia; Cai, Yin; Ellenberg, JanMolecular Biology of the Cell (2014), 25 (22), 3610-3618, 9 pp.CODEN: MBCEEV; ISSN:1939-4586. (American Society for Cell Biology)Fluorescence tagging of proteins is a widely used tool to study protein function and dynamics in live cells. However, the extent to which different mammalian transgene methods faithfully report on the properties of endogenous proteins has not been studied comparatively. Here we use quant. live-cell imaging and single-mol. spectroscopy to analyze how different transgene systems affect imaging of the functional properties of the mitotic kinase Aurora B. We show that the transgene method fundamentally influences level and variability of expression and can severely compromise the ability to report on endogenous binding and localization parameters, providing a guide for quant. imaging studies in mammalian cells.
Supporting Information
Supporting Information
ARTICLE SECTIONSDetailed experimental protocols, supplementary figures, and tables. This material is available free of charge at The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acschembio.5b00143.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.