An Introduction to Fluorescence Resonance Energy Transfer (FRET) Technology and its Application in Bioscience

Similar documents
Chapter 4 Fluorescence Resonance Energy Transfer (FRET) by Minor Groove-Associated Cyanine-Polyamide Conjugates

The Green Fluorescent Protein. w.chem.uwec.edu/chem412_s99/ppt/green.ppt

Challenges to measuring intracellular Ca 2+ Calmodulin: nature s Ca 2+ sensor

Using Quantum Dots in Fluorescence Resonance Energy Transfer Studies

Supplementary Figure 1. The normalized absorption and emission spectra of 605QD

Fluorescence quenching, Fluorescence anisotropy, Fluorescence resonance energy transfer (FRET)

F* techniques: FRAP, FLIP, FRET, FLIM,

Contact Details. Dr Alexander Galkin. Office: MBC Room 186. Tel: (028) Frequency and wavelength.

FRET and FRET based Microscopy Techniques

FRET from Multiple Pathways in Fluorophore Labeled DNA

Quantitation of ssdna using OliGreen Fluorescent Stain

UV Fluorescence Polarization as a Means to Investigate Protein Conformational and Mass Change

Imaging of BacMam Transfected U-2 OS Cells

Concept review: Fluorescence

Roche Molecular Biochemicals Technical Note No. LC 6/99

If you like us, please share us on social media. The latest UCD Hyperlibrary newsletter is now complete, check it out.

Synergy NEO HTS Multi-Mode Microplate Reader and Life Technologies Reagents

Reminder: absorption. OD = A = - log (I / I 0 ) = ε (λ) c x. I = I ε(λ) c x. Definitions. Fluorescence quenching and FRET.

Biochemistry. Biochemical Techniques. 18 Spectrofluorimetry

Comparison of LANCE Ultra TR-FRET to PerkinElmer s Classical LANCE TR-FRET Platform for Kinase Applications

Lab 1: Ensemble Fluorescence Basics

Design for Manufacturability (DFM) in the Life Sciences

Macromolecular environments influence proteins

Application of Quantum Mechanics to Biology

Fluorescence Spectroscopy. Student: Marin Cristina Antonia Coordinator:S.l. Preda Liliana

Real-Time PCR Principles and Applications

Welcome! openmicberkeley.wordpress.com. Open Berkeley

Ratio and Spectral Scanning

Validation and Comparison of Single-Step Flash and Dual-Spectral Luciferase Reporter Gene Assays using the Synergy Line of Microplate Readers

Quantitative Real time PCR. Only for teaching purposes - not for reproduction or sale

DELFIA, LANCE and TruPoint technologies Sensitivity and dynamic range using PerkinElmer instrumentation and microplates

Time-resolved Measurements Using the Agilent Cary Eclipse Fluorescence Spectrophotometer A Versatile Instrument for Accurate Measurements

Technical Review. Real time PCR

Stopped-Flow Fluorescence Polarisation/Anisotropy

Methods in virus diagnosis PCR techniques

Chapter 4. the biological community to assay for protein-protein interactions. FRET describes the

Binding of bioactive coomassie brilliant blue G with protein:

Fluorescent protein. Origin of fluorescent proteins. Structural and biophysical analysis of FPs. Application in molecular biology and biotechnology

The most extensively used technique for tissue analysis is light microscopy.

ALP (alkaline phosphatase) calibrators were analyzed manually in microtiter plates to find the linearity range by following this protocol:

ADVANCED PRACTICAL COURSE IN BIOPHYSICS: FRET

Directional Surface Plasmon Coupled Emission

Module1TheBasicsofRealTimePCR Monday, March 19, 2007

FLUORESCENCE. Matyas Molnar and Dirk Pacholsky

Absorption of an electromagnetic wave

Fluorescence Resonance Energy Transfer (FRET) Microscopy

Real Time PCR. Advanced Biotechnology Lab I Florida Atlantic University April 2, 2008

Special Techniques 1. Mark Scott FILM Facility

Introduction to Fluorescence Jablonski Diagram

Utility of Variable Bandwidth Monochromators for Quantification of Fluorescent Probes in Produced Effluent Water

Investigating the distance limit of a metal nanoparticle based spectroscopic ruler

Gene expression. What is gene expression?

Design. Construction. Characterization

Polymerase Chain Reaction: Application and Practical Primer Probe Design qrt-pcr

Development of FRET-based Ion Channel, GPCR, and Kinase Assays on the GENios Pro Multifunctional Reader from Tecan

Monitoring and Optimizing the Lipopolysaccharides-plasmid DNA interaction by FLIM-FRET

More on fluorescence

Optimizing Dual-Glo Luciferase Assays with the Synergy HT Multi-Detection Microplate Reader

20.320, notes for 9/13

Dino-Lite knowledge & education. Fluorescence Microscopes

Masayoshi Honda, Jeehae Park, Robert A. Pugh, Taekjip Ha, and Maria Spies

BoTest A/E Botulinum Neurotoxin Detection Kit- Detailed Description

Executive Summary. clinical supply services

Multiplexed 3D FRET imaging in deep tissue of live embryos Ming Zhao, Xiaoyang Wan, Yu Li, Weibin Zhou and Leilei Peng

Technical tips Session 4

Using Quantum Dots for a FRET Based Immunoassay Technique

The Synergy 2, A Novel Approach to Microplate Multi- Detection for HTS and Drug Discovery

A photoprotection strategy for microsecond-resolution single-molecule fluorescence spectroscopy

Linearly polarized light

Only for teaching purposes - not for reproduction or sale

FLUORESCENT PEPTIDES. Outstanding Performance and Wide Application Range

Partha Roy

Roche Molecular Biochemicals Technical Note No. LC 10/2000

Expressed genes profiling (Microarrays) Overview Of Gene Expression Control Profiling Of Expressed Genes

Supplementary Figure 1. FRET probe labeling locations in the Cas9-RNA-DNA complex.

Lecture Four. Molecular Approaches I: Nucleic Acids

Fluorescence Quenching of Human Serum Albumin by Caffeine

Presented at the 51st Annual ASMS Conference on Mass Spectrometry and Allied Topics Montreal, 2003

Chapter 20 Recombinant DNA Technology. Copyright 2009 Pearson Education, Inc.

Visualizing mechanical tension across membrane receptors with a fluorescent sensor

Contents... vii. List of Figures... xii. List of Tables... xiv. Abbreviatons... xv. Summary... xvii. 1. Introduction In vitro evolution...

Confocal Microscopy Analyzes Cells

Limit of Detection. Theoretical Lowest Concentration

FINAL PROGRESS REPORT. GRANT # DAAD P00001 TITLE: Interactions of Multiple Fluorescent Labels in Biological Sensing

Z -LYTE fluorescent kinase assay technology. Z -LYTE Kinase Assay Platform

Illumatool ΤΜ Tunable Light System: A Non-Destructive Light Source For Molecular And Cellular Biology Applications. John Fox, Lightools Research.

Applications and Uses. (adapted from Roche RealTime PCR Application Manual)

Practice Problems 5. Location of LSA-GFP fluorescence

Automated Imaging and Dual-Mask Analysis of γh2ax Foci to Determine DNA Damage on an Individual Cell Basis

Fluorescense. Aromatic molecules often fluoresce (but DNA bases don't) Excitation ~ absorbance (Differences due to: ν h ' Fluorescence

Color-Rich Fluoro-Max Dyed Microparticles March 2008

Visualizing Cells Molecular Biology of the Cell - Chapter 9

Self-assembly of oligonucleotides

Q-PCR QUANTITATIVE-PCR 세포생물학및실험 2 박태식교수님

1. INTRODUCTION 2. EXPERIMENTAL 3. REFERENCES

Supplementary information. for the paper:

A Cost-effective Workflow for High-Throughput Screening of G- Protein Coupled Receptors (GPCRs)

The Two-Hybrid System

Technical Seminar 22th Jan 2013 DNA Origami

Tide Fluor and Tide Quencher Dyes, Optimized for Maximizing The Power of Fluorescence Resonance Energy Transfer (FRET)

Transcription:

W h i t e P a p e r An Introduction to Fluorescence Resonance Energy Transfer (FRET) Technology and its Application in Bioscience Paul Held, Laboratory Manager, Applications Department, BioTek Instruments, Inc., Winooski, VT Fluorescence Resonance Energy Transfer (FRET) is a physical phenomenon first described over 50 years ago, that is being used more and more in biomedical research and drug discovery today. FRET relies on the distance-dependent transfer of energy from a donor molecule to an acceptor molecule. Due to its sensitivity to distance, FRET has been used to investigate molecular interactions. FRET is the radiationless transmission of energy from a donor molecule to an acceptor molecule. The donor molecule is the dye or chromophore that initially absorbs the energy and the acceptor is the chromophore to which the energy is subsequently transferred. This resonance interaction occurs over greater than interatomic distances, without conversion to thermal energy, and without any molecular collision. The transfer of energy leads to a reduction in the donor s fluorescence intensity and excited state lifetime, and an increase in the acceptor s emission intensity. A pair of molecules that interact in such a manner that FRET occurs is often referred to as a donor/acceptor pair. While there are many factors that influence FRET, the primary conditions that need to be met in order for FRET to occur are relatively few. The donor and acceptor molecules must be in close proximity to one another (typically 10-100 Å). The absorption or excitation spectrum of the acceptor must overlap the fluorescence emission spectrum of the donor (Figure 1). The degree to which they overlap is referred to as the spectral overlap integral (J). The donor and acceptor transition dipole orientations must be approximately parallel. Assuming that the donor acceptor pairs are compatible the most critical element necessary for FRET to occur is close proximity of the pairs. Förster, demonstrated that the efficiency of the process (E) depends on the inverse sixth-distance between donor and acceptor (see Equation 1). [1] Eq 1. E = Ro 6 /(Ro 6 + r 6 ) Where Ro is the Förster distance at which half the energy is transferred and r is the actual distance between donor and acceptor. The distance at which energy transfer is 50% efficient is referred to as the Förster radius (Ro). The magnitude of the Ro is dependent on the spectral properties of the donor and the acceptor. Förster distances ranging from 20 to 90 Å are most useful for studies of biological macromolecules. These distances are comparable to the diameters of many proteins, the thickness of biological membranes, and the distances between sites on multisubunit proteins. Any process that affects the energy transfer rate allows the process to be quantified. As a result, FRET is often referred to as a spectroscopic ruler. Note that the Förster distance (Ro) is dependant on a number of factors, including the fluorescence quantum yield of the donor in the absence of acceptor (fd), the refractive index of the solution (n), the dipole angular orientation of each molecule (k 2 ), and the spectral overlap integral of the donor and acceptor (J). See Equation 2. Eq 2. Ro=9.78 x 10 3 (n -4 * fd * k2 * J)1/6 Å BioTek Instruments, Inc. P.O. Box 998, Highland Park, Winooski, Vermont 05404-0998 USA Phone: 888-451-5171 Outside the USA: 802-655-4740 Email: customercare@biotek.com www.biotek.com Copyright 2012

Figure 1. Schematic representation of the spectral overlap integral As an example of the affect of distance on the efficiency of energy transfer one can use some arbitrary numbers for the Förster distance (Ro) and the actual distance (r). If Ro is arbitrarily set to 1 (Ro =1), and the distance between the donor and acceptor is also equal to 1, then r=ro, and the equation for efficiency is E =1 6 /(1 6 + 1 6 ), which is equal to 0.5 (i.e. 50%). This half maximal value is what the Förster distance is defined as being. If the distance is 10 x closer (e.g. r=0.1ro), then E = 1 6 /(1 6 + 0.1 6 ) = 0.999999, considerably more efficient. However, if the distance between the donor and the acceptor is 10 x further away (i.e. r=10ro), then E = 1 6 /(1 6 + 10 6 ) = 0.0000001. This extreme sensitivity for distance is what allows FRET to be used for proximity experiments. In general the donor and acceptor moieties are different, in which case FRET can be detected by the appearance of fluorescence of the acceptor or by quenching of donor fluorescence. The donor probe is always a fluorescent molecule. Note that luminescent molecules behave as a fluorescent molecule in regards to their emission. With appropriate excitation, its electrons jump from the ground state (So) to a higher vibrational level. Very rapidly (within picoseconds) these electrons decay to the lowest vibrational levels (S1) and eventually decay (within nanoseconds) back to the So state and a photon of light is emitted. When conditions for FRET to occur are met, decay as donor fluorescence and energy transfer to the acceptor will compete for the decay of excitation energy. With resonance energy transfer, the photon is NOT emitted, but rather the energy is transferred to the acceptor molecule, whose electrons in turn become excited as described for the donor molecule. The subsequent return to the ground state emits a photon (Figure 2). 2 Figure 2. Schematic depicting electron vibrational energy states that occur during FRET.

Measurement The detection and quantitation of FRET can certainly be accomplished in a number of different ways. Because FRET can result in both a decrease in fluorescence of the donor molecule as well as an increase in fluorescence of the acceptor, a ratio metric determination of the two signals can be made. The advantage of this method is that a measure of interaction can be made that is independent of the absolute concentration of the sensor. Because not all acceptor moieties are fluorescent, they can be used as a means to quench fluorescence. In these instances, those interactions that result in a fluorescent donor molecule coming in close proximity to such a molecule would result in a loss of signal. Inversely, reactions that remove the proximity of a fluorescent donor and a quencher would result in an increase in fluorescence. One such example would be protease assays. These assays usually involve a fluorescent moiety on one terminus and a quenching molecule on the other terminus separated by a peptide containing the protease cleavage sequence. Some Examples Genetically encoded fluorescent dyes, such as Green Fluorescent Protein (GFP) and related molecules blue, cyan, yellow and red have provided the ability to perform FRET in vitro, particularly in living cells [2]. These proteins form FRET pairs with each other as well as with conventional dyes. They can be attached to other proteins genetically or covalently yet still retain their fluorescent capability. These dyes have the utility of being genetic elements that can be linked with other genes to form chimeric proteins. These chimeric proteins contain a GFP (or related fluorescent protein element) and a putative binding domain. With different chimeric proteins (one donor and one acceptor) protein-protein interactions can be investigated. Only when the donor/acceptor pairs interacted through protein-protein interactions would FRET result. (Figure 3) Figure 3. Schematic representation of the interaction of two different fluorescent protein chimeras. Protein-protein interactions between proteins labeled A and B bring Blue fluorescent protein and Green fluorescent proteins in close enough proximity to allow for FRET to occur. In this example, excitation of blue fluorescent protein results in the emission of fluorescence by Green fluorescent protein. The organic cyanine dyes Cy3, Cy5, Cy5.5 and Cy7, which emit in the red range (>550 nm), offer a number of advantages. Their emission range is such that background fluorescence is often reduced. Additionally large distances (>100 Å) can be measured as a result of the high extinction coefficients and good quantum yields. Even donor-acceptor pairs with separated emission spectra (i.e. low overlap integral) result in acceptable Förster distances. For example, Cy3, which emits maximally at 570 nm and Cy5, which emits at 670 nm, have a Förster distance >50 Å. Large separation between pairs allows the measurement of acceptor emission as a result of FRET without interference from donor emission. In addition these molecules can be linked directly to specific locations in synthetically produced nucleic acids, allowing FRET to be used to assess nucleic acid annealing. 3

Figure 4. Schematic representation of FRET occurring between Cy3 and Cy5 fluorescent moieties when labeled oligonucleotides are annealed. In the example depicted in Figure 4, two complementary RNA oligonucleotides are labeled with Cy3 and Cy5 respectively. When these labeled molecules are not annealed (Figure 4A), excitation of an RNA oligonucleotide labeled with Cy3 with light at 540 nm results only in the emission of light by Cy3 at 590 nm, while the complementary RNA-oligo labeled with Cy5 does not emit any light at 590 nm or its true emission wavelength of 680 nm. However, when the two oligonucleotides are allowed to anneal, the close proximity of the molecules allows for FRET transfer to occur. This results in the emission of light at 680 nm when the annealed molecule is excited with 540 nm light. Note that not all of the emission of Cy3 at 590 nm is lost, but a significant portion is. Figure 5. Schematic Diagram of FRET activity utilized by VSPs. 4

Voltage Sensor Probes (VSPs) are a Fluorescence Resonance Energy Transfer (FRET)-based assay technology used for high-throughput ion channel drug discovery. The FRET donor is a membrane-bound, coumarin-phospholipid (CC2- DMPE), which binds only to the exterior of the cell membrane. The FRET acceptor is a mobile, negatively charged, hydrophobic oxonol [either DiSBAC 2 (3) or DiSBAC 4 (3)], which will bind to either side of the plasma membrane in response to changes in membrane potential (Figure 5). It is the distance dependence that is utilized with VSPs. Only when the acceptor DISBAC 2 (3) is located on the exterior of the cell membrane is FRET capable of taking place. Resting cells have a relatively negative potential, so the two probes associate with the exterior of the cell membrane, resulting in efficient FRET. Exciting the CC2-DMPE donor probe (at ~400 nm) generates a strong red fluorescence signal (at ~590 nm) from the oxonol acceptor probe. When the membrane potential becomes more positive, as occurs with cell depolarization, the oxonol probe rapidly translocates (on a subsecond time scale) to the other face of the membrane (Figure 1). Thus, each oxonol probe senses and responds to voltage changes in the cell. This translocation separates the FRET pair, so exciting the CC2-DMPE donor probe now generates a strong blue fluorescence signal (at ~460 nm) from the CC2-DMPE probe. Depolarization of the cell, which causes DISBAC 2 (3) to relocate to the inner side of the membrane, would be expected to result in a decrease in FRET activity. Lanthanide compounds, such as europium and terbium, have been effectively used as donors in FRET reactions. These compounds provide very good signal/noise ratios as a result of their long fluorescent half-life and spectral characteristics. The emission of these compounds has a sharply spiked profile with a large Stoke s shift from excitation. In addition, the lengthy fluorescent half-life allows for measurement to begin after the cessation of excitatory light. The delay between excitation and measurement (msec range) allows for background fluorescence from the organic acceptor molecule, with a half-life in the nanosecond range, to dissipate. Thus only donor-acceptor emission is measured when the appropriate delay is enabled. The donor molecule need not always involve a fluorescent compound. Luminescent molecules emit photons in a fashion very similar to fluorescence. The primary difference is that the electron excitation is not the result of photon absorption, but rather from the release of chemical energy contained within the molecule. When the excited electrons return to their ground energy can be released as a photon of light or transferred via RET to an acceptor molecule if the conditions are correct. While more limited in the number of molecules that can be utilized, this technology has the advantage that there is no external excitation of the acceptor molecule. Issues When designing FRET experiments there are a number of issues that need to be considered. The most obvious issue is the matter of close proximity. Depending on the assay design, close proximity will either be established or removed during the assay, resulting in a change in signal that can be measured. Appropriate donor/acceptor pairs need to be selected. The pairs need to have enough spectral overlap for efficient energy transfer to take place, yet have enough of a difference in spectrums as to be distinguishable from one another. The choice of filters for fluorescent wavelength selection is also critical to the success or failure of experimental detection of FRET. The excitation filter for the donor has to be able to selectively excite the donor molecule, while minimizing direct excitation of the acceptor molecule. Contaminating direct excitation of the acceptor molecule can be accounted for using appropriate controls, but large amounts will make interpretation of the data more difficult. As shown in figure 1, the filters used for Cy3 excitation, minimized excitation of Cy5, while providing sufficient excitation signal for Cy3. Another important concern in regards to detection of FRET involves analyte concentration. Only those molecules that interact with one another will result in FRET. If large amounts of donor and acceptor molecules are present, but do not interact, the amount of FRET taking place would be quite low. In this example, while the donor and the acceptor molecules may be very easy to detect separately the actual amount of FRET activity may not be sufficient to detect. In regards to FRET the actual analyte being measured are donor/acceptor pairs rather than the individual components. Additionally, both the donor and acceptor molecules need to be in sufficient concentration in order for FRET to take place. Most binding events are a dynamic process that reaches a steady state. If one of the components of the reaction is in short supply then the total amount bound will be naturally low. For example, transient transfections with two different genetic elements that result in one of the elements not being efficiently translated into protein will result in poor levels of FRET. Cellular localization can play a role as well. If one molecule is located in the cytoplasm while the other molecule has a nuclear location, there will be no interaction with each other despite sufficient quantities of each. 5

FRET Applications Structure and conformation of proteins [3] Spatial distribution and assembly of proteins [4] Receptor/ligand interactions [5] Immunoassays [6] Structure and conformation of nucleic acids [7] Real-time PCR assays and SNP detection [8,9] Nucleic acid hybridization [10] Distribution and transport of lipids [11] Membrane fusion assays [12] Membrane potential sensing [13, 14] Fluorogenic protease assays [15] Indicators for cyclic AMP [16]. References 1. Förster, T. (1948) Intermolecular Energy Migration and Fluorescence Ann. Phys 2:55-75. 2. Tsien, R. (1998) Ann. Review Biochem, 67:509-544 3. Jonsson T, Waldburger CD, Sauer RT, (1996) Nonlinear free energy relationships in Arc repressor unfolding imply the existence of unstable, native-like folding intermediates.. Biochemistry 35:4795-4802. 4. Watson BS, Hazlett TL, Eccleston JF, Davis C, Jameson DM, Johnson AE. (1995) Macromolecular arrangement in the aminoacyl-trna.elongation factor Tu.GTP ternary complex. A fluorescence energy transfer study. Biochemistry 34:7904-7912. 5. Berger W, Prinz H, Striessnig J, Kang HC, Haugland R, Glossmann H. (1994) Complex molecular mechanism for dihydropyridine binding to L-type Ca(2+)-channels as revealed by fluorescence resonance energy transfer. Biochemistry 33: 1875-11883. 6. Khanna PL, Ullman EF. (1980) 4,5 -Dimethoxy-6-carboxyfluorescein: a novel dipole-dipole coupled fluorescence energy transfer acceptor useful for fluorescence immunoassays. Anal Biochem 108:156-161. 7. Clegg RM, Murchie AI, Lilley DM. (1994) The solution structure of the four-way DNA junction at low-salt conditions: a fluorescence resonance energy transfer analysis. Biophys J 66:99-109. 8. Lee LG, Livak KJ, Mullah B, Graham RJ, Vinayak RS, Woudenberg TM. (1999) Seven-color, homogeneous detection of six PCR products. Biotechniques 27:342-349. 9. Myakishev MV, Khripin Y, Hu S, Hamer DH. 2001) High-throughput SNP genotyping by allele-specific PCR with universal energy-transfer-labeled primers. Genome Res 11:163-169. 10. Parkhurst KM, Parkhurst LJ. (1995) Kinetic studies by fluorescence resonance energy transfer employing a doublelabeled oligonucleotide: hybridization to the oligonucleotide complement and to single-stranded DNA. Biochemistry 34:285-292. 11. Nichols JW, Pagano RE. (1983) Resonance energy transfer assay of protein-mediated lipid transfer between vesicles. J Biol Chem 258:5368-5371. 12. Uster PS (1993) In situ resonance energy transfer microscopy: monitoring membrane fusion in living cells. Methods Enzymol. 221:239-246. 13. Hoffman, R. and Held, P. BioTek Application Note. www.biotek.com/products/tech_res_detail.php?id=135 14. Gonzalez JE, Tsien RY. (1995) Voltage sensing by fluorescence resonance energy transfer in single cells. Biophys J 69:1272-1280. 15. Matayoshi ED, Wang GT, Krafft GA, Erickson J. (1990) Novel fluorogenic substrates for assaying retroviral proteases by resonance energy transfer. Science 247, 954-958. 16. Adams SR, et al. (1993) Optical Probes for Cyclic AMP, Fluorescent and Luminescent Probes for Biological Activity, Mason WT, Ed. pp. 133-149. 6 Rev. 06/15/05