ACS Publications. Most Trusted. Most Cited. Most Read
My Activity
CONTENT TYPES

Figure 1Loading Img

NanoBRET—A Novel BRET Platform for the Analysis of Protein–Protein Interactions

View Author Information
Promega Corporation, 2800 Woods Hollow, Madison, Wisconsin 53711, United States
Promega Biosciences Incorporated, 277 Granada Drive, San Luis Obispo, California 93401, United States
Cite this: ACS Chem. Biol. 2015, 10, 8, 1797–1804
Publication Date (Web):May 25, 2015
https://doi.org/10.1021/acschembio.5b00143

Copyright © 2015 American Chemical Society. This publication is licensed under these Terms of Use.

  • Open Access

Article Views

39384

Altmetric

-

Citations

LEARN ABOUT THESE METRICS
PDF (2 MB)
Supporting Info (1)»

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.

Dynamic behaviors in cells are mechanistically entwined with a vast interplay of transient interactions occurring between intracellular proteins. Protein–protein interactions (PPI) are thus fundamental to understanding cellular physiology and are attractive as pharmaceutical targets. Although numerous methods are available for the discovery of protein interactions, most notably through analysis by mass spectrometry, it remains challenging to study the dynamics of these interactions in a cellular context. Resonance energy transfer has been an attractive approach for this purpose, where excited-state energy is transferred from one fluorophore to another when brought into close proximity. (1) The rigorous distance constraint of energy transfer restricts proximity detection to roughly the physical span of a typical protein (∼5 nm), making this approach well suited for the crowded molecular space within cells.
Energy transfer is most often achieved using autofluorescent proteins (e.g., YFP), which can be linked to the interacting proteins through genetic fusions. The method allows visualization of subcellular location and interaction dynamics by microscopy imaging but often requires relatively high levels of expression for reliable measurements. This can make analysis at physiological concentrations challenging within the cells and may lead to artifacts of protein behavior. The intensity of the illuminating light source also causes problems with phototoxicity, photobleaching of the fluorophores, direct excitation of the acceptor fluorophore, and background fluorescence from the sample. These issues make sensitive and reliable measurements difficult, particularly for kinetic processes.
Although FRET is generally recognized as requiring an external light source, such as a laser, to generate the excited state of the donor, the energy can be supplied instead by a chemical reaction. For BRET (bioluminescence resonance energy transfer), using a luciferase as the donor sidesteps many of the deficiencies associated with direct illumination of the sample. (2-4) Furthermore, quantitative measurements even at very low expression levels in cells should be possible due to the extraordinary sensitivity typical of luciferase assays. Yet despite these predictable advantages, BRET has not been widely adopted largely due to limited sensitivity and dynamic range of the presently available methods.
Current methods routinely utilize Renilla luciferase (Rluc) as the energy donor, more recently relying on the Rluc8 mutant to provide higher light intensity. (5) In the most commonly used BRET format, BRET1, Rluc is typically matched with a yellow fluorescent protein (YFP) to achieve efficient energy transfer by significant overlap of its excitation spectrum with the bioluminescence. However, the spectral proximity of Rluc and YFP (∼55 nm peak separation) produces a substantial measurement background by the extension of donor signal into the acceptor channel. The high background increases assay noise, which diminished dynamic range and sensitivity.
An alternative BRET, referred to as BRET2, was developed to increase spectral separation between the luciferase and the fluorescent acceptor. (6) The system utilizes a substrate analog, bisdeoxycoelenterazine, to shift the luminescence to much shorter wavelengths (400 nm peak from 480 nm), and GFP2 or GFP10, which exhibit excitation and emission maxima of 395 and 510 nm, respectively. Although the large Stokes shift of GFP2/GFP10 improves spectral resolution (115 nm), this advantage is largely negated by the very low quantum yield and poor luminescence stability. (7) The recent adoption of Rluc8 (8) for BRET2 has improved performance, but light intensities are still far below other BRET configurations. (9)
The recent development of the Nanoluc luciferase (Nluc) offers an opportunity to further improve the performance of BRET assays. (10) Nluc is an engineered protein (19 kDa), which uses a novel coelenterazine derivative (furimazine) as its substrate. Its small size and high physical stability are desirable for use as a fusion tag, imparting minimal influence when tethered onto other proteins. Although it is only about half the size of Rluc8 (36 kDa), it produces sustained luminescence with much greater intensity, allowing very small quantities to be accurately quantified. Moreover, its bioluminescence spectrum is narrower than Rluc8, (10) permitting better spectral discrimination with acceptor fluorophores. To provide distinction from the other configurations of BRET, we refer to energy transfer using Nluc as the donor as NanoBRET.
With Nluc available as a more effective BRET donor, we focused on matching this with an optimal fluorescent acceptor. The HaloTag fusion tag allowed us to rapidly evaluate a range of fluorophores for this purpose. HaloTag (HT) is a two component system consisting of a stable protein (36 kDa) which can be genetically tethered onto other proteins and a chloroalkane ligand that can be chemically coupled onto fluorophores or other molecules of interest. (11) The chloroalkane irreversibly links to the HaloTag through rapid formation of a covalent bond. By using this modular system, we found that long-wavelength fluorophores were preferred for BRET measurements by providing better spectral separation from the luciferase donor. Best performance was achieved with a chloroalkane derivative of nonchloro TOM (NCT) dye, which provides peak light emission at 635 nm. Thus, peak emission from the donor and acceptor were spectrally separated by 175 nm, significantly greater than provided even by the BRET2 system.
The high luminescence intensity of NanoBRET allows detection of protein interactions at low levels comparable to endogenous physiological conditions. The assay exhibits a linear dynamic range extending over several orders of magnitude and provides greater sensitivity than Rluc8/YFP by typically more than an order of magnitude for analogous molecular configurations. The strong performance of NanoBRET was utilized to develop the first assays for quantifying the binding dynamics of bromodomain protein with chromatin in living cells. Furthermore, the high intensity of NanoBRET also raises the potential for quantifying energy transfer in individual cells by microscopy imaging.

Results and Discussion

ARTICLE SECTIONS
Jump To

Optimization of the Fluorescent Acceptor

Genetically fusing HaloTag directly onto Nluc (Nluc-HT) offered a convenient means for providing a fixed geometry between the donor and acceptor. Fifty-seven different chloroalkane–fluorophore conjugates were synthesized and screened in HEK293 cells transiently expressing the Nluc-HT fusion. By using a cell-based assay, we were able to evaluate the fluorophores for their ability to permeate cellular membranes and produce a BRET signal within the intracellular environment. Each fluorophore was evaluated using a set of four long-pass filters (cut-on 570, 590, 610, and 630 nm) for the acceptor emission, where the BRET response was determined by comparing measurements made both in the presence and absence of the fluorophore. The HaloTag TMR Ligand (590 nm emission peak) was used as a positive control, having been previously shown to bind efficiently and stoichiometrically to HaloTag in living cells. From these experiments, a nonchloro TOM (NCT) ligand was identified as providing the greatest signal over the background. The NCT ligand has an excitation maximum of 595 nm with an emission maximum of 635 nm when bound to HaloTag (Supporting Information Figure S1a). Labeling of HaloTag fusion proteins in living cells is rapid with saturation occurring in less than 10 min after the addition of 250 nM HaloTag NCT ligand (Supporting Information Figure S1b). No obvious cytotoxicity was exhibited by the NanoBRET expression constructs in combination of NCT ligand and furimazine (the principal reagents required for NanoBRET) for periods of up to 24 h (Supporting Information Figure S2).
The Förster equation predicts that the greatest energy transfer will occur with maximal overlap between the emission spectrum of the donor and excitation spectrum of the acceptor. The HaloTag Oregon Green Ligand (512 nm peak emission) has greater overlap, and the NCT ligand has less overlap, with Nluc than does the HaloTag TMR Ligand (Supporting Information Figure S1). Accordingly, in the Nluc-HT fusion, more energy is transferred from Nluc to the Oregon Green Ligand, and less to the NCT ligand, compared to the TMR Ligand (Figure 1A). However, since the decline in energy transfer efficiency is substantially smaller compared to the decrease in Nluc light intensity across the same spectral range, the signal to background ratio increases substantially. Thus, the highest signal to background was achieved with the NCT ligand, even though it does not support the most efficient energy transfer (Supporting Information Figure S3). Moreover, the modest reduction in transfer efficiency is more than compensated by the increased light intensity of Nluc. The specific activity of Nluc is about 150-fold greater than Rluc, and 30-fold greater than Rluc8. Signal decay kinetics are comparable between Nluc and Rluc8, with NanoLuc showing a slightly improved decay profile in biological buffers (Supporting Information Figure S4a–c).

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.

In BRET measurements of molecular interactions, since energy transfer occurs when the donor is occupied by an acceptor, assay sensitivity can be specified as the lowest detectable degree of occupancy. This can be modeled using the Nluc-HT fusion, where fully bound to a fluorophore represents 100% occupancy and fully bound with a nonfluorophore represents 0% occupancy. Because the covalently bound HaloTag ligands cannot be exchanged once attached, different ratios of the “occupied” and “unoccupied” fusion protein can be combined to create fractional occupancy. Thus, a series of stable compositions can be readily generated that hold the total amount of Nluc constant while accurately varying the portion occupied by an acceptor. By this method, NanoBRET exhibits linearity with donor occupancy extending beyond 3 orders of magnitude (Figure 1B; see Supporting Information Figure S5 for uncorrected acceptor/donor channel emission ratios). The smallest degree of measurable occupancy can be determined using a statistical metric of discrimination from the unoccupied donor. A commonly used metric is Z′-factor, where quantitatively reliable discrimination is attained for values above 0.5. An evaluation of Z′-factor values relative to fractional occupancy indicates that the NCT ligand provides about 20-fold greater sensitivity than the Oregon Green Ligand and about 4-fold over the TMR Ligand (Table 1).
Table 1. Sensitivity Limits for NanoBRET Acceptors
Z′-factora
 acceptor fluorophore
fractional occupancy (%)NCTTMROGYFPRFP
0.050.46<0<0<0<0
0.100.65<0<0<0<0
0.250.780.57<0<0<0
0.500.850.71<0<0<0
1.00.920.810.046<0<0
2.50.950.900.78<0<0
5.00.960.930.880.230.21
100.980.950.900.750.65
250.980.970.960.890.82
500.980.980.980.920.91
1000.990.990.970.970.93
a

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).

Since autofluorescent proteins are most commonly used as the acceptor in BRET assays, their performance was also evaluated in NanoBRET. TurboYFP is a fast maturing YFP variant derived from Phialidium sp., with brightness comparable to Venus and spectral properties similar to the Oregon Green Ligand. TagRFP is derived from Entacmaea quadricolor and has spectral properties similar to the TMR Ligand. (12-14) These were directly fused genetically to Nluc, and fractional occupancy was established as before, using Nluc-HT as an unoccupied donor. Despite the spectral similarity with the fluorescent HaloTag ligands, assay sensitivity was substantially diminished (Table 1). This may be explained in part by having lower quantum yields relative to the HaloTag ligands, inefficient chromophore maturation as well as differences in geometries of these fusions relative to Nluc-HT. (15) In addition, the chromophore in autofluorescent proteins may also be influenced by intracellular conditions, as suggested by an apparent linkage we observed between energy transfer and expression levels for TurboYFP and TagRFP (Supporting Information Figure S6). Both exhibited decreased energy transfer with lower expression while HaloTag provided nearly constant energy transfer. BRET is expected to be independent of concentration, so long as the orientation between the donor and acceptor remains fixed, since the ratiometric values are normalized to donor intensity. This was the case for these fusions when measured as purified proteins (Supporting Information Figure S7).

Performance of NanoBRET for Quantifying Protein Interactions

Having established HaloTag-NCT as an optimal energy acceptor for Nluc, this combination should enhance the performance of BRET for analyzing intracellular protein–protein interactions (PPI). Furthermore, labeling HaloTag with NCT as the energy acceptor is straightforward, requiring only the addition of the tagged fluorophore to the culture medium prior to making measurements (Figure 2). We evaluated this by benchmarking performance to the commonly used pairing of Rluc8 with YFP. For this purpose, we selected the rapamycin-induced interaction between FKBP12 and Frb (Rapamycin-binding domain of mTOR) as the biological model. The Frb/FKBP interaction model has been frequently employed for the evaluation of new PPI analytical methods due to its simplicity and robustness. (16)

Figure 2

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

Because the efficiency of energy transfer can be affected by geometric orientation, it is important to examine all eight possible configurations of donor and acceptor fused onto the interacting proteins. Specifically, these are the donor and acceptor fused to each of the proteins at either terminus of the polypeptide. For all configurations of Nluc/HT-NCT and Rluc8/YFP fused to FKBP and Frb, induction of PPI by rapamycin led to an increase in energy transfer. As expected, the different configurations of the fusions show considerable variation in their relative response to rapamycin (Figure 3a, see Supporting Information Figure S8a,b for uncorrected acceptor/donor channel emission ratios). For all configurations of Rluc8/YFP, the response was lower than for the corresponding configuration of Nluc/HT-NCT and varied by 4 to 70 fold between analogously oriented pairs. The most efficient configurations of Nluc/HT-NCT and Rluc8/YFP (Figure 3a, bars with hatch pattern), respectively, exhibited a 7-fold difference in response to rapamycin.

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.

The donors and acceptors used in these comparisons are structurally distinct, possibly resulting in some differences in their relative geometry when fused to FKBP and Frb. Nonetheless, it is likely that NanoBRET consistently exhibits greater response dynamics due to its greater spectral separation between the donor and acceptor, narrower emission spectrum of Nluc, and higher quantum yield of NCT ligand. By either choice of donor and acceptor, the EC50 values were essentially unchanged and in agreement with previously published data (Figure 3b, see Supporting Information Figure 9A–D for raw RLU values and uncorrected acceptor/donor channel emission ratios), indicating that the pharmacology of rapamycin was not influenced by the reporter fusions. (16) In addition, NanoBRET measurements of this stable interaction remained relatively constant for 60 min both in the presence and in the absence of rapamycin, indicating the ability to continuously monitor intracellular PPIs over an extended period of time (Supporting Information Figure S10).
As molecular interactions are subject to concentration, having appropriate expression levels of the donor and acceptor is important for achieving biologically relevant data. Because regulatory proteins in particular tend to be present at low levels in cells, energy transfer must be measurable at low expression levels. Furthermore, unphysiologically high expression can lead to biological artifacts such as ligand independent signaling, improper recruitment of other cellular components and mis-folding or mis-localization of the proteins. To explore the capacity for detecting PPI at physiologically relevant levels, we measured rapamycin-induced BRET over a titrated range of donor and acceptor expression plasmids, combined in equal amounts and serially diluted with carrier DNA. For both Nluc/HT-NCT and Rluc8/YFP, a negative correlation between expression level and rapamycin response was evident, possibly due to competition between the introduced fusions and their endogenous counterparts at lower transfection amounts. Western blots for detection of FKBP showed that fusions to either acceptor were present in the sample at amounts comparable to the endogenous protein near the middle of the titration range (Supporting Information Figure S11A,B). Consistency in expression of FKBP fused to either acceptor, over the titrated range of DNA, was also confirmed by flow cytometry analysis of FKBP-HaloTag and FKBP-turboYFP expression (Supporting Information Figure 12A,B).
However, there was a marked difference in signal separation between the BRET pairs, especially at low levels of fusion protein. The NanoBRET pair exhibited complete data separation between rapamycin treated and untreated samples for the entire range of DNA dilutions (Figure 4a, Supplemental Figure 12c), whereas the Rluc8/YFP pair showed convergence to a single value at much higher analyte levels (Figure 4b, Supplemental Figure 12d). A Z′-factor analysis was used to estimate the capacity for reliably distinguishing a rapamycin-induced interaction from an untreated control. (17) The NanoBRET based assay maintained good sensitivity (Z′ > 0.65) even at concentrations of FKBP-HT well below the endogenous FKBP. In comparison, the Rluc8/YFP pair exhibited decreased assay sensitivity even for analyte amounts well above endogenous levels. These results show that NanoBRET can provide better assay sensitivity and response dynamics even at analyte levels comparable to or below physiological concentrations within cells. The ability to perform such measurements at low concentrations of reporter may be particularly significant for technically challenging cell types, such as stem cells and primary cells, which are often difficult to transfect or have limited capacity for expressing transgenes.

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

The role of PPI in many disease relevant signaling pathways has motivated interest in compounds that target these interactions for therapeutic purposes. (18) NanoBRET provides a means for exploring the action of these compounds within the intracellular environment. This is exemplified by characterizing compounds able to inhibit binding of bromodomain proteins to chromatin, which reveals the importance of the cellular context and how biochemical studies can sometimes be misleading. The binding of bromodomain proteins to acetylated histones is one of the key mechanisms for epigenetic regulation of transcriptional activity. The ability to block binding by specific bromodomain proteins is an emerging area of drug therapy for a variety of cancers, which has been most elegantly shown for the BET family members, BRD2, BRD3, and BRD4. (19-21)
We show that the interaction between BRD4 and chromatin can be monitored with NanoBRET by expressing Nluc-BRD4 in mammalian cells with human histone H3.3-HT fusions (Figure 5A). Treatment of the cells with increasing doses of known inhibitor I-BET151, which competes for binding to acetyl lysine, showed a dose dependent decrease of BRET signal with an IC50 close to published values (Figures 5A, see Supporting Information Figure S13A for uncorrected acceptor/donor channel emission ratios). (22) In similar studies with a non-BET family bromodomain protein, CBP, we do not see any displacement from chromatin with I-BET151, rather a slight increase we believe is due to increased occupancy of CBP after BET family displacement (Figures 5B, see Supporting Information Figure S13B for uncorrected acceptor/donor channel emission ratios). Similar results in both systems were obtained using histone H4-HT as the acceptor partner in place of histone H3.3-HT (Supporting Information Figure S13C and D). These results demonstrate that the action of I-BET151 can be observed by NanoBRET for specific pairs of bromodomain proteins and histones in living cells.

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

The ability to image BRET within individual cells has previously been restricted by the relatively low photon flux provided by bioluminescence. However, the bright luminescence of Nluc combined with recent advances in cellular imaging technologies is beginning to change this situation. We recently demonstrated that Nluc enables visualization of subcellular translocation events within living cells with subsecond temporal resolution. (10) In this study, the well-established ligand-induced recruitment of β-arrestin 2 (ARRB2) to vasopressin receptor 2 (AVPR2) was used as a model for visualizing protein interactions. (23)
In the absence of a ligand, Nluc-ARRB2 showed the anticipated uniform distribution in cells (Figure 6A). A weak signal was also observed in the acceptor channel, which can be attributed to “bleed-through” of the Nluc luminescence. The addition of arginine vasopressin (AVP) induced a substantial signal increase in the acceptor channel, accompanied by the appearance of a vesicular pattern in both detection channels, which indicates association of Nluc-ARRB2 with AVPR2-HT, followed by trafficking into clathrin-coated pits (Figure 6A, Supporting Information Figure S14A). (24) The sequential imaging with short exposure times allowed the recording of arrestin recruitment kinetics in individual cells. Although transient transfection leads to variable expression levels between cells, the relative response kinetics were found to be similar and corresponded well with data obtained for a cell population in a 96-well plate format (Figure 6B, see Supporting Information Figure S14B and C for uncorrected acceptor/donor channel emission ratios).

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.

In addition to the vasopressin receptor model, NanoBRET imaging was successfully applied to the rapamycin-induced interaction of FKBP with Frb, and the EGF-induced recruitment of grb2 to the EGF receptor (Supporting Information Figure S15A and B). The sustained high luminescence output of Nluc allows continuous monitoring of protein–protein interactions for up to 2 h and also improves spatial and temporal resolution. The enhanced feasibility for imaging of PPI in living cells afforded by NanoBRET may be of particular use for signaling events that are confined to specific subcellular compartments, especially if only a small subset of the donor-tagged protein is engaged in the interaction of interest.

Conclusions

BRET has been used for nearly 15 years to analyze protein–protein interaction in living cells. Benefiting from the capacity of luminescent chemistries to elucidate intracellular events, BRET assays are further advantaged by their ratiometric format for enhancing data precision and robustness. By representing acceptor proximity in relation to an available donor, the ratiometric normalization provides automatic correction for variable cell numbers or gene expression, inconsistent reagent potency, inadvertent luciferase inhibitors, and so forth. Yet, despite these advantages, usage of this technology has remained largely confined to relatively specific areas of investigation, most notably the analysis of cell surface receptor interactions. The main reason may be the comparatively limited sensitivity and dynamic range provided by current BRET systems.
With the recent development of Nluc, which emits bright, stable, and spectrally narrow luminescence, we saw an opportunity to design a BRET platform with substantially improved capabilities. This was achieved by pairing Nluc with a spectrally well separated acceptor, thus effectively reducing the background caused by “bleed through” of the donor signal into the acceptor channel. Our results indicate that NanoBRET (Figure 2) provides a reliable platform for analyzing protein–protein interactions at low expression levels in living cells, which is important for maintaining appropriate interaction dynamics. This applies not only to the tagged proteins subject to measurement but also to the many untagged endogenous proteins whose contributing influence comprises the physiological environment. The increased sensitivity of NanoBRET should be useful in combination with genomic editing technologies such as TALEN or CRISPR, allowing protein expression to be directed by the corresponding endogenous gene locus. (25)
Due to the delicate energy relationships associated with transient protein interactions, the influence of synthetic molecules on these interactions may be particularly reliant on the intracellular environment. Drug development founded on biochemical approaches may not adequately encompass the essential elements necessary to recapitulate a living molecular process. We have demonstrated that NanoBRET can deliver precise quantitative measurements of dynamic interactions as they occur within cells. Moreover, the technology delivers sufficient sensitivity to reveal these interactions occurring even to relatively small extents (i.e., at low fractional occupancies). The high emission intensity of NanoBRET also opens possibilities for imaging protein interactions within individual cells, and we speculate that the red-shifted emission of the acceptor may be suitable for imaging in living animals. In addition, linking fluorophores onto other types of molecules may prove useful for revealing a broader range of intracellular interactions.
The technical challenge of working with isolated full-length proteins also promotes the common usage of protein fragments as surrogates. Such simplified assays may not capture the multidomain and multifunctional nature of intracellular proteins and the many cellular factors which regulate their interactions. This is clearly demonstrated in our examples using NanoBRET to monitor interactions with chromatin. These interactions are highly modulated and dictated by the chromatin environment, including nucleosome positioning, presence of other transcription factors and transcription machinery, and the array of modifications present at any given time on the various histones. This environment cannot be replicated in a biochemical setting and thus highlights the necessity for monitoring such interactions within cells. NanoBRET configured with histones as energy acceptors should provide the ability to monitor interactions not only of bromodomains, as demonstrated here, but other chromatin interacting proteins as well.

Material and Methods

ARTICLE SECTIONS
Jump To

Fractional Occupancy Assay

A 400 pM dilution of recombinant Nluc-HT, Nluc-turboYFP, and Nluc-tagRFP protein was prepared in PBS + 0.1% BSA (Promega). The Nluc-HT solution was split in separate samples and labeled for 180 min at RT with 5 μM of either the indicated fluorescent HaloTag ligands (Oregon Green, TMR, and NCT) or the nonfluorescent HaloTag PEG-biotin ligand. To prepare solutions representing different degrees of occupancy, the stock solutions of Nluc-HT-ligand, Nluc-TurboYFP, and Nluc-tagRFP were combined with Nluc-HT-PEG-biotin (as unoccupied donor) at defined ratios and a total concentration of 2 nM in PBS + 0.1% BSA. The percentage of acceptor occupied donor relative to total donor is referred to as fractional occupancy. BRET measurements were performed in white 96-well plates by combining 50 μL of the donor/acceptor mixtures with 50 μL of 20 μM furimazine in PBS. All measurements were performed at RT on a Thermo Scientific Varioskan Flash plate reader equipped with a 460/80 nm band-pass filter (donor) and 530 nm (HaloTag Oregon Green Ligand/TurboYFP), 590 nm (HaloTag TMR Ligand/Tag RFP), or 610 nm (HaloTag NCT Ligand) long-pass filters (acceptor) using an integration time of 0.5 s.

Live Cell Labeling with HaloTag NCT Ligand

For labeling of cells with the HaloTag NCT ligand, we developed two alternative protocols based on the needs of experimental PPI models (protocol use is also indicated in the description of the PPI models). Rapid labeling protocol: Cells were labeled by replacing the growth medium with labeling medium (Opti-MEM supplemented with 250 nM HaloTag NCT ligand) followed by incubation for 60–90 min at 37 °C. Overnight labeling protocol: Cells were labeled by replacing the growth medium with labeling medium (Opti-MEM supplemented with 4% FBS and 100 nM HaloTag NCT ligand) followed by incubation for 12–18 h at 37 °C. All HaloTag NCT ligand dilutions were performed using a stock solution of 5 mM HaloTag NCT ligand in DMSO. Subsequent treatment and processing of samples was performed as outlined in assay specific experimental procedures. Removal of excess ligand is not required since no unspecific bystander BRET was observed between the HaloTag NCT ligand and Nluc in biochemical or cell based assays for HaloTag NCT ligand concentrations of up to 500 nM.

Frb/FKBP Interaction Assays

HEK293 cells were transiently transfected with expression constructs for the indicated Frb and FKBP fusions and plated into a white 96-well tissue culture plate (Corning Costar #3917) at 2 × 104 cells/well in 100 μL volume. After 24 h of incubation, the growth medium was replaced by 50 μL Opti-MEM (Life Technologies). HaloTag containing samples were labeled by the addition of 50 μL of Opti-MEM containing HaloTag NCT Ligand at a concentration of 250 nM followed by incubation for 90 min at 37 °C. Interaction of Frb and FKBP was induced by the addition of rapamycin at the indicated concentration. After 15 min of incubation at 37 °C, furimazine (NanoBRET) or Coelenterazine h (BRET1) was added to each sample to a final concentration of 10 or 20 μM, respectively. All measurements were taken at RT using a Thermo Scientific Varioskan Flash plate reader using the filter setting as outlined above.

Supporting Information

ARTICLE SECTIONS
Jump To

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.

Author Information

ARTICLE SECTIONS
Jump To

  • Corresponding Author
    • Thomas Machleidt - Promega Corporation, 2800 Woods Hollow, Madison, Wisconsin 53711, United States Email: [email protected]
  • Authors
    • Carolyn C. Woodroofe - Promega Biosciences Incorporated, 277 Granada Drive, San Luis Obispo, California 93401, United States
    • Marie K. Schwinn - Promega Corporation, 2800 Woods Hollow, Madison, Wisconsin 53711, United States
    • Jacqui Méndez - Promega Corporation, 2800 Woods Hollow, Madison, Wisconsin 53711, United States
    • Matthew B. Robers - Promega Corporation, 2800 Woods Hollow, Madison, Wisconsin 53711, United States
    • Kris Zimmerman - Promega Corporation, 2800 Woods Hollow, Madison, Wisconsin 53711, United States
    • Paul Otto - Promega Corporation, 2800 Woods Hollow, Madison, Wisconsin 53711, United States
    • Danette L. Daniels - Promega Corporation, 2800 Woods Hollow, Madison, Wisconsin 53711, United States
    • Thomas A. Kirkland - Promega Biosciences Incorporated, 277 Granada Drive, San Luis Obispo, California 93401, United States
    • Keith V. Wood - Promega Corporation, 2800 Woods Hollow, Madison, Wisconsin 53711, United States
  • Notes
    The authors declare no competing financial interest.

References

ARTICLE SECTIONS
Jump To

This article references 25 other publications.

  1. 1
    Ciruela, F. (2008) Fluorescence-based methods in the study of protein-protein interactions in living cells Curr. Opin. Biotechnol. 19, 338 343
  2. 2
    Angers, 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 3689
  3. 3
    Xu, 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, 151 156
  4. 4
    Wampler, 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 2909
  5. 5
    Pfleger, K. D. and Eidne, K. A. (2006) Illuminating insights into protein-protein interactions using bioluminescence resonance energy transfer (BRET) Nat. Methods 3, 165 174
  6. 6
    Bertrand, 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, 533 541
  7. 7
    Hamdan, 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, 463 475
  8. 8
    Loening, 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, 391 400
  9. 9
    Kocan, 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, 888 898
  10. 10
    Hall, 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 1857
  11. 11
    Los, 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, 373 382
  12. 12
    Shagin, 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, 841 850
  13. 13
    Merzlyak, 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, 555 557
  14. 14
    Nagai, 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 90
  15. 15
    Shaner, N. C., Steinbach, P. A., and Tsien, R. Y. (2005) A guide to choosing fluorescent proteins Nat. Methods 2, 905 909
  16. 16
    Galarneau, 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, 619 622
  17. 17
    Zhang, 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 73
  18. 18
    Wells, J. A. and McClendon, C. L. (2007) Reaching for high-hanging fruit in drug discovery at protein-protein interfaces Nature 450, 1001 1009
  19. 19
    Sanchez, R., Meslamani, J., and Zhou, M. M. (2014) The bromodomain: from epigenome reader to druggable target Biochim. Biophys. Acta 1839, 676 685
  20. 20
    Dawson, M. A., Kouzarides, T., and Huntly, B. J. (2012) Targeting epigenetic readers in cancer N. Engl. J. Med. 367, 647 657
  21. 21
    Filippakopoulos, 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 1073
  22. 22
    Dawson, 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, 529 533
  23. 23
    Oakley, 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 32257
  24. 24
    Laporte, 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 3717
  25. 25
    Mahen, 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, 3610

Cited By

ARTICLE SECTIONS
Jump To

This article is cited by 300 publications.

  1. 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
  2. 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
  3. 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
  4. 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
  5. 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
  6. 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
  7. 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
  8. 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
  9. 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
  10. 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
  11. 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
  12. 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
  13. 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
  14. 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
  15. 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
  16. 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
  17. 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
  18. 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
  19. 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
  20. 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
  21. 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
  22. 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
  23. 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
  24. 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
  25. 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
  26. 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
  27. 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
  28. 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
  29. 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
  30. 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
  31. 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
  32. 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
  33. 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
  34. 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
  35. 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
  36. 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
  37. 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
  38. 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
  39. 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
  40. 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
  41. 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
  42. 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
  43. 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
  44. 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
  45. Peter J. Tonge . Drug–Target Kinetics in Drug Discovery. ACS Chemical Neuroscience 2018, 9 (1) , 29-39. https://doi.org/10.1021/acschemneuro.7b00185
  46. 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
  47. 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
  48. 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
  49. 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
  50. 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
  51. 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
  52. 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
  53. 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
  54. 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
  55. 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
  56. 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
  57. 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
  58. 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
  59. 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
  60. 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
  61. 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
  62. 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
  63. 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
  64. 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
  65. 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
  66. 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
  67. 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
  68. 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
  69. 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
  70. 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
  71. 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
  72. 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
  73. 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
  74. 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
  75. 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
  76. 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
  77. 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
  78. 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
  79. 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
  80. 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
  81. 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
  82. 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
  83. 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
  84. 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
  85. 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
  86. 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
  87. 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
  88. Xin Yu, Jin Wang. Quantitative measurement of PROTAC intracellular accumulation. 2023, 189-214. https://doi.org/10.1016/bs.mie.2022.11.001
  89. 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
  90. 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
  91. 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
  92. 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
  93. 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
  94. 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
  95. 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
  96. 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
  97. 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
  98. 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
  99. 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
  100. 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
Load more citations
  • 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 SECTIONS
    Jump To

    This article references 25 other publications.

    1. 1
      Ciruela, F. (2008) Fluorescence-based methods in the study of protein-protein interactions in living cells Curr. Opin. Biotechnol. 19, 338 343
    2. 2
      Angers, 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 3689
    3. 3
      Xu, 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, 151 156
    4. 4
      Wampler, 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 2909
    5. 5
      Pfleger, K. D. and Eidne, K. A. (2006) Illuminating insights into protein-protein interactions using bioluminescence resonance energy transfer (BRET) Nat. Methods 3, 165 174
    6. 6
      Bertrand, 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, 533 541
    7. 7
      Hamdan, 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, 463 475
    8. 8
      Loening, 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, 391 400
    9. 9
      Kocan, 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, 888 898
    10. 10
      Hall, 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 1857
    11. 11
      Los, 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, 373 382
    12. 12
      Shagin, 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, 841 850
    13. 13
      Merzlyak, 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, 555 557
    14. 14
      Nagai, 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 90
    15. 15
      Shaner, N. C., Steinbach, P. A., and Tsien, R. Y. (2005) A guide to choosing fluorescent proteins Nat. Methods 2, 905 909
    16. 16
      Galarneau, 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, 619 622
    17. 17
      Zhang, 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 73
    18. 18
      Wells, J. A. and McClendon, C. L. (2007) Reaching for high-hanging fruit in drug discovery at protein-protein interfaces Nature 450, 1001 1009
    19. 19
      Sanchez, R., Meslamani, J., and Zhou, M. M. (2014) The bromodomain: from epigenome reader to druggable target Biochim. Biophys. Acta 1839, 676 685
    20. 20
      Dawson, M. A., Kouzarides, T., and Huntly, B. J. (2012) Targeting epigenetic readers in cancer N. Engl. J. Med. 367, 647 657
    21. 21
      Filippakopoulos, 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 1073
    22. 22
      Dawson, 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, 529 533
    23. 23
      Oakley, 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 32257
    24. 24
      Laporte, 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 3717
    25. 25
      Mahen, 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, 3610
  • Supporting Information

    Supporting Information

    ARTICLE SECTIONS
    Jump To

    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.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect