SlideShare a Scribd company logo
1 of 9
Download to read offline
NATURE PROTOCOLS | VOL.1 NO.1 | 2006 | 337
PROTOCOL
INTRODUCTION
What is BRET?
BRET is a naturally occurring phenomenon used in the labora-
tory for real-time monitoring of protein-protein interactions
in live cells, cell extracts or purified preparations1−3. The nonra-
diative (dipole-dipole) transfer of energy from donor enzyme to
complementary acceptor fluorophore occurs after substrate oxida-
tion, and its efficiency is inversely proportional to the sixth power
of the distance between donor and acceptor dipoles. This energy
only excites the acceptor fluorophore if it is in close proximity to
the donor, resulting in light emission at a longer wavelength that
can be detected and expressed relative to the donor light emission.
By generating cDNA fusion constructs, which are transfected into
cells within a suitable expression vector4, fusion proteins consist-
ing of proteins of interest linked to donor or acceptor molecules
are expressed. Donor to acceptor energy transfer and consequent
emission from the acceptor generally indicates separation of less
than 10 nm, thus indicating that the proteins of interest are likely
to be interacting with each other (directly or as part of a complex)5.
Indeed, this high distance dependence means that various combi-
nations of fusion proteins with donor or acceptor molecules in dif-
ferent positions should be tested to establish the best combination
for BRET. The BRET procedure is summarized in Figure 1.
Diversity of proteins investigated
BRET has been used to study a wide range of protein-protein inter-
actions in bacterial6, plant7 and mammalian cells8,9. The first use
of BRET studied KaiB circadian clock protein dimerization6. Since
then, well over a hundred protein-protein interaction studies have
used BRET,with approximately half monitoring interactions involv-
ing G protein−coupled receptors (GPCRs), particularly dimeriza-
tion or oligomerization, but also interactions with both membrane
and cytosolic proteins10. The other half of studies that have used
BRET covers a diverse range of interactions too large to mention
here, but includes those involving other membrane-bound recep-
tors11,12, cytosolic receptors13,14, integrins11,15, enzymes15,16, endo-
philins17 and nuclear cofactors18. This diversity illustrates that any
protein-protein interaction can potentially be observed using BRET
provided that the proteins can be suitably labeled and expressed in a
functionally relevant manner1.
Suitability of BRET
The BRET technique is most suited to investigating ligand-modu-
lated (or reagent-modulated) interactions as changes in BRET sig-
nal after reagent addition provide good evidence for interaction
specificity, provided that the reagent is not cytotoxic and does not
inhibit substrate oxidation. Changes in BRET signal may repre-
Bioluminescence resonance energy transfer (BRET)
for the real-time detection of protein-protein
interactions
Kevin D G Pfleger, Ruth M Seeber & Karin A Eidne
7TM Laboratory/Laboratory for Molecular Endocrinology, Western Australian Institute for Medical Research (WAIMR) and UWA Centre for Medical Research, University
of Western Australia, Nedlands, Perth, Western Australia 6009, Australia. Correspondence should be addressed to K.D.G.P. (kpfleger@waimr.uwa.edu.au).
Published online 27 June 2006; doi:10.1038/nprot.2006.52
A substantial range of protein-protein interactions can be readily monitored in real time using bioluminescence resonance
energy transfer (BRET). The procedure involves heterologous coexpression of fusion proteins, which link proteins of interest to
a bioluminescent donor enzyme or acceptor fluorophore. Energy transfer between these proteins is then detected. This protocol
encompasses BRET1, BRET2 and the recently described eBRET, including selection of the donor, acceptor and substrate combination,
fusion construct generation and validation, cell culture, fluorescence and luminescence detection, BRET detection and data analysis.
The protocol is particularly suited to studying protein-protein interactions in live cells (adherent or in suspension), but cell extracts
and purified proteins can also be used. Furthermore, although the procedure is illustrated with references to mammalian cell culture
conditions, this protocol can be readily used for bacterial or plant studies. Once fusion proteins are generated and validated, the
procedure typically takes 48–72 h depending on cell culture requirements.
Figure 1 | An illustration of the BRET protocol using a microplate
luminometer. Reprinted with permission from ref. 1.
©
2006
Nature
Publishing
Group
http://www.nature.com/natureprotocols
338 | VOL.1 NO.1 | 2006 | NATURE PROTOCOLS
PROTOCOL
sent an increase or decrease in the number and/or rate of inter-
actions between the proteins of interest, but they may also result
from conformational changes that influence the relative position-
ing of the donor and acceptor molecules. This needs to be taken
into account when interpreting the data. Studies of constitutive
interactions using BRET must provide evidence for interaction
specificity1, typically using saturation or competition assays as dis-
cussed below. In particular, suitable negative controls are required,
such as similar proteins expressed at similar levels in the same cel-
lular compartment. Positive controls should also be included when
possible. The donor covalently fused to the acceptor is a useful
construct for checking that the protocol is working and to com-
pare between assays. A better positive control is to use a combi-
nation of fusion proteins, which are known to interact, and are
expressed at similar levels and in the same cellular compartment
as the proteins under investigation. The BRET technology is most
suited to studying protein-protein interactions in live cells where
they are expressed in a near-physiological environment in the cor-
rect cellular compartment. Cell extracts or purified proteins can
be used to increase protein expression levels, but these situations
are less likely to provide physiologically relevant data and are par-
ticularly prone to the problems associated with overexpression and
artifactual interactions. The BRET technique is not suitable for
investigating endogenous proteins because of the need for fusion
proteins. Furthermore, unless cells are fractionated19, BRET can-
not currently provide information about subcellular location.
Saturation assays
The use of saturation assays to provide evidence for the specificity
of constitutive interactions has been put forward by Bouvier and
coworkers20. Using the procedure described in this protocol, sev-
eral samples that coexpress a constant amount of donor-labeled
protein with increasing amounts of acceptor-labeled protein are
assayed. Ideally, the lowest level of donor-labeled protein expres-
sion that results in a reliable luminescence signal should be used.
It is thought that specific interactions result in BRET signal satu-
ration at high concentrations of acceptor-labeled protein. By con-
trast,nonspecific interactions are believed to result in BRET signals
that increase with increasing concentrations of acceptor-labeled
protein in a quasi-linear manner20. Relative acceptor fluorescence
and donor luminescence levels should be established in addition to
the BRET signals. Consequently, saturation curves can be gener-
ated using nonlinear regression, plotting BRET signal against the
ratio of acceptor fluorescence over donor luminescence.
Competition assays
Evidence for specificity can also be provided by showing com-
petitive inhibition of interactions. Using the procedure described
in this protocol, donor- and acceptor-labeled proteins are coex-
pressed with unlabeled protein that competes for interactions
with the labeled proteins. Unlabeled protein can be expressed
at a single concentration (usually excess)9 or at increasing con-
centrations21. In parallel, a negative control sample coexpressing
TABLE 1 | Comparison of BRET1, eBRET and BRET2.
BRET1 eBRET BRET2
Donor Renilla luciferase Renilla luciferase Renilla luciferase
Acceptor YFP (or EGFP) YFP (or EGFP) GFP2 or GFP10
Substrate Coelenterazine h EnduRen DeepBlueC
Example filters
‘Short-wavelength emission’ (nm): 440−500 (YFP)
400−475 (EGFP)
440−500 (YFP)
400−475 (EGFP)
370−450
‘Long-wavelength emission’ (nm): 510−590 (YFP)
500−550 (EGFP)
510−590 (YFP)
500−550 (EGFP)
500−530
Characteristics
Donor and acceptor emission peak separation
(nm)
∼55 (YFP)
∼35 (EGFP)
∼55 (YFP)
∼35 (EGFP)
∼115
Substrate relative quantum yield >100 >100 1
Possible detection duration Up to 1 h Several hours Few seconds
Substrate injection required No No Yes
Substrate preincubation 0 1.5 h 0
Possible in live cells Yes Yes Yes
Possible in cell extracts or purified proteins Yes No Yes
Optimal final substrate concentration (µM) 5 60 5
Typical assay buffer D-PBS with CaCl2, MgCl2 and D-
glucose
HEPES-buffered phenol
red−free medium
D-PBS with CaCl2, MgCl2 and D-
glucose
YFP, yellow fluorescent protein (includes Topaz, Venus and Citrine); EGFP, enhanced green fluorescent protein. See ref. 1 for a detailed
discussion.
©
2006
Nature
Publishing
Group
http://www.nature.com/natureprotocols
NATURE PROTOCOLS | VOL.1 NO.1 | 2006 | 339
PROTOCOL
donor- and acceptor-labeled proteins with unlabeled protein that
does not compete for interactions with the labeled proteins is includ-
ed. It is important to measure relative protein expression levels (as
determined by fluorescence or luminescence detection) to ensure
that an attenuated BRET signal is not a consequence of reducing
labeled-protein expression as the unlabeled protein is introduced.
The different BRET derivations
Three BRET derivations, BRET1, BRET2 and eBRET, are now avail-
able for general laboratory use and their characteristics are summa-
rized in Table 1. The advantage of BRET2 over BRET1 and eBRET
is the superior donor and acceptor emission peak separation when
using the substrate DeepBlueC. This enables donor and acceptor
emissions to be distinguished much more easily, thereby reducing
the background22. But the low quantum yield and rapid decay of
DeepBlueC mean that more cells are needed to achieve sufficiently
high luminescence levels for BRET detection, and highly sensi-
tive instrumentation is required22. Furthermore, BRET2 assays are
effectively impractical without injection of substrate into samples.
The advantage of eBRET over both BRET1 and BRET2 is the
capacity to monitor a particular interaction in real time over
several hours or measure multiple samples on the scale of high-
throughput screening without the need for frequent substrate
addition23. The limitation with both scenarios is cell viability, but
by assaying cells in HEPES-buffered medium, cell viability does
not appear to be compromised substantially for at least 6 h at
37 °C (ref. 23). eBRET should be carried out using live cells at 37
°C. It cannot be used for assaying cell extracts or purified proteins
because of its dependence on endogenous esterases.
BRET imaging
The use of an intensified charge-coupled device (CCD) camera to
image BRET is an exciting technological derivation. BRET from
single cells can be visualized21 and the potential for detecting BRET
in whole animals has recently been demonstrated by imaging trans-
fected cells injected intravenously or subcutaneously into mice24.
Furthermore, the development of higher-quantum-yield donors
may well allow subcellular BRET detection in the near future. The
specialized instrumentation required for BRET imaging clearly
necessitates a considerably different protocol to that needed for
BRET detected by luminometry or spectroscopy. Consequently, this
derivation is beyond the scope of the protocol described herein.
Fluorescence and luminescence detection
It is desirable to detect relative fluorescence and luminescence
levels for evaluating relative expression levels, and this is par-
ticularly important when carrying out saturation and competi-
tion assays20. Fluorescence can be detected using a fluorometer,
scanning spectrometer or flow cytometer. Using a fluorometer
is the most rapid and straightforward method, with scanning
spectrometry providing additional spectral information, should
that be required. Both of these methods allow subsequent lumi-
nescence measurements to be made on the same aliquot of cells,
which enables relative fluorescence over luminescence ratios to
be established for the same sample. Flow cytometry addition-
ally allows evaluation of the percentage of cells expressing fluo-
rophore and the mean fluorescence. Subsequent measurement
of luminescence on the same aliquot of cells, however, is less
practical.
BRET detection
BRET can be detected using a luminometer (microplate or sin-
gle-tube) or scanning spectrometer. The luminometer must have
the capacity to sequentially or simultaneously detect filtered light
within two distinct wavelength windows and ideally include tem-
perature control and kinetics software (required for eBRET).
Injectors are required for BRET2 and are desirable for BRET1 and
eBRET. Single-tube luminometers are more sensitive than micro-
plate luminometers, but they are clearly less practical for assay-
ing multiple samples. Scanning spectroscopy provides additional
spectral information, but the time required to complete each scan
limits the number of repeats per sample, the number of samples
that can be measured and the ability to generate time courses. The
rapid decay of DeepBlueC limits the use of scanning spectroscopy
for BRET2.
Donor-only controls
When investigating constitutive interactions, cells expressing only
the donor-labeled proteins should be included to establish the
background signal. There is a second option for analyzing BRET
data if investigating a ligand-induced interaction23. This does not
require ‘donor-only’ samples and is theoretically more accurate
as aliquots of the same preparation containing the same protein
expression are used to assess the ‘interacting protein’ and ‘back-
ground’ emissions.
MATERIALS
REAGENTS
• cDNA for functionally validated fusion proteins in suitable expression vectors
• Suitable cells for transfection (e.g.,HEK293 and COS-7)
• 6-well cell culture plates (BD Falcon,cat.no.351146)
• Suitable growth medium,such as Complete Medium (Dulbecco’s modified
Eagle’s medium (DMEM; Gibco,cat.no.11960-044) containing 0.3 mg ml−1
glutamine (Gibco,cat.no.25030-081),100 IU ml−1 penicillin,100 µg ml−1
streptomycin (Gibco,cat.no.15140-122) and 10% fetal calf serum (Gibco,cat.
no.16000-044)) for HEK293 and COS-7 cells
• Transfection system or reagent; examples of reagents include Genejuice
(Novagen,cat.no.70967) and Polyfect (Qiagen,cat.no.301 107)
! CAUTION Wear suitable protective clothing,gloves and safety glasses.
• 96-well white cell culture plate (Nunc,cat.no.136101) for assaying adherent
cells
• Phenol red−free medium,such as the Complete Medium described above,but
containing DMEM without phenol red (Gibco,cat.no.31053-028); requires 25
mM HEPES (Sigma,cat.no.H-3034) when used as eBRET assay buffer
• 0.05% trypsin−0.53 mM EDTA (Gibco,cat.no.15400-054) ! CAUTION Wear
suitable protective clothing,gloves and safety glasses.
• Dulbecco’s phosphate-buffered saline (D-PBS) containing 0.1 g l−1 CaCl2,
0.1 g l−1 MgCl2·6H2O and 1 g l−1 D-glucose (Invitrogen,cat.no.14287-080)
• 96-well isoplates: white for BRET (Perkin Elmer,cat.no.1450-581) and black
for fluorescence detection (OptiPlate-96F; Perkin Elmer,cat.no.6005270)
• For BRET1 only: 500 µM coelenterazine h (Molecular Probes,cat.no.C-6780)
in methanol as luciferase substrate stock solution (Sigma,cat.no.10158.6B)
! CAUTION Wear suitable protective clothing,gloves and safety glasses.
▲ CRITICAL Store at -20 °C protected from light.
• For BRET2 only: 1 mM DeepBlueC (PerkinElmer,cat.no.6310-100C) in
©
2006
Nature
Publishing
Group
http://www.nature.com/natureprotocols
340 | VOL.1 NO.1 | 2006 | NATURE PROTOCOLS
PROTOCOL
anhydrous or absolute ethanol as luciferase substrate stock solution (Merck,cat.
no.1.00986) ! CAUTION Wear suitable protective clothing,gloves and safety
glasses.▲ CRITICAL Store at –20 °C protected from light.
•For eBRET only: 60 mM EnduRen (Promega,cat.no.E6482) in
dimethylsulfoxide (DMSO) as luciferase substrate stock solution (Sigma,
cat.no.D-2650) ! CAUTION Readily absorbed through skin.Wear suitable
protective clothing,gloves and safety glasses.▲ CRITICAL Store at –20 °C
protected from light.
•Ligand or other modulating reagent depending on interaction being assayed
! CAUTION Wear suitable protective clothing,gloves and safety glasses.
EQUIPMENT
•Standard cell culture facility including Class II biological safety cabinet,such
as BH2000 series (Clyde-Apac),and 37 °C incubator with 5% CO2,such as the
Heracell (Heraeus)
•Microplate luminometer; examples include theVICTOR Light (PerkinElmer,
cat.no.1420-060),Mithras LB 940 (Berthold Technologies) and FLUOstar
Optima or POLARstar Optima (BMG Labtech)
•(Optional) Single-tube luminometer with filter switch capability such as the
Sirius C (Berthold Detection Systems,cat.no.11040050)
•(Optional) Scanning spectrometer with 96-well plate capability; examples
include the Spex fluorolog or fluoromax (JobinYvon),the Cary Eclipse
(Varian) and the FlexStation II (Molecular Devices)
•(Optional) Fluorometer; examples include the EnVision (Perkin Elmer,cat.no.
2102-0010),Mithras LB 940 (Berthold Technologies) and FLUOstar Optima or
POLARstar Optima (BMG Labtech)
•(Optional) Flow cytometer,such as the FACS Calibur (Becton Dickinson,cat.
no.1641)
PROCEDURE
Preparation
1| Select the donor, acceptor and substrate combination, and consequently the filter combination if using a luminometer for
BRET detection1 (Table 1).
2| Generate fusion constructs in a suitable expression vector consisting of the cDNA for the protein of interest inserted in-
frame with the cDNA for the donor or acceptor molecule, incorporating the cDNA for a suitable linker region, if appropriate.
Remove the stop codon separating the cDNA sequences by mutagenesis so that a single fusion protein is expressed after
transfection.
3| Validate the labeling of proteins of interest, including suitable control proteins, by comparing labeled and wild-type
proteins in functional assays. Check that luminescence or fluorescence is detectable. If possible, use confocal microscopy to
visualize correct cellular localization of acceptor-labeled protein.
▲ CRITICAL STEP Establish that protein function is not compromised by the addition of donor or acceptor molecules.
Insufficient demonstration of correct cellular function must be taken into account when interpreting data.
? TROUBLESHOOTING
4| Reconstitute and store the luciferase substrate stock solution containing coelenterazine h for BRET1 or DeepBlueC for
BRET2 (both option A), or EnduRen for eBRET (option B).
(A) For BRET1 or BRET2:
(i) Allow the vial of lyophilized coelenterazine h or DeepBlueC to equilibrate to ambient temperature before opening to
avoid condensation.
▲ CRITICAL STEP Protect from light.
(ii) Reconstitute coelenterazine h with methanol, and DeepBlueC with anhydrous or absolute ethanol. Coelenterazine h can
also be reconstituted in ethanol.
▲ CRITICAL STEP Do not dissolve in dimethylsulfoxide (DMSO).
(iii) Agitate gently until resuspended, potentially a few minutes.
(iv) Aliquot (volumes depend on expected rate of use) and store desiccated and protected from light at –20 °C.
(B) For eBRET:
(i) Allow the vial of lyophilized EnduRen to equilibrate to ambient temperature before opening to avoid condensation.
▲ CRITICAL STEP Protect from light.
(ii) Reconstitute in tissue-culture grade dimethylsulfoxide (DMSO).
(iii) Vortex to resuspend.
▲ CRITICAL STEP Repeated vortexing may be required. Resuspension may take up to 10 min and may require warming to 37 °C.
(iv) Aliquot and store protected from light at –20 °C.
Cell culture
5| Aliquot cells into a 6-well cell culture plate in a suitable growth medium. The number of cells required in each well will
depend on size, growth rate and transfection system to be used, but typically they should be 50–80% confluent after 24 h
if being transiently transfected. For example, COS-7 cells are typically plated out at a density of 120,000 cells per well. Cells
stably expressing fusion proteins can also be used, with clonal stable cell lines having the advantage of homogeneous protein
expression levels. Maintain at 37 °C, 5% CO2 (for mammalian cells).
©
2006
Nature
Publishing
Group
http://www.nature.com/natureprotocols
NATURE PROTOCOLS | VOL.1 NO.1 | 2006 | 341
PROTOCOL
6| Typically 24 h after plating, coexpress the donor- and acceptor-labeled proteins of interest in cells using a suitable
transfection or expression system if these proteins are not already stably expressed. If required, produce a population of
cells expressing only the donor-labeled proteins at similar expression levels to those in the samples coexpressing donor- and
acceptor-labeled proteins. This requirement depends on the form of analysis to be used. Aim to achieve a physiologically
relevant expression level that still provides a detectable BRET signal. Samples with various combinations of protein expression
levels may be required if saturation or competition assays are being carried out. Express suitable positive- and negative-control
proteins in parallel. Maintain a population of untransfected cells in parallel for establishing background levels of fluorescence
and luminescence.
? TROUBLESHOOTING
7| If pretreating with ligand or other reagent, add these at an appropriate time between transfection and BRET detection.
Include vehicle-treated samples in parallel.
8| Interactions can be monitored in adherent cells (option A), in cell suspension (option B), in cell extracts or between
purified proteins (option C). The optimal expression time for transiently transfected fusion proteins should be established as it
varies from 24 to 72 h.
(A) In adherent cells:
(i) Detach the cells (e.g., using trypsin-EDTA) typically 24 h after transfection.
(ii) Resuspend the cells in phenol red−free medium (HEPES-buffered if eBRET is to be carried out in this medium) and
aliquot into a 96-well white tissue culture plate (40−100 µl per well). Initially titrate cells to establish a suitable
dilution. Higher cell concentrations are required for BRET2 compared with BRET1 or eBRET.
(iii) Incubate at 37 °C, 5% CO2 (mammalian cells) for an additional 24 h to allow attachment, and either assay in the
existing phenol red−free medium or remove medium and replace with assay buffer.
▲ CRITICAL STEP Extreme care must be taken not to detach cells. If removing the medium, washing is not recommended.
? TROUBLESHOOTING
(B) In cell suspension:
(i) Detach the cells (e.g., using trypsin-EDTA) immediately before the assay (24–48 h after transfection) and resuspend in
assay buffer.
(ii) Aliquot cells into a 96-well white isoplate (40–100 µl per well).
(C) Cell extracts or purified proteins:
(i) Typically prepare cell extracts or purified proteins 24−48 h after transfection.
(ii) Aliquot into a 96-well white isoplate (40–100 µl per well).
Fluorescence and luminescence detection
9| Fluorescence can be measured in a fluorometer or scanning spectrometer (both option A), or flow cytometer (option B).
Use a fluorometer or scanning spectrometer with 96-well capability if subsequent detection of unfiltered luminescence is to be
carried out using the same aliquots (Step 10).
(A) Fluorometer or scanning spectrometer:
(i) Measure the relative fluorescence from aliquots of each sample in D-PBS (40−100 µl per well in a 96-well black
isoplate), directly exciting the acceptor fluorophore with laser light of a suitable wavelength.
(ii) Filter the emission to avoid detection of excitation light and correct for background fluorescence from
untransfected cells.
? TROUBLESHOOTING
(B) Flow cytometer:
(i) Dilute aliquots of samples in D-PBS to achieve an appropriate volume.
(ii) Measure the relative fluorescence, directly exciting the acceptor fluorophore with laser light of a suitable wavelength.
(iii) Filter the emission to avoid detection of excitation light and correct for background fluorescence from untransfected
cells.
? TROUBLESHOOTING
10| Taking the same aliquots as assessed in the fluorometer or scanning spectrometer, add coelenterazine h or DeepBlueC and
measure unfiltered luminescence in a luminometer such as that used for BRET detection. Correct for background luminescence
from untransfected cells unless this is shown to be negligible.
? TROUBLESHOOTING
©
2006
Nature
Publishing
Group
http://www.nature.com/natureprotocols
342 | VOL.1 NO.1 | 2006 | NATURE PROTOCOLS
PROTOCOL
BRET detection
11| Dilute the substrate stock (coelenterazine h, DeepBlueC or EnduRen) to a working concentration in assay buffer, typically
10× final concentration (Table 1).
▲ CRITICAL STEP Dilute immediately before adding to samples and protect from light. For EnduRen, preincubate the assay
buffer at 37 °C to avoid precipitation.
12| Add the luciferase substrate to the samples. For eBRET, use option A; for BRET1 use option B; and for BRET2 use option C.
(A) For eBRET, add EnduRen to live cells at least 1.5 h before BRET detection1. Substrate addition can be verified by
luminescence detection. Counts are low at this pre-equilibrium stage, but they are substantially higher than background. For
mammalian cells, incubate at 37 °C, 5% CO2.
(B) For BRET1, add coelenterazine h to the samples immediately before BRET detection. As BRET1 data are generally reliable
for 30–60 min after coelenterazine h addition (depending on the initial signal), injection of the substrate is optional.
(C) For BRET2, add DeepBlueC to each well immediately before detecting emission from that well.
▲ CRITICAL STEP In practical terms, this requires the use of injectors.
13| Detect the light emissions using either a luminometer (option A) or scanning spectrometer (option B). The rapid decay of
DeepBlueC limits the use of scanning spectroscopy for BRET2.
▲ CRITICAL STEP eBRET should be carried out at 37 °C to avoid EnduRen precipitation.
(A) If using a luminometer with the appropriate filter combination (Table 1), detect the light emission through both
filters (simultaneously or sequentially, typically 0.5–5 s per filter) before proceeding to the next sample. Simultaneous
detection reduces measurement times and is potentially more accurate, particularly for BRET2 because of the rapid decay
of DeepBlueC.
? TROUBLESHOOTING
(B) If using scanning spectrometry for BRET1 or eBRET, detect light emission from 400–600 nm, typically using a 10 nm slit
width and 2 s per increment.
? TROUBLESHOOTING
14| Repeat readings as required, within the timeframe allowed by substrate stability (Table 1) and cell viability. BRET2 repeats
require re-addition of DeepBlueC. Repeats can be automated with appropriate kinetics software.
15| Add ligands or other reagents, if desired, and continue collecting readings. Use a luminometer and inject these agents if
early post-addition time points are required (<1 min). Include vehicle-treated samples in parallel.
● TIMING
Preparation (Steps 1–4): potentially several weeks depending on the complexity of cloning and validation required.
Cell culture (Steps 5–8): typically 48–72 h (less if stable cell lines, cell extracts or purified proteins have already been prepared).
Fluorescence and luminescence detection (steps 9–10): a few minutes (depending on the number of samples).
BRET detection (Steps 11–15): minutes to hours depending on the number of samples, number of repeats and length of time
course. eBRET requires addition of EnduRen at least 1.5 h before commencing BRET detection.
? TROUBLESHOOTING
For troubleshooting guidance see Table 2.
TABLE 2 | Troubleshooting table.
STEP PROBLEM POSSIBLE REASON SOLUTION
3 Fusion protein is not
functioning correctly
There was an error in construct
generation
Sequence and check entire fusion protein cDNA
Label is interfering with protein
function
Reposition the label to the other end of the protein (or
possibly an internal peripheral loop or linker region), or
increase linker length between protein of interest and label
6 Cells are dying after
transfection
Cytotoxicity of transfection reagent Refer to the manufacturer’s guidelines, or consider using
alternative reagents
Cytotoxicity of GFP fusion proteins Titrate down expression of GFP fusion proteins
©
2006
Nature
Publishing
Group
http://www.nature.com/natureprotocols
NATURE PROTOCOLS | VOL.1 NO.1 | 2006 | 343
PROTOCOL
ANTICIPATED RESULTS
Calculate the BRET signals using option A (below) if a luminometer was used or option B if a scanning spectrometer was used.
If autoluminescence from untransfected cells is not negligible, this should be subtracted from emissions before calculating the
BRET signal.
(A) If a luminometer was used, calculate the BRET signal using the following equation:
For BRET1 and eBRET, note that light is emitted through the ‘long-wavelength emission’ filter, even in the absence of
acceptor expression or ligand addition. This is ‘bleed-through’ from the donor emission1.
There are two options for using the equation. All interactions can be analyzed using option (i), but ligand-induced interactions
can also be analyzed using option (ii). Option (ii) eliminates the need for ‘donor-only’ samples and is theoretically more
accurate as aliquots of the same preparation containing the same amount of protein are used to assess the ‘interacting protein’
and ‘background emissions’23.
(i) The ‘emissions from interacting proteins’ are from samples coexpressing donor and acceptor fusion proteins and the
‘background emissions’ from samples expressing only donor fusion proteins.
(ii) If the interaction is ligand-induced, the ‘emissions from interacting proteins’ can be from ligand-treated samples and
the ‘background emissions’ from vehicle-treated samples.
(B) Data derived using scanning spectroscopy can be analyzed using option (i) or (ii):
(i) Normalize the emission spectra by defining the donor peak emission intensity as 1. Calculate the BRET signal as the
area under the curve to which the acceptor emission contributes (typically 500−550 nm) minus the corresponding area
observed when only the donor is present.
(ii) Alternatively, establish areas under the curve within wavelength windows that correspond to the luminometer filters and
calculate the BRET ratio as described for luminometer-derived data (option A).
TABLE 2 | Troubleshooting table (continued).
STEP PROBLEM POSSIBLE REASON SOLUTION
8 Cells are detaching despite
extreme care.
Cells are not sufficiently adherent Consider using different plastics or coating wells (e.g., with
poly(L-lysine))
9, 10 and 13 There are low relative
luminescence or
fluorescence counts
There is poor protein expression Optimize or consider an alternative transfection strategy
The cell number is low Increase cell concentratio
Substrate was not added Add substrate to each well
Substrate is not viable Check expiry and storage conditions. Consider using new
aliquot of substrate
Reducing agent present Ensure buffers do not include reducing agents such as ascorbic
acid
Instrumentation is not functioning
correctly
Check instrument setup and consider recalibration
10 and 13 Luminescence signal is
detected from untransfected
cells upon substrate addition
There is spontaneous
autoluminescence
Correct for signal from untransfected cells
Consider removing serum or BSA if present in assay buffer
Problem should be reduced or alleviated by using EnduRen
Results BRET signal is not detected
despite validated fusion
proteins generating
high luminescence or
fluorescence counts
Distance between donor and
acceptor is too great
Test alternative combinations of fusion proteins with labels in
different positions
Relative orientation of donor and
acceptor is unsuitable for BRE
Increase linker length between protein of interest and label
May be due to ligand if interaction
is ligand-induce
Check and optimize potency of ligand required
BRET signal (or BRET radio) =
‘long-wavelength emission from interacting proteins’
‘short-wavelength emission from interacting proteins’
‘background long-wavelength emission’
‘background short-wavelength emission’
–
©
2006
Nature
Publishing
Group
http://www.nature.com/natureprotocols
344 | VOL.1 NO.1 | 2006 | NATURE PROTOCOLS
PROTOCOL
An example of raw BRET data is shown in Table 3 and is derived from data generated in three independent experiments. Two
aliquots of the same cell sample were treated with ligand or vehicle and monitored in real-time. The data shown was observed
20 min after ligand or vehicle addition. In this example, option A(ii) has been used for data analysis. Emissions were detected
through the two filters and the ‘long-wavelength emission’ over ‘short-wavelength emission’ ratios calculated. The BRET ratio
was then calculated as: Ligand-treated emission ratio - vehicle-treated emission ratio = 2.3429 - 2.1396 = 0.203.
Statistical analysis of BRET data should be carried out using ANOVA with suitable post-tests, or using Student’s t-tests
where appropriate. One method is to determine the ‘long-wavelength emission’ over ‘short-wavelength emission’ ratio for the
interacting proteins and background separately. The variance in these ratios can then be compared to establish whether they
are significantly different. For the example shown in Table 3, the ligand-treated and vehicle-treated emission ratios were found
to be significantly different (P = 0.0086) using an unpaired Student’s t-test.
Anticipated results for ligand- (or reagent-) modulated interactions are illustrated in Figure 2. eBRET is particularly suitable
if kinetic profiles beyond 30−60 min are required.
Anticipated results for constitutive interactions should not only exhibit significantly higher BRET signals than
appropriate negative controls, but also
demonstrate interaction specificity
by incorporating competition or
saturation data generation into the
experimental design (see ref. 1 for
extensive discussion).
It is important to note that a
negative BRET result is not conclusive
evidence for a lack of protein-protein
interaction. The extremely high
dependency on distance between donor
and acceptor means that functionally
validated fusion proteins may interact
without a BRET signal being detectable.
See Table 2 for troubleshooting
guidance.
TABLE 3 | An example of raw BRET data and its analysis.
Experiment Ligand-treated Vehicle-treated
Long-wavelength
emission
Short-wavelength
emission
Long:short
wavelength ratio
Long-wavelength
emission
Short-wavelength
emission
Long:short
wavelength ratio
1 313,159 134,012 2.3368 404,242 191,367 2.1124
2 154,900 65,013 2.3826 130,677 59,082 2.2118
3 256,707 111,166 2.3092 353,669 168,849 2.0946
Mean 2.3429 2.1396
s.d. 0.0371 0.0632
a b
Figure 2 | Examples of theoretical BRET data from ligand (reagent) modulated interactions.
(a) Dose-response curves can be generated by nonlinear regression, permitting calculation of EC50
(half-maximal effective concentration) values. (b) Changes in BRET ratio can be monitored over time
before and after addition of ligand and/or other modulators (the dashed line denotes data produced
upon addition of vehicle instead of ligand). Apparent association (or dissociation) rate constants can
be calculated from such data.
COMPETING INTERESTS STATEMENT The authors declare competing financial
interests (see the HTML version of this article for details).
Published online at http://www.natureprotocols.com/
Reprints and permissions information is available online at http://npg.nature.
com/reprintsandpermissions/
1. Pfleger, K.D. & Eidne, K.A. Illuminating insights into protein-protein
interactions using bioluminescence resonance energy transfer (BRET). Nat.
Methods 3, 165−174 (2006).
2. Milligan, G. & Bouvier, M. Methods to monitor the quaternary structure of
G-protein−coupled receptors. FEBS J. 272, 2914−2925 (2005).
3. Boute, N., Jockers, R. & Issad, T. The use of resonance energy transfer in
high-throughput screening: BRET versus FRET. Trends Pharmacol. Sci. 23,
351−354 (2002).
4. Pfleger, K.D.G. & Eidne, K.A. New technologies: bioluminescence resonance
energy transfer (BRET) for the detection of real time interactions involving G-
protein coupled receptors. Pituitary 6, 141−151 (2003).
5. Wu, P. & Brand, L. Resonance energy transfer: methods and applications.
Anal. Biochem. 218, 1−13 (1994).
6. Xu, Y., Piston, D.W. & Johnson, C.H. A bioluminescence resonance energy
transfer (BRET) system: application to interacting circadian clock proteins.
Proc. Natl. Acad. Sci. USA 96, 151−156. (1999).
7. Subramanian, C. et al. The Arabidopsis repressor of light signaling, COP1,
is regulated by nuclear exclusion: mutational analysis by bioluminescence
resonance energy transfer. Proc. Natl. Acad. Sci. USA 101, 6798−6802 (2004).
8. Angers, S. et al. Detection of beta 2-adrenergic receptor dimerization in
living cells using bioluminescence resonance energy transfer (BRET). Proc.
Natl. Acad. Sci. USA 97, 3684−3689 (2000).
9. Kroeger, K.M., Hanyaloglu, A.C., Seeber, R.M., Miles, L.E. &
Eidne, K.A. Constitutive and agonist-dependent homo-oligomerization
of the thyrotropin-releasing hormone receptor. Detection in living cells
using bioluminescence resonance energy transfer. J. Biol. Chem. 276,
12736−12743 (2001).
10. Pfleger, K.D.G. & Eidne, K.A. Monitoring the formation of dynamic
G-protein-coupled receptor-protein complexes in living cells. Biochem. J.
©
2006
Nature
Publishing
Group
http://www.nature.com/natureprotocols
NATURE PROTOCOLS | VOL.1 NO.1 | 2006 | 345
PROTOCOL
385, 625−637 (2005).
11. Scaffidi, A.K. et al. α(v)β(3) Integrin interacts with the transforming growth
factor β (TGFβ) type II receptor to potentiate the proliferative effects of
TGFβ1 in living human lung fibroblasts. J. Biol. Chem. 279, 37726−37733
(2004).
12. Brown, R.J. et al. Model for growth hormone receptor activation based on
subunit rotation within a receptor dimer. Nat. Struct. Mol. Biol. 12, 814−821
(2005).
13. Michelini, E., Mirasoli, M., Karp, M., Virta, M. & Roda, A. Development of a
bioluminescence resonance energy-transfer assay for estrogen-like compound
in vivo monitoring. Anal. Chem. 76, 7069−7076 (2004).
14. Garside, H. et al. Glucocorticoid ligands specify different interactions with
NF-kappaB by allosteric effects on the glucocorticoid receptor DNA binding
domain. J. Biol. Chem. 279, 50050−50059 (2004).
15. de Virgilio, M., Kiosses, W.B. & Shattil, S.J. Proximal, selective, and dynamic
interactions between integrin alphaIIbbeta3 and protein tyrosine kinases in
living cells. J. Cell. Biol. 165, 305−311 (2004).
16. Yung, T.M., Sato, S. & Satoh, M.S. Poly(ADP-ribosyl)ation as a DNA damage-
induced post-translational modification regulating poly(ADP-ribose)
polymerase-1-topoisomerase I interaction. J. Biol. Chem. 279, 39686−39696
(2004).
17. Trevaskis, J. et al. Src homology 3-domain growth factor receptor-bound
2-like (endophilin) interacting protein 1, a novel neuronal protein that
regulates energy balance. Endocrinology 146, 3757−3764 (2005).
18. Germain-Desprez, D., Bazinet, M., Bouvier, M. & Aubry, M. Oligomerization
of transcriptional intermediary factor 1 regulators and interaction with
ZNF74 nuclear matrix protein revealed by bioluminescence resonance energy
transfer in living cells. J. Biol. Chem. 278, 22367−22373 (2003).
19. Terrillon, S. et al. Oxytocin and vasopressin V1a and V2 receptors from
constitutive homo- and heterodimers during biosynthesis. Mol. Endocrinol.
17, 677−691 (2003).
20. Mercier, J.F., Salahpour, A., Angers, S., Breit, A. & Bouvier, M. Quantitative
assessment of β1- and β2-adrenergic receptor homo- and heterodimerization
by bioluminescence resonance energy transfer. J. Biol. Chem. 277,
44925−44931 (2002).
21. Ayoub, M.A. et al. Monitoring of ligand-independent dimerization and
ligand-induced conformational changes of melatonin receptors in living
cells by bioluminescence resonance energy transfer. J. Biol. Chem. 277,
21522−21528 (2002).
22. Hamdan, F.F., Audet, M., Garneau, P., Pelletier, J. & Bouvier, M. High-
throughput screening of G protein-coupled receptor antagonists using a
bioluminescence resonance energy transfer 1−based β-arrestin2 recruitment
assay. J. Biomol. Screen. 10, 463−475 (2005).
23. Pfleger, K.D. et al. Extended bioluminescence resonance energy transfer
(eBRET) for monitoring prolonged protein-protein interactions in live cells.
Cell. Signal; Advance online publication 21 February 2006 (doi: 10.1016/
j.cellsig.2006.01.004).
24. De, A. & Gambhir, S.S. Noninvasive imaging of protein-protein interactions
from live cells and living subjects using bioluminescence resonance energy
transfer. FASEB J. 19, 2017−2019 (2005).
©
2006
Nature
Publishing
Group
http://www.nature.com/natureprotocols

More Related Content

Similar to Bioluminescence Resonance Energy Transfer (BRET) ARTUCLO 1.pdf

лекция 2 методы анализа
лекция 2 методы анализалекция 2 методы анализа
лекция 2 методы анализаnizhgma.ru
 
biophysical &biomedical analysis.pptx
biophysical  &biomedical analysis.pptxbiophysical  &biomedical analysis.pptx
biophysical &biomedical analysis.pptxbreenaawan
 
Analysis of bioreactor parameters online and offline
Analysis of bioreactor parameters online and offlineAnalysis of bioreactor parameters online and offline
Analysis of bioreactor parameters online and offlinevikash_94
 
Protein protein interaction, functional proteomics
Protein protein interaction, functional proteomicsProtein protein interaction, functional proteomics
Protein protein interaction, functional proteomicsKAUSHAL SAHU
 
Specificity and Evolvability in Eukaryotic Protein Interaction Networks
Specificity and Evolvability in Eukaryotic Protein Interaction NetworksSpecificity and Evolvability in Eukaryotic Protein Interaction Networks
Specificity and Evolvability in Eukaryotic Protein Interaction Networkspedrobeltrao
 
High throughput approaches to understanding gene function and mapping archite...
High throughput approaches to understanding gene function and mapping archite...High throughput approaches to understanding gene function and mapping archite...
High throughput approaches to understanding gene function and mapping archite...Tintumann
 
Functional proteomics, and tools
Functional proteomics, and toolsFunctional proteomics, and tools
Functional proteomics, and toolsKAUSHAL SAHU
 
Main Exam Applied biochemistry final year
Main Exam Applied biochemistry final yearMain Exam Applied biochemistry final year
Main Exam Applied biochemistry final yearmarwaahmad357
 
Methods to Detect Protein-Protein Interaction
Methods to Detect Protein-Protein InteractionMethods to Detect Protein-Protein Interaction
Methods to Detect Protein-Protein InteractionCreative Proteomics
 
Structure-Function Analysis of POR Mutants
Structure-Function Analysis of POR MutantsStructure-Function Analysis of POR Mutants
Structure-Function Analysis of POR MutantsAYang999
 
Mol Cell Proteomics-2013-Koytiger-1204-13
Mol Cell Proteomics-2013-Koytiger-1204-13Mol Cell Proteomics-2013-Koytiger-1204-13
Mol Cell Proteomics-2013-Koytiger-1204-13Greg Koytiger
 
Techniques in proteomics
Techniques in proteomicsTechniques in proteomics
Techniques in proteomicsroshni mohan
 
Western blotting principle
Western blotting principleWestern blotting principle
Western blotting principleJames Waita
 
Online Size-Exclusion High-Performance Liquid Chromatography Light Scattering...
Online Size-Exclusion High-Performance Liquid Chromatography Light Scattering...Online Size-Exclusion High-Performance Liquid Chromatography Light Scattering...
Online Size-Exclusion High-Performance Liquid Chromatography Light Scattering...KBI Biopharma
 
Western blotting
Western blotting Western blotting
Western blotting Ravi kumar
 

Similar to Bioluminescence Resonance Energy Transfer (BRET) ARTUCLO 1.pdf (20)

лекция 2 методы анализа
лекция 2 методы анализалекция 2 методы анализа
лекция 2 методы анализа
 
biophysical &biomedical analysis.pptx
biophysical  &biomedical analysis.pptxbiophysical  &biomedical analysis.pptx
biophysical &biomedical analysis.pptx
 
Analysis of bioreactor parameters online and offline
Analysis of bioreactor parameters online and offlineAnalysis of bioreactor parameters online and offline
Analysis of bioreactor parameters online and offline
 
Protein protein interaction, functional proteomics
Protein protein interaction, functional proteomicsProtein protein interaction, functional proteomics
Protein protein interaction, functional proteomics
 
Specificity and Evolvability in Eukaryotic Protein Interaction Networks
Specificity and Evolvability in Eukaryotic Protein Interaction NetworksSpecificity and Evolvability in Eukaryotic Protein Interaction Networks
Specificity and Evolvability in Eukaryotic Protein Interaction Networks
 
High throughput approaches to understanding gene function and mapping archite...
High throughput approaches to understanding gene function and mapping archite...High throughput approaches to understanding gene function and mapping archite...
High throughput approaches to understanding gene function and mapping archite...
 
Nanoparticle corona study -
Nanoparticle corona study - Nanoparticle corona study -
Nanoparticle corona study -
 
REV
REVREV
REV
 
Functional proteomics, and tools
Functional proteomics, and toolsFunctional proteomics, and tools
Functional proteomics, and tools
 
Main Exam Applied biochemistry final year
Main Exam Applied biochemistry final yearMain Exam Applied biochemistry final year
Main Exam Applied biochemistry final year
 
Methods to Detect Protein-Protein Interaction
Methods to Detect Protein-Protein InteractionMethods to Detect Protein-Protein Interaction
Methods to Detect Protein-Protein Interaction
 
Structure-Function Analysis of POR Mutants
Structure-Function Analysis of POR MutantsStructure-Function Analysis of POR Mutants
Structure-Function Analysis of POR Mutants
 
Mol Cell Proteomics-2013-Koytiger-1204-13
Mol Cell Proteomics-2013-Koytiger-1204-13Mol Cell Proteomics-2013-Koytiger-1204-13
Mol Cell Proteomics-2013-Koytiger-1204-13
 
Site-Directed-Mutagenesis.pptx
Site-Directed-Mutagenesis.pptxSite-Directed-Mutagenesis.pptx
Site-Directed-Mutagenesis.pptx
 
Techniques in proteomics
Techniques in proteomicsTechniques in proteomics
Techniques in proteomics
 
M2-Cinical Immunology
M2-Cinical ImmunologyM2-Cinical Immunology
M2-Cinical Immunology
 
Western Blotting Principle
Western Blotting PrincipleWestern Blotting Principle
Western Blotting Principle
 
Western blotting principle
Western blotting principleWestern blotting principle
Western blotting principle
 
Online Size-Exclusion High-Performance Liquid Chromatography Light Scattering...
Online Size-Exclusion High-Performance Liquid Chromatography Light Scattering...Online Size-Exclusion High-Performance Liquid Chromatography Light Scattering...
Online Size-Exclusion High-Performance Liquid Chromatography Light Scattering...
 
Western blotting
Western blotting Western blotting
Western blotting
 

Recently uploaded

How to Give a Domain for a Field in Odoo 17
How to Give a Domain for a Field in Odoo 17How to Give a Domain for a Field in Odoo 17
How to Give a Domain for a Field in Odoo 17Celine George
 
Activity 01 - Artificial Culture (1).pdf
Activity 01 - Artificial Culture (1).pdfActivity 01 - Artificial Culture (1).pdf
Activity 01 - Artificial Culture (1).pdfciinovamais
 
Google Gemini An AI Revolution in Education.pptx
Google Gemini An AI Revolution in Education.pptxGoogle Gemini An AI Revolution in Education.pptx
Google Gemini An AI Revolution in Education.pptxDr. Sarita Anand
 
Jual Obat Aborsi Hongkong ( Asli No.1 ) 085657271886 Obat Penggugur Kandungan...
Jual Obat Aborsi Hongkong ( Asli No.1 ) 085657271886 Obat Penggugur Kandungan...Jual Obat Aborsi Hongkong ( Asli No.1 ) 085657271886 Obat Penggugur Kandungan...
Jual Obat Aborsi Hongkong ( Asli No.1 ) 085657271886 Obat Penggugur Kandungan...ZurliaSoop
 
UGC NET Paper 1 Mathematical Reasoning & Aptitude.pdf
UGC NET Paper 1 Mathematical Reasoning & Aptitude.pdfUGC NET Paper 1 Mathematical Reasoning & Aptitude.pdf
UGC NET Paper 1 Mathematical Reasoning & Aptitude.pdfNirmal Dwivedi
 
Unit-V; Pricing (Pharma Marketing Management).pptx
Unit-V; Pricing (Pharma Marketing Management).pptxUnit-V; Pricing (Pharma Marketing Management).pptx
Unit-V; Pricing (Pharma Marketing Management).pptxVishalSingh1417
 
ICT Role in 21st Century Education & its Challenges.pptx
ICT Role in 21st Century Education & its Challenges.pptxICT Role in 21st Century Education & its Challenges.pptx
ICT Role in 21st Century Education & its Challenges.pptxAreebaZafar22
 
Dyslexia AI Workshop for Slideshare.pptx
Dyslexia AI Workshop for Slideshare.pptxDyslexia AI Workshop for Slideshare.pptx
Dyslexia AI Workshop for Slideshare.pptxcallscotland1987
 
HMCS Max Bernays Pre-Deployment Brief (May 2024).pptx
HMCS Max Bernays Pre-Deployment Brief (May 2024).pptxHMCS Max Bernays Pre-Deployment Brief (May 2024).pptx
HMCS Max Bernays Pre-Deployment Brief (May 2024).pptxEsquimalt MFRC
 
Making communications land - Are they received and understood as intended? we...
Making communications land - Are they received and understood as intended? we...Making communications land - Are they received and understood as intended? we...
Making communications land - Are they received and understood as intended? we...Association for Project Management
 
Basic Civil Engineering first year Notes- Chapter 4 Building.pptx
Basic Civil Engineering first year Notes- Chapter 4 Building.pptxBasic Civil Engineering first year Notes- Chapter 4 Building.pptx
Basic Civil Engineering first year Notes- Chapter 4 Building.pptxDenish Jangid
 
Vishram Singh - Textbook of Anatomy Upper Limb and Thorax.. Volume 1 (1).pdf
Vishram Singh - Textbook of Anatomy  Upper Limb and Thorax.. Volume 1 (1).pdfVishram Singh - Textbook of Anatomy  Upper Limb and Thorax.. Volume 1 (1).pdf
Vishram Singh - Textbook of Anatomy Upper Limb and Thorax.. Volume 1 (1).pdfssuserdda66b
 
How to Manage Global Discount in Odoo 17 POS
How to Manage Global Discount in Odoo 17 POSHow to Manage Global Discount in Odoo 17 POS
How to Manage Global Discount in Odoo 17 POSCeline George
 
1029-Danh muc Sach Giao Khoa khoi 6.pdf
1029-Danh muc Sach Giao Khoa khoi  6.pdf1029-Danh muc Sach Giao Khoa khoi  6.pdf
1029-Danh muc Sach Giao Khoa khoi 6.pdfQucHHunhnh
 
Food safety_Challenges food safety laboratories_.pdf
Food safety_Challenges food safety laboratories_.pdfFood safety_Challenges food safety laboratories_.pdf
Food safety_Challenges food safety laboratories_.pdfSherif Taha
 
How to Create and Manage Wizard in Odoo 17
How to Create and Manage Wizard in Odoo 17How to Create and Manage Wizard in Odoo 17
How to Create and Manage Wizard in Odoo 17Celine George
 
1029 - Danh muc Sach Giao Khoa 10 . pdf
1029 -  Danh muc Sach Giao Khoa 10 . pdf1029 -  Danh muc Sach Giao Khoa 10 . pdf
1029 - Danh muc Sach Giao Khoa 10 . pdfQucHHunhnh
 
SKILL OF INTRODUCING THE LESSON MICRO SKILLS.pptx
SKILL OF INTRODUCING THE LESSON MICRO SKILLS.pptxSKILL OF INTRODUCING THE LESSON MICRO SKILLS.pptx
SKILL OF INTRODUCING THE LESSON MICRO SKILLS.pptxAmanpreet Kaur
 
Application orientated numerical on hev.ppt
Application orientated numerical on hev.pptApplication orientated numerical on hev.ppt
Application orientated numerical on hev.pptRamjanShidvankar
 

Recently uploaded (20)

How to Give a Domain for a Field in Odoo 17
How to Give a Domain for a Field in Odoo 17How to Give a Domain for a Field in Odoo 17
How to Give a Domain for a Field in Odoo 17
 
Activity 01 - Artificial Culture (1).pdf
Activity 01 - Artificial Culture (1).pdfActivity 01 - Artificial Culture (1).pdf
Activity 01 - Artificial Culture (1).pdf
 
Google Gemini An AI Revolution in Education.pptx
Google Gemini An AI Revolution in Education.pptxGoogle Gemini An AI Revolution in Education.pptx
Google Gemini An AI Revolution in Education.pptx
 
Jual Obat Aborsi Hongkong ( Asli No.1 ) 085657271886 Obat Penggugur Kandungan...
Jual Obat Aborsi Hongkong ( Asli No.1 ) 085657271886 Obat Penggugur Kandungan...Jual Obat Aborsi Hongkong ( Asli No.1 ) 085657271886 Obat Penggugur Kandungan...
Jual Obat Aborsi Hongkong ( Asli No.1 ) 085657271886 Obat Penggugur Kandungan...
 
UGC NET Paper 1 Mathematical Reasoning & Aptitude.pdf
UGC NET Paper 1 Mathematical Reasoning & Aptitude.pdfUGC NET Paper 1 Mathematical Reasoning & Aptitude.pdf
UGC NET Paper 1 Mathematical Reasoning & Aptitude.pdf
 
Unit-V; Pricing (Pharma Marketing Management).pptx
Unit-V; Pricing (Pharma Marketing Management).pptxUnit-V; Pricing (Pharma Marketing Management).pptx
Unit-V; Pricing (Pharma Marketing Management).pptx
 
ICT Role in 21st Century Education & its Challenges.pptx
ICT Role in 21st Century Education & its Challenges.pptxICT Role in 21st Century Education & its Challenges.pptx
ICT Role in 21st Century Education & its Challenges.pptx
 
Dyslexia AI Workshop for Slideshare.pptx
Dyslexia AI Workshop for Slideshare.pptxDyslexia AI Workshop for Slideshare.pptx
Dyslexia AI Workshop for Slideshare.pptx
 
HMCS Max Bernays Pre-Deployment Brief (May 2024).pptx
HMCS Max Bernays Pre-Deployment Brief (May 2024).pptxHMCS Max Bernays Pre-Deployment Brief (May 2024).pptx
HMCS Max Bernays Pre-Deployment Brief (May 2024).pptx
 
Making communications land - Are they received and understood as intended? we...
Making communications land - Are they received and understood as intended? we...Making communications land - Are they received and understood as intended? we...
Making communications land - Are they received and understood as intended? we...
 
Basic Civil Engineering first year Notes- Chapter 4 Building.pptx
Basic Civil Engineering first year Notes- Chapter 4 Building.pptxBasic Civil Engineering first year Notes- Chapter 4 Building.pptx
Basic Civil Engineering first year Notes- Chapter 4 Building.pptx
 
Vishram Singh - Textbook of Anatomy Upper Limb and Thorax.. Volume 1 (1).pdf
Vishram Singh - Textbook of Anatomy  Upper Limb and Thorax.. Volume 1 (1).pdfVishram Singh - Textbook of Anatomy  Upper Limb and Thorax.. Volume 1 (1).pdf
Vishram Singh - Textbook of Anatomy Upper Limb and Thorax.. Volume 1 (1).pdf
 
How to Manage Global Discount in Odoo 17 POS
How to Manage Global Discount in Odoo 17 POSHow to Manage Global Discount in Odoo 17 POS
How to Manage Global Discount in Odoo 17 POS
 
1029-Danh muc Sach Giao Khoa khoi 6.pdf
1029-Danh muc Sach Giao Khoa khoi  6.pdf1029-Danh muc Sach Giao Khoa khoi  6.pdf
1029-Danh muc Sach Giao Khoa khoi 6.pdf
 
Food safety_Challenges food safety laboratories_.pdf
Food safety_Challenges food safety laboratories_.pdfFood safety_Challenges food safety laboratories_.pdf
Food safety_Challenges food safety laboratories_.pdf
 
Spatium Project Simulation student brief
Spatium Project Simulation student briefSpatium Project Simulation student brief
Spatium Project Simulation student brief
 
How to Create and Manage Wizard in Odoo 17
How to Create and Manage Wizard in Odoo 17How to Create and Manage Wizard in Odoo 17
How to Create and Manage Wizard in Odoo 17
 
1029 - Danh muc Sach Giao Khoa 10 . pdf
1029 -  Danh muc Sach Giao Khoa 10 . pdf1029 -  Danh muc Sach Giao Khoa 10 . pdf
1029 - Danh muc Sach Giao Khoa 10 . pdf
 
SKILL OF INTRODUCING THE LESSON MICRO SKILLS.pptx
SKILL OF INTRODUCING THE LESSON MICRO SKILLS.pptxSKILL OF INTRODUCING THE LESSON MICRO SKILLS.pptx
SKILL OF INTRODUCING THE LESSON MICRO SKILLS.pptx
 
Application orientated numerical on hev.ppt
Application orientated numerical on hev.pptApplication orientated numerical on hev.ppt
Application orientated numerical on hev.ppt
 

Bioluminescence Resonance Energy Transfer (BRET) ARTUCLO 1.pdf

  • 1. NATURE PROTOCOLS | VOL.1 NO.1 | 2006 | 337 PROTOCOL INTRODUCTION What is BRET? BRET is a naturally occurring phenomenon used in the labora- tory for real-time monitoring of protein-protein interactions in live cells, cell extracts or purified preparations1−3. The nonra- diative (dipole-dipole) transfer of energy from donor enzyme to complementary acceptor fluorophore occurs after substrate oxida- tion, and its efficiency is inversely proportional to the sixth power of the distance between donor and acceptor dipoles. This energy only excites the acceptor fluorophore if it is in close proximity to the donor, resulting in light emission at a longer wavelength that can be detected and expressed relative to the donor light emission. By generating cDNA fusion constructs, which are transfected into cells within a suitable expression vector4, fusion proteins consist- ing of proteins of interest linked to donor or acceptor molecules are expressed. Donor to acceptor energy transfer and consequent emission from the acceptor generally indicates separation of less than 10 nm, thus indicating that the proteins of interest are likely to be interacting with each other (directly or as part of a complex)5. Indeed, this high distance dependence means that various combi- nations of fusion proteins with donor or acceptor molecules in dif- ferent positions should be tested to establish the best combination for BRET. The BRET procedure is summarized in Figure 1. Diversity of proteins investigated BRET has been used to study a wide range of protein-protein inter- actions in bacterial6, plant7 and mammalian cells8,9. The first use of BRET studied KaiB circadian clock protein dimerization6. Since then, well over a hundred protein-protein interaction studies have used BRET,with approximately half monitoring interactions involv- ing G protein−coupled receptors (GPCRs), particularly dimeriza- tion or oligomerization, but also interactions with both membrane and cytosolic proteins10. The other half of studies that have used BRET covers a diverse range of interactions too large to mention here, but includes those involving other membrane-bound recep- tors11,12, cytosolic receptors13,14, integrins11,15, enzymes15,16, endo- philins17 and nuclear cofactors18. This diversity illustrates that any protein-protein interaction can potentially be observed using BRET provided that the proteins can be suitably labeled and expressed in a functionally relevant manner1. Suitability of BRET The BRET technique is most suited to investigating ligand-modu- lated (or reagent-modulated) interactions as changes in BRET sig- nal after reagent addition provide good evidence for interaction specificity, provided that the reagent is not cytotoxic and does not inhibit substrate oxidation. Changes in BRET signal may repre- Bioluminescence resonance energy transfer (BRET) for the real-time detection of protein-protein interactions Kevin D G Pfleger, Ruth M Seeber & Karin A Eidne 7TM Laboratory/Laboratory for Molecular Endocrinology, Western Australian Institute for Medical Research (WAIMR) and UWA Centre for Medical Research, University of Western Australia, Nedlands, Perth, Western Australia 6009, Australia. Correspondence should be addressed to K.D.G.P. (kpfleger@waimr.uwa.edu.au). Published online 27 June 2006; doi:10.1038/nprot.2006.52 A substantial range of protein-protein interactions can be readily monitored in real time using bioluminescence resonance energy transfer (BRET). The procedure involves heterologous coexpression of fusion proteins, which link proteins of interest to a bioluminescent donor enzyme or acceptor fluorophore. Energy transfer between these proteins is then detected. This protocol encompasses BRET1, BRET2 and the recently described eBRET, including selection of the donor, acceptor and substrate combination, fusion construct generation and validation, cell culture, fluorescence and luminescence detection, BRET detection and data analysis. The protocol is particularly suited to studying protein-protein interactions in live cells (adherent or in suspension), but cell extracts and purified proteins can also be used. Furthermore, although the procedure is illustrated with references to mammalian cell culture conditions, this protocol can be readily used for bacterial or plant studies. Once fusion proteins are generated and validated, the procedure typically takes 48–72 h depending on cell culture requirements. Figure 1 | An illustration of the BRET protocol using a microplate luminometer. Reprinted with permission from ref. 1. © 2006 Nature Publishing Group http://www.nature.com/natureprotocols
  • 2. 338 | VOL.1 NO.1 | 2006 | NATURE PROTOCOLS PROTOCOL sent an increase or decrease in the number and/or rate of inter- actions between the proteins of interest, but they may also result from conformational changes that influence the relative position- ing of the donor and acceptor molecules. This needs to be taken into account when interpreting the data. Studies of constitutive interactions using BRET must provide evidence for interaction specificity1, typically using saturation or competition assays as dis- cussed below. In particular, suitable negative controls are required, such as similar proteins expressed at similar levels in the same cel- lular compartment. Positive controls should also be included when possible. The donor covalently fused to the acceptor is a useful construct for checking that the protocol is working and to com- pare between assays. A better positive control is to use a combi- nation of fusion proteins, which are known to interact, and are expressed at similar levels and in the same cellular compartment as the proteins under investigation. The BRET technology is most suited to studying protein-protein interactions in live cells where they are expressed in a near-physiological environment in the cor- rect cellular compartment. Cell extracts or purified proteins can be used to increase protein expression levels, but these situations are less likely to provide physiologically relevant data and are par- ticularly prone to the problems associated with overexpression and artifactual interactions. The BRET technique is not suitable for investigating endogenous proteins because of the need for fusion proteins. Furthermore, unless cells are fractionated19, BRET can- not currently provide information about subcellular location. Saturation assays The use of saturation assays to provide evidence for the specificity of constitutive interactions has been put forward by Bouvier and coworkers20. Using the procedure described in this protocol, sev- eral samples that coexpress a constant amount of donor-labeled protein with increasing amounts of acceptor-labeled protein are assayed. Ideally, the lowest level of donor-labeled protein expres- sion that results in a reliable luminescence signal should be used. It is thought that specific interactions result in BRET signal satu- ration at high concentrations of acceptor-labeled protein. By con- trast,nonspecific interactions are believed to result in BRET signals that increase with increasing concentrations of acceptor-labeled protein in a quasi-linear manner20. Relative acceptor fluorescence and donor luminescence levels should be established in addition to the BRET signals. Consequently, saturation curves can be gener- ated using nonlinear regression, plotting BRET signal against the ratio of acceptor fluorescence over donor luminescence. Competition assays Evidence for specificity can also be provided by showing com- petitive inhibition of interactions. Using the procedure described in this protocol, donor- and acceptor-labeled proteins are coex- pressed with unlabeled protein that competes for interactions with the labeled proteins. Unlabeled protein can be expressed at a single concentration (usually excess)9 or at increasing con- centrations21. In parallel, a negative control sample coexpressing TABLE 1 | Comparison of BRET1, eBRET and BRET2. BRET1 eBRET BRET2 Donor Renilla luciferase Renilla luciferase Renilla luciferase Acceptor YFP (or EGFP) YFP (or EGFP) GFP2 or GFP10 Substrate Coelenterazine h EnduRen DeepBlueC Example filters ‘Short-wavelength emission’ (nm): 440−500 (YFP) 400−475 (EGFP) 440−500 (YFP) 400−475 (EGFP) 370−450 ‘Long-wavelength emission’ (nm): 510−590 (YFP) 500−550 (EGFP) 510−590 (YFP) 500−550 (EGFP) 500−530 Characteristics Donor and acceptor emission peak separation (nm) ∼55 (YFP) ∼35 (EGFP) ∼55 (YFP) ∼35 (EGFP) ∼115 Substrate relative quantum yield >100 >100 1 Possible detection duration Up to 1 h Several hours Few seconds Substrate injection required No No Yes Substrate preincubation 0 1.5 h 0 Possible in live cells Yes Yes Yes Possible in cell extracts or purified proteins Yes No Yes Optimal final substrate concentration (µM) 5 60 5 Typical assay buffer D-PBS with CaCl2, MgCl2 and D- glucose HEPES-buffered phenol red−free medium D-PBS with CaCl2, MgCl2 and D- glucose YFP, yellow fluorescent protein (includes Topaz, Venus and Citrine); EGFP, enhanced green fluorescent protein. See ref. 1 for a detailed discussion. © 2006 Nature Publishing Group http://www.nature.com/natureprotocols
  • 3. NATURE PROTOCOLS | VOL.1 NO.1 | 2006 | 339 PROTOCOL donor- and acceptor-labeled proteins with unlabeled protein that does not compete for interactions with the labeled proteins is includ- ed. It is important to measure relative protein expression levels (as determined by fluorescence or luminescence detection) to ensure that an attenuated BRET signal is not a consequence of reducing labeled-protein expression as the unlabeled protein is introduced. The different BRET derivations Three BRET derivations, BRET1, BRET2 and eBRET, are now avail- able for general laboratory use and their characteristics are summa- rized in Table 1. The advantage of BRET2 over BRET1 and eBRET is the superior donor and acceptor emission peak separation when using the substrate DeepBlueC. This enables donor and acceptor emissions to be distinguished much more easily, thereby reducing the background22. But the low quantum yield and rapid decay of DeepBlueC mean that more cells are needed to achieve sufficiently high luminescence levels for BRET detection, and highly sensi- tive instrumentation is required22. Furthermore, BRET2 assays are effectively impractical without injection of substrate into samples. The advantage of eBRET over both BRET1 and BRET2 is the capacity to monitor a particular interaction in real time over several hours or measure multiple samples on the scale of high- throughput screening without the need for frequent substrate addition23. The limitation with both scenarios is cell viability, but by assaying cells in HEPES-buffered medium, cell viability does not appear to be compromised substantially for at least 6 h at 37 °C (ref. 23). eBRET should be carried out using live cells at 37 °C. It cannot be used for assaying cell extracts or purified proteins because of its dependence on endogenous esterases. BRET imaging The use of an intensified charge-coupled device (CCD) camera to image BRET is an exciting technological derivation. BRET from single cells can be visualized21 and the potential for detecting BRET in whole animals has recently been demonstrated by imaging trans- fected cells injected intravenously or subcutaneously into mice24. Furthermore, the development of higher-quantum-yield donors may well allow subcellular BRET detection in the near future. The specialized instrumentation required for BRET imaging clearly necessitates a considerably different protocol to that needed for BRET detected by luminometry or spectroscopy. Consequently, this derivation is beyond the scope of the protocol described herein. Fluorescence and luminescence detection It is desirable to detect relative fluorescence and luminescence levels for evaluating relative expression levels, and this is par- ticularly important when carrying out saturation and competi- tion assays20. Fluorescence can be detected using a fluorometer, scanning spectrometer or flow cytometer. Using a fluorometer is the most rapid and straightforward method, with scanning spectrometry providing additional spectral information, should that be required. Both of these methods allow subsequent lumi- nescence measurements to be made on the same aliquot of cells, which enables relative fluorescence over luminescence ratios to be established for the same sample. Flow cytometry addition- ally allows evaluation of the percentage of cells expressing fluo- rophore and the mean fluorescence. Subsequent measurement of luminescence on the same aliquot of cells, however, is less practical. BRET detection BRET can be detected using a luminometer (microplate or sin- gle-tube) or scanning spectrometer. The luminometer must have the capacity to sequentially or simultaneously detect filtered light within two distinct wavelength windows and ideally include tem- perature control and kinetics software (required for eBRET). Injectors are required for BRET2 and are desirable for BRET1 and eBRET. Single-tube luminometers are more sensitive than micro- plate luminometers, but they are clearly less practical for assay- ing multiple samples. Scanning spectroscopy provides additional spectral information, but the time required to complete each scan limits the number of repeats per sample, the number of samples that can be measured and the ability to generate time courses. The rapid decay of DeepBlueC limits the use of scanning spectroscopy for BRET2. Donor-only controls When investigating constitutive interactions, cells expressing only the donor-labeled proteins should be included to establish the background signal. There is a second option for analyzing BRET data if investigating a ligand-induced interaction23. This does not require ‘donor-only’ samples and is theoretically more accurate as aliquots of the same preparation containing the same protein expression are used to assess the ‘interacting protein’ and ‘back- ground’ emissions. MATERIALS REAGENTS • cDNA for functionally validated fusion proteins in suitable expression vectors • Suitable cells for transfection (e.g.,HEK293 and COS-7) • 6-well cell culture plates (BD Falcon,cat.no.351146) • Suitable growth medium,such as Complete Medium (Dulbecco’s modified Eagle’s medium (DMEM; Gibco,cat.no.11960-044) containing 0.3 mg ml−1 glutamine (Gibco,cat.no.25030-081),100 IU ml−1 penicillin,100 µg ml−1 streptomycin (Gibco,cat.no.15140-122) and 10% fetal calf serum (Gibco,cat. no.16000-044)) for HEK293 and COS-7 cells • Transfection system or reagent; examples of reagents include Genejuice (Novagen,cat.no.70967) and Polyfect (Qiagen,cat.no.301 107) ! CAUTION Wear suitable protective clothing,gloves and safety glasses. • 96-well white cell culture plate (Nunc,cat.no.136101) for assaying adherent cells • Phenol red−free medium,such as the Complete Medium described above,but containing DMEM without phenol red (Gibco,cat.no.31053-028); requires 25 mM HEPES (Sigma,cat.no.H-3034) when used as eBRET assay buffer • 0.05% trypsin−0.53 mM EDTA (Gibco,cat.no.15400-054) ! CAUTION Wear suitable protective clothing,gloves and safety glasses. • Dulbecco’s phosphate-buffered saline (D-PBS) containing 0.1 g l−1 CaCl2, 0.1 g l−1 MgCl2·6H2O and 1 g l−1 D-glucose (Invitrogen,cat.no.14287-080) • 96-well isoplates: white for BRET (Perkin Elmer,cat.no.1450-581) and black for fluorescence detection (OptiPlate-96F; Perkin Elmer,cat.no.6005270) • For BRET1 only: 500 µM coelenterazine h (Molecular Probes,cat.no.C-6780) in methanol as luciferase substrate stock solution (Sigma,cat.no.10158.6B) ! CAUTION Wear suitable protective clothing,gloves and safety glasses. ▲ CRITICAL Store at -20 °C protected from light. • For BRET2 only: 1 mM DeepBlueC (PerkinElmer,cat.no.6310-100C) in © 2006 Nature Publishing Group http://www.nature.com/natureprotocols
  • 4. 340 | VOL.1 NO.1 | 2006 | NATURE PROTOCOLS PROTOCOL anhydrous or absolute ethanol as luciferase substrate stock solution (Merck,cat. no.1.00986) ! CAUTION Wear suitable protective clothing,gloves and safety glasses.▲ CRITICAL Store at –20 °C protected from light. •For eBRET only: 60 mM EnduRen (Promega,cat.no.E6482) in dimethylsulfoxide (DMSO) as luciferase substrate stock solution (Sigma, cat.no.D-2650) ! CAUTION Readily absorbed through skin.Wear suitable protective clothing,gloves and safety glasses.▲ CRITICAL Store at –20 °C protected from light. •Ligand or other modulating reagent depending on interaction being assayed ! CAUTION Wear suitable protective clothing,gloves and safety glasses. EQUIPMENT •Standard cell culture facility including Class II biological safety cabinet,such as BH2000 series (Clyde-Apac),and 37 °C incubator with 5% CO2,such as the Heracell (Heraeus) •Microplate luminometer; examples include theVICTOR Light (PerkinElmer, cat.no.1420-060),Mithras LB 940 (Berthold Technologies) and FLUOstar Optima or POLARstar Optima (BMG Labtech) •(Optional) Single-tube luminometer with filter switch capability such as the Sirius C (Berthold Detection Systems,cat.no.11040050) •(Optional) Scanning spectrometer with 96-well plate capability; examples include the Spex fluorolog or fluoromax (JobinYvon),the Cary Eclipse (Varian) and the FlexStation II (Molecular Devices) •(Optional) Fluorometer; examples include the EnVision (Perkin Elmer,cat.no. 2102-0010),Mithras LB 940 (Berthold Technologies) and FLUOstar Optima or POLARstar Optima (BMG Labtech) •(Optional) Flow cytometer,such as the FACS Calibur (Becton Dickinson,cat. no.1641) PROCEDURE Preparation 1| Select the donor, acceptor and substrate combination, and consequently the filter combination if using a luminometer for BRET detection1 (Table 1). 2| Generate fusion constructs in a suitable expression vector consisting of the cDNA for the protein of interest inserted in- frame with the cDNA for the donor or acceptor molecule, incorporating the cDNA for a suitable linker region, if appropriate. Remove the stop codon separating the cDNA sequences by mutagenesis so that a single fusion protein is expressed after transfection. 3| Validate the labeling of proteins of interest, including suitable control proteins, by comparing labeled and wild-type proteins in functional assays. Check that luminescence or fluorescence is detectable. If possible, use confocal microscopy to visualize correct cellular localization of acceptor-labeled protein. ▲ CRITICAL STEP Establish that protein function is not compromised by the addition of donor or acceptor molecules. Insufficient demonstration of correct cellular function must be taken into account when interpreting data. ? TROUBLESHOOTING 4| Reconstitute and store the luciferase substrate stock solution containing coelenterazine h for BRET1 or DeepBlueC for BRET2 (both option A), or EnduRen for eBRET (option B). (A) For BRET1 or BRET2: (i) Allow the vial of lyophilized coelenterazine h or DeepBlueC to equilibrate to ambient temperature before opening to avoid condensation. ▲ CRITICAL STEP Protect from light. (ii) Reconstitute coelenterazine h with methanol, and DeepBlueC with anhydrous or absolute ethanol. Coelenterazine h can also be reconstituted in ethanol. ▲ CRITICAL STEP Do not dissolve in dimethylsulfoxide (DMSO). (iii) Agitate gently until resuspended, potentially a few minutes. (iv) Aliquot (volumes depend on expected rate of use) and store desiccated and protected from light at –20 °C. (B) For eBRET: (i) Allow the vial of lyophilized EnduRen to equilibrate to ambient temperature before opening to avoid condensation. ▲ CRITICAL STEP Protect from light. (ii) Reconstitute in tissue-culture grade dimethylsulfoxide (DMSO). (iii) Vortex to resuspend. ▲ CRITICAL STEP Repeated vortexing may be required. Resuspension may take up to 10 min and may require warming to 37 °C. (iv) Aliquot and store protected from light at –20 °C. Cell culture 5| Aliquot cells into a 6-well cell culture plate in a suitable growth medium. The number of cells required in each well will depend on size, growth rate and transfection system to be used, but typically they should be 50–80% confluent after 24 h if being transiently transfected. For example, COS-7 cells are typically plated out at a density of 120,000 cells per well. Cells stably expressing fusion proteins can also be used, with clonal stable cell lines having the advantage of homogeneous protein expression levels. Maintain at 37 °C, 5% CO2 (for mammalian cells). © 2006 Nature Publishing Group http://www.nature.com/natureprotocols
  • 5. NATURE PROTOCOLS | VOL.1 NO.1 | 2006 | 341 PROTOCOL 6| Typically 24 h after plating, coexpress the donor- and acceptor-labeled proteins of interest in cells using a suitable transfection or expression system if these proteins are not already stably expressed. If required, produce a population of cells expressing only the donor-labeled proteins at similar expression levels to those in the samples coexpressing donor- and acceptor-labeled proteins. This requirement depends on the form of analysis to be used. Aim to achieve a physiologically relevant expression level that still provides a detectable BRET signal. Samples with various combinations of protein expression levels may be required if saturation or competition assays are being carried out. Express suitable positive- and negative-control proteins in parallel. Maintain a population of untransfected cells in parallel for establishing background levels of fluorescence and luminescence. ? TROUBLESHOOTING 7| If pretreating with ligand or other reagent, add these at an appropriate time between transfection and BRET detection. Include vehicle-treated samples in parallel. 8| Interactions can be monitored in adherent cells (option A), in cell suspension (option B), in cell extracts or between purified proteins (option C). The optimal expression time for transiently transfected fusion proteins should be established as it varies from 24 to 72 h. (A) In adherent cells: (i) Detach the cells (e.g., using trypsin-EDTA) typically 24 h after transfection. (ii) Resuspend the cells in phenol red−free medium (HEPES-buffered if eBRET is to be carried out in this medium) and aliquot into a 96-well white tissue culture plate (40−100 µl per well). Initially titrate cells to establish a suitable dilution. Higher cell concentrations are required for BRET2 compared with BRET1 or eBRET. (iii) Incubate at 37 °C, 5% CO2 (mammalian cells) for an additional 24 h to allow attachment, and either assay in the existing phenol red−free medium or remove medium and replace with assay buffer. ▲ CRITICAL STEP Extreme care must be taken not to detach cells. If removing the medium, washing is not recommended. ? TROUBLESHOOTING (B) In cell suspension: (i) Detach the cells (e.g., using trypsin-EDTA) immediately before the assay (24–48 h after transfection) and resuspend in assay buffer. (ii) Aliquot cells into a 96-well white isoplate (40–100 µl per well). (C) Cell extracts or purified proteins: (i) Typically prepare cell extracts or purified proteins 24−48 h after transfection. (ii) Aliquot into a 96-well white isoplate (40–100 µl per well). Fluorescence and luminescence detection 9| Fluorescence can be measured in a fluorometer or scanning spectrometer (both option A), or flow cytometer (option B). Use a fluorometer or scanning spectrometer with 96-well capability if subsequent detection of unfiltered luminescence is to be carried out using the same aliquots (Step 10). (A) Fluorometer or scanning spectrometer: (i) Measure the relative fluorescence from aliquots of each sample in D-PBS (40−100 µl per well in a 96-well black isoplate), directly exciting the acceptor fluorophore with laser light of a suitable wavelength. (ii) Filter the emission to avoid detection of excitation light and correct for background fluorescence from untransfected cells. ? TROUBLESHOOTING (B) Flow cytometer: (i) Dilute aliquots of samples in D-PBS to achieve an appropriate volume. (ii) Measure the relative fluorescence, directly exciting the acceptor fluorophore with laser light of a suitable wavelength. (iii) Filter the emission to avoid detection of excitation light and correct for background fluorescence from untransfected cells. ? TROUBLESHOOTING 10| Taking the same aliquots as assessed in the fluorometer or scanning spectrometer, add coelenterazine h or DeepBlueC and measure unfiltered luminescence in a luminometer such as that used for BRET detection. Correct for background luminescence from untransfected cells unless this is shown to be negligible. ? TROUBLESHOOTING © 2006 Nature Publishing Group http://www.nature.com/natureprotocols
  • 6. 342 | VOL.1 NO.1 | 2006 | NATURE PROTOCOLS PROTOCOL BRET detection 11| Dilute the substrate stock (coelenterazine h, DeepBlueC or EnduRen) to a working concentration in assay buffer, typically 10× final concentration (Table 1). ▲ CRITICAL STEP Dilute immediately before adding to samples and protect from light. For EnduRen, preincubate the assay buffer at 37 °C to avoid precipitation. 12| Add the luciferase substrate to the samples. For eBRET, use option A; for BRET1 use option B; and for BRET2 use option C. (A) For eBRET, add EnduRen to live cells at least 1.5 h before BRET detection1. Substrate addition can be verified by luminescence detection. Counts are low at this pre-equilibrium stage, but they are substantially higher than background. For mammalian cells, incubate at 37 °C, 5% CO2. (B) For BRET1, add coelenterazine h to the samples immediately before BRET detection. As BRET1 data are generally reliable for 30–60 min after coelenterazine h addition (depending on the initial signal), injection of the substrate is optional. (C) For BRET2, add DeepBlueC to each well immediately before detecting emission from that well. ▲ CRITICAL STEP In practical terms, this requires the use of injectors. 13| Detect the light emissions using either a luminometer (option A) or scanning spectrometer (option B). The rapid decay of DeepBlueC limits the use of scanning spectroscopy for BRET2. ▲ CRITICAL STEP eBRET should be carried out at 37 °C to avoid EnduRen precipitation. (A) If using a luminometer with the appropriate filter combination (Table 1), detect the light emission through both filters (simultaneously or sequentially, typically 0.5–5 s per filter) before proceeding to the next sample. Simultaneous detection reduces measurement times and is potentially more accurate, particularly for BRET2 because of the rapid decay of DeepBlueC. ? TROUBLESHOOTING (B) If using scanning spectrometry for BRET1 or eBRET, detect light emission from 400–600 nm, typically using a 10 nm slit width and 2 s per increment. ? TROUBLESHOOTING 14| Repeat readings as required, within the timeframe allowed by substrate stability (Table 1) and cell viability. BRET2 repeats require re-addition of DeepBlueC. Repeats can be automated with appropriate kinetics software. 15| Add ligands or other reagents, if desired, and continue collecting readings. Use a luminometer and inject these agents if early post-addition time points are required (<1 min). Include vehicle-treated samples in parallel. ● TIMING Preparation (Steps 1–4): potentially several weeks depending on the complexity of cloning and validation required. Cell culture (Steps 5–8): typically 48–72 h (less if stable cell lines, cell extracts or purified proteins have already been prepared). Fluorescence and luminescence detection (steps 9–10): a few minutes (depending on the number of samples). BRET detection (Steps 11–15): minutes to hours depending on the number of samples, number of repeats and length of time course. eBRET requires addition of EnduRen at least 1.5 h before commencing BRET detection. ? TROUBLESHOOTING For troubleshooting guidance see Table 2. TABLE 2 | Troubleshooting table. STEP PROBLEM POSSIBLE REASON SOLUTION 3 Fusion protein is not functioning correctly There was an error in construct generation Sequence and check entire fusion protein cDNA Label is interfering with protein function Reposition the label to the other end of the protein (or possibly an internal peripheral loop or linker region), or increase linker length between protein of interest and label 6 Cells are dying after transfection Cytotoxicity of transfection reagent Refer to the manufacturer’s guidelines, or consider using alternative reagents Cytotoxicity of GFP fusion proteins Titrate down expression of GFP fusion proteins © 2006 Nature Publishing Group http://www.nature.com/natureprotocols
  • 7. NATURE PROTOCOLS | VOL.1 NO.1 | 2006 | 343 PROTOCOL ANTICIPATED RESULTS Calculate the BRET signals using option A (below) if a luminometer was used or option B if a scanning spectrometer was used. If autoluminescence from untransfected cells is not negligible, this should be subtracted from emissions before calculating the BRET signal. (A) If a luminometer was used, calculate the BRET signal using the following equation: For BRET1 and eBRET, note that light is emitted through the ‘long-wavelength emission’ filter, even in the absence of acceptor expression or ligand addition. This is ‘bleed-through’ from the donor emission1. There are two options for using the equation. All interactions can be analyzed using option (i), but ligand-induced interactions can also be analyzed using option (ii). Option (ii) eliminates the need for ‘donor-only’ samples and is theoretically more accurate as aliquots of the same preparation containing the same amount of protein are used to assess the ‘interacting protein’ and ‘background emissions’23. (i) The ‘emissions from interacting proteins’ are from samples coexpressing donor and acceptor fusion proteins and the ‘background emissions’ from samples expressing only donor fusion proteins. (ii) If the interaction is ligand-induced, the ‘emissions from interacting proteins’ can be from ligand-treated samples and the ‘background emissions’ from vehicle-treated samples. (B) Data derived using scanning spectroscopy can be analyzed using option (i) or (ii): (i) Normalize the emission spectra by defining the donor peak emission intensity as 1. Calculate the BRET signal as the area under the curve to which the acceptor emission contributes (typically 500−550 nm) minus the corresponding area observed when only the donor is present. (ii) Alternatively, establish areas under the curve within wavelength windows that correspond to the luminometer filters and calculate the BRET ratio as described for luminometer-derived data (option A). TABLE 2 | Troubleshooting table (continued). STEP PROBLEM POSSIBLE REASON SOLUTION 8 Cells are detaching despite extreme care. Cells are not sufficiently adherent Consider using different plastics or coating wells (e.g., with poly(L-lysine)) 9, 10 and 13 There are low relative luminescence or fluorescence counts There is poor protein expression Optimize or consider an alternative transfection strategy The cell number is low Increase cell concentratio Substrate was not added Add substrate to each well Substrate is not viable Check expiry and storage conditions. Consider using new aliquot of substrate Reducing agent present Ensure buffers do not include reducing agents such as ascorbic acid Instrumentation is not functioning correctly Check instrument setup and consider recalibration 10 and 13 Luminescence signal is detected from untransfected cells upon substrate addition There is spontaneous autoluminescence Correct for signal from untransfected cells Consider removing serum or BSA if present in assay buffer Problem should be reduced or alleviated by using EnduRen Results BRET signal is not detected despite validated fusion proteins generating high luminescence or fluorescence counts Distance between donor and acceptor is too great Test alternative combinations of fusion proteins with labels in different positions Relative orientation of donor and acceptor is unsuitable for BRE Increase linker length between protein of interest and label May be due to ligand if interaction is ligand-induce Check and optimize potency of ligand required BRET signal (or BRET radio) = ‘long-wavelength emission from interacting proteins’ ‘short-wavelength emission from interacting proteins’ ‘background long-wavelength emission’ ‘background short-wavelength emission’ – © 2006 Nature Publishing Group http://www.nature.com/natureprotocols
  • 8. 344 | VOL.1 NO.1 | 2006 | NATURE PROTOCOLS PROTOCOL An example of raw BRET data is shown in Table 3 and is derived from data generated in three independent experiments. Two aliquots of the same cell sample were treated with ligand or vehicle and monitored in real-time. The data shown was observed 20 min after ligand or vehicle addition. In this example, option A(ii) has been used for data analysis. Emissions were detected through the two filters and the ‘long-wavelength emission’ over ‘short-wavelength emission’ ratios calculated. The BRET ratio was then calculated as: Ligand-treated emission ratio - vehicle-treated emission ratio = 2.3429 - 2.1396 = 0.203. Statistical analysis of BRET data should be carried out using ANOVA with suitable post-tests, or using Student’s t-tests where appropriate. One method is to determine the ‘long-wavelength emission’ over ‘short-wavelength emission’ ratio for the interacting proteins and background separately. The variance in these ratios can then be compared to establish whether they are significantly different. For the example shown in Table 3, the ligand-treated and vehicle-treated emission ratios were found to be significantly different (P = 0.0086) using an unpaired Student’s t-test. Anticipated results for ligand- (or reagent-) modulated interactions are illustrated in Figure 2. eBRET is particularly suitable if kinetic profiles beyond 30−60 min are required. Anticipated results for constitutive interactions should not only exhibit significantly higher BRET signals than appropriate negative controls, but also demonstrate interaction specificity by incorporating competition or saturation data generation into the experimental design (see ref. 1 for extensive discussion). It is important to note that a negative BRET result is not conclusive evidence for a lack of protein-protein interaction. The extremely high dependency on distance between donor and acceptor means that functionally validated fusion proteins may interact without a BRET signal being detectable. See Table 2 for troubleshooting guidance. TABLE 3 | An example of raw BRET data and its analysis. Experiment Ligand-treated Vehicle-treated Long-wavelength emission Short-wavelength emission Long:short wavelength ratio Long-wavelength emission Short-wavelength emission Long:short wavelength ratio 1 313,159 134,012 2.3368 404,242 191,367 2.1124 2 154,900 65,013 2.3826 130,677 59,082 2.2118 3 256,707 111,166 2.3092 353,669 168,849 2.0946 Mean 2.3429 2.1396 s.d. 0.0371 0.0632 a b Figure 2 | Examples of theoretical BRET data from ligand (reagent) modulated interactions. (a) Dose-response curves can be generated by nonlinear regression, permitting calculation of EC50 (half-maximal effective concentration) values. (b) Changes in BRET ratio can be monitored over time before and after addition of ligand and/or other modulators (the dashed line denotes data produced upon addition of vehicle instead of ligand). Apparent association (or dissociation) rate constants can be calculated from such data. COMPETING INTERESTS STATEMENT The authors declare competing financial interests (see the HTML version of this article for details). Published online at http://www.natureprotocols.com/ Reprints and permissions information is available online at http://npg.nature. com/reprintsandpermissions/ 1. Pfleger, K.D. & Eidne, K.A. Illuminating insights into protein-protein interactions using bioluminescence resonance energy transfer (BRET). Nat. Methods 3, 165−174 (2006). 2. Milligan, G. & Bouvier, M. Methods to monitor the quaternary structure of G-protein−coupled receptors. FEBS J. 272, 2914−2925 (2005). 3. Boute, N., Jockers, R. & Issad, T. The use of resonance energy transfer in high-throughput screening: BRET versus FRET. Trends Pharmacol. Sci. 23, 351−354 (2002). 4. Pfleger, K.D.G. & Eidne, K.A. New technologies: bioluminescence resonance energy transfer (BRET) for the detection of real time interactions involving G- protein coupled receptors. Pituitary 6, 141−151 (2003). 5. Wu, P. & Brand, L. Resonance energy transfer: methods and applications. Anal. Biochem. 218, 1−13 (1994). 6. Xu, Y., Piston, D.W. & Johnson, C.H. A bioluminescence resonance energy transfer (BRET) system: application to interacting circadian clock proteins. Proc. Natl. Acad. Sci. USA 96, 151−156. (1999). 7. Subramanian, C. et al. The Arabidopsis repressor of light signaling, COP1, is regulated by nuclear exclusion: mutational analysis by bioluminescence resonance energy transfer. Proc. Natl. Acad. Sci. USA 101, 6798−6802 (2004). 8. Angers, S. et al. Detection of beta 2-adrenergic receptor dimerization in living cells using bioluminescence resonance energy transfer (BRET). Proc. Natl. Acad. Sci. USA 97, 3684−3689 (2000). 9. Kroeger, K.M., Hanyaloglu, A.C., Seeber, R.M., Miles, L.E. & Eidne, K.A. Constitutive and agonist-dependent homo-oligomerization of the thyrotropin-releasing hormone receptor. Detection in living cells using bioluminescence resonance energy transfer. J. Biol. Chem. 276, 12736−12743 (2001). 10. Pfleger, K.D.G. & Eidne, K.A. Monitoring the formation of dynamic G-protein-coupled receptor-protein complexes in living cells. Biochem. J. © 2006 Nature Publishing Group http://www.nature.com/natureprotocols
  • 9. NATURE PROTOCOLS | VOL.1 NO.1 | 2006 | 345 PROTOCOL 385, 625−637 (2005). 11. Scaffidi, A.K. et al. α(v)β(3) Integrin interacts with the transforming growth factor β (TGFβ) type II receptor to potentiate the proliferative effects of TGFβ1 in living human lung fibroblasts. J. Biol. Chem. 279, 37726−37733 (2004). 12. Brown, R.J. et al. Model for growth hormone receptor activation based on subunit rotation within a receptor dimer. Nat. Struct. Mol. Biol. 12, 814−821 (2005). 13. Michelini, E., Mirasoli, M., Karp, M., Virta, M. & Roda, A. Development of a bioluminescence resonance energy-transfer assay for estrogen-like compound in vivo monitoring. Anal. Chem. 76, 7069−7076 (2004). 14. Garside, H. et al. Glucocorticoid ligands specify different interactions with NF-kappaB by allosteric effects on the glucocorticoid receptor DNA binding domain. J. Biol. Chem. 279, 50050−50059 (2004). 15. de Virgilio, M., Kiosses, W.B. & Shattil, S.J. Proximal, selective, and dynamic interactions between integrin alphaIIbbeta3 and protein tyrosine kinases in living cells. J. Cell. Biol. 165, 305−311 (2004). 16. Yung, T.M., Sato, S. & Satoh, M.S. Poly(ADP-ribosyl)ation as a DNA damage- induced post-translational modification regulating poly(ADP-ribose) polymerase-1-topoisomerase I interaction. J. Biol. Chem. 279, 39686−39696 (2004). 17. Trevaskis, J. et al. Src homology 3-domain growth factor receptor-bound 2-like (endophilin) interacting protein 1, a novel neuronal protein that regulates energy balance. Endocrinology 146, 3757−3764 (2005). 18. Germain-Desprez, D., Bazinet, M., Bouvier, M. & Aubry, M. Oligomerization of transcriptional intermediary factor 1 regulators and interaction with ZNF74 nuclear matrix protein revealed by bioluminescence resonance energy transfer in living cells. J. Biol. Chem. 278, 22367−22373 (2003). 19. Terrillon, S. et al. Oxytocin and vasopressin V1a and V2 receptors from constitutive homo- and heterodimers during biosynthesis. Mol. Endocrinol. 17, 677−691 (2003). 20. Mercier, J.F., Salahpour, A., Angers, S., Breit, A. & Bouvier, M. Quantitative assessment of β1- and β2-adrenergic receptor homo- and heterodimerization by bioluminescence resonance energy transfer. J. Biol. Chem. 277, 44925−44931 (2002). 21. Ayoub, M.A. et al. Monitoring of ligand-independent dimerization and ligand-induced conformational changes of melatonin receptors in living cells by bioluminescence resonance energy transfer. J. Biol. Chem. 277, 21522−21528 (2002). 22. Hamdan, F.F., Audet, M., Garneau, P., Pelletier, J. & Bouvier, M. High- throughput screening of G protein-coupled receptor antagonists using a bioluminescence resonance energy transfer 1−based β-arrestin2 recruitment assay. J. Biomol. Screen. 10, 463−475 (2005). 23. Pfleger, K.D. et al. Extended bioluminescence resonance energy transfer (eBRET) for monitoring prolonged protein-protein interactions in live cells. Cell. Signal; Advance online publication 21 February 2006 (doi: 10.1016/ j.cellsig.2006.01.004). 24. De, A. & Gambhir, S.S. Noninvasive imaging of protein-protein interactions from live cells and living subjects using bioluminescence resonance energy transfer. FASEB J. 19, 2017−2019 (2005). © 2006 Nature Publishing Group http://www.nature.com/natureprotocols