Skip to main content
Advertisement

Main menu

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET

User menu

  • My alerts
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Molecular Pharmacology
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET
  • My alerts
  • Log in
  • Log out
  • My Cart
Molecular Pharmacology

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Visit molpharm on Facebook
  • Follow molpharm on Twitter
  • Follow molpharm on LinkedIn
Rapid CommunicationMinireview—Exploring the biology of GPCRs: from in vitro to in vivo

A Perspective on Studying G-Protein–Coupled Receptor Signaling with Resonance Energy Transfer Biosensors in Living Organisms

Jakobus van Unen, Jeanette Woolard, Ago Rinken, Carsten Hoffmann, Stephen J. Hill, Joachim Goedhart, Michael R. Bruchas, Michel Bouvier and Merel J. W. Adjobo-Hermans
Molecular Pharmacology September 2015, 88 (3) 589-595; DOI: https://doi.org/10.1124/mol.115.098897
Jakobus van Unen
Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.J.W.A.-H.); Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada (M.B.); Department of Anesthesiology, Washington University, St. Louis, Missouri (M.R.B.); Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands (J.U., J.G.); Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen’s Medical Centre, University of Nottingham, Nottingham, United Kingdom (J.W., S.J.H.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum and Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.H.); and Institute of Chemistry, University of Tartu, Tartu, Estonia (A.R.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jeanette Woolard
Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.J.W.A.-H.); Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada (M.B.); Department of Anesthesiology, Washington University, St. Louis, Missouri (M.R.B.); Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands (J.U., J.G.); Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen’s Medical Centre, University of Nottingham, Nottingham, United Kingdom (J.W., S.J.H.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum and Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.H.); and Institute of Chemistry, University of Tartu, Tartu, Estonia (A.R.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ago Rinken
Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.J.W.A.-H.); Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada (M.B.); Department of Anesthesiology, Washington University, St. Louis, Missouri (M.R.B.); Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands (J.U., J.G.); Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen’s Medical Centre, University of Nottingham, Nottingham, United Kingdom (J.W., S.J.H.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum and Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.H.); and Institute of Chemistry, University of Tartu, Tartu, Estonia (A.R.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Carsten Hoffmann
Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.J.W.A.-H.); Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada (M.B.); Department of Anesthesiology, Washington University, St. Louis, Missouri (M.R.B.); Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands (J.U., J.G.); Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen’s Medical Centre, University of Nottingham, Nottingham, United Kingdom (J.W., S.J.H.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum and Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.H.); and Institute of Chemistry, University of Tartu, Tartu, Estonia (A.R.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Stephen J. Hill
Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.J.W.A.-H.); Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada (M.B.); Department of Anesthesiology, Washington University, St. Louis, Missouri (M.R.B.); Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands (J.U., J.G.); Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen’s Medical Centre, University of Nottingham, Nottingham, United Kingdom (J.W., S.J.H.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum and Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.H.); and Institute of Chemistry, University of Tartu, Tartu, Estonia (A.R.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Joachim Goedhart
Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.J.W.A.-H.); Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada (M.B.); Department of Anesthesiology, Washington University, St. Louis, Missouri (M.R.B.); Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands (J.U., J.G.); Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen’s Medical Centre, University of Nottingham, Nottingham, United Kingdom (J.W., S.J.H.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum and Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.H.); and Institute of Chemistry, University of Tartu, Tartu, Estonia (A.R.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Michael R. Bruchas
Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.J.W.A.-H.); Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada (M.B.); Department of Anesthesiology, Washington University, St. Louis, Missouri (M.R.B.); Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands (J.U., J.G.); Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen’s Medical Centre, University of Nottingham, Nottingham, United Kingdom (J.W., S.J.H.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum and Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.H.); and Institute of Chemistry, University of Tartu, Tartu, Estonia (A.R.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Michel Bouvier
Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.J.W.A.-H.); Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada (M.B.); Department of Anesthesiology, Washington University, St. Louis, Missouri (M.R.B.); Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands (J.U., J.G.); Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen’s Medical Centre, University of Nottingham, Nottingham, United Kingdom (J.W., S.J.H.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum and Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.H.); and Institute of Chemistry, University of Tartu, Tartu, Estonia (A.R.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Merel J. W. Adjobo-Hermans
Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.J.W.A.-H.); Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada (M.B.); Department of Anesthesiology, Washington University, St. Louis, Missouri (M.R.B.); Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands (J.U., J.G.); Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen’s Medical Centre, University of Nottingham, Nottingham, United Kingdom (J.W., S.J.H.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum and Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.H.); and Institute of Chemistry, University of Tartu, Tartu, Estonia (A.R.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF
Loading

Visual Overview

Figure
  • Download figure
  • Open in new tab
  • Download powerpoint

Abstract

The last frontier for a complete understanding of G-protein–coupled receptor (GPCR) biology is to be able to assess GPCR activity, interactions, and signaling in vivo, in real time within biologically intact systems. This includes the ability to detect GPCR activity, trafficking, dimerization, protein-protein interactions, second messenger production, and downstream signaling events with high spatial resolution and fast kinetic readouts. Resonance energy transfer (RET)–based biosensors allow for all of these possibilities in vitro and in cell-based assays, but moving RET into intact animals has proven difficult. Here, we provide perspectives on the optimization of biosensor design, of signal detection in living organisms, and the multidisciplinary development of in vitro and cell-based assays that more appropriately reflect the physiologic situation. In short, further development of RET-based probes, optical microscopy techniques, and mouse genome editing hold great potential over the next decade to bring real-time in vivo GPCR imaging to the forefront of pharmacology.

Introduction

The study of G-protein–coupled receptor (GPCR) signaling has a long tradition in pharmacology and has led to the development of numerous drug classes that make up almost 30% of all marketed drugs. Initially, the signaling properties of GPCRs were studied in tissue preparations or whole organs with limited knowledge of the individual components and events that were involved in the signaling process. With the advent of molecular biology techniques, it became possible to study and manipulate individual GPCR subtypes and their signaling cascades in transfected cells. The accumulated molecular knowledge of GPCRs and their signaling cascades, together with the advances in fluorescent and luminescent proteins, allowed for the development of resonance energy transfer (RET) sensors by several groups (reviewed by Marullo and Bouvier, 2007; Lohse et al., 2012). RET, based on Förster resonance energy transfer, is the nonradiative transfer of energy from an emitting donor to an absorbing acceptor molecule. RET is perfectly suited to study intramolecular and intermolecular changes in distance and/or orientation due to conformational changes and protein-protein interactions (Hamers et al., 2014). RET-based sensors permit the study of signaling events in living cells with a kinetic resolution down to the millisecond time range (Lohse et al., 2012 and references therein).

Analyses of RET-based sensors are a proven technology to study cellular processes in vitro or in cells. However, as we will discuss, implementation for in vivo use has been challenging. To bridge the gap, the contrast and brightness of RET-based sensors need to be improved, advances in optical detection methods are required, and realistic in vitro or cell-based models need to be developed that more closely resemble the physiologic properties of whole organisms.

In this perspective article we briefly discuss the currently available RET-based approaches used to study GPCR signaling. More importantly, we discuss what would be desirable or required as improvements to extend the use of RET-based sensors in a more physiologically relevant in vitro as well as in vivo setting. This perspective is not meant to be exhaustive but rather to highlight the recent breakthroughs that we judge most relevant when moving RET-biosensors into more physiologically relevant contexts.

State of the Art: RET Sensors

RET-based biosensors to monitor GPCR signaling have been developed for many of the events that occur upon receptor activation, up to the level of second messenger production and downstream protein regulation (Lohse et al., 2012; Clister et al., 2015). The initial events that can be monitored are ligand binding to the receptor (Ma et al., 2014), receptor oligomerization (Angers et al., 2000), receptor activation, heterotrimeric G-protein activation, G-protein–coupled receptor kinase and β-arrestin recruitment (Lohse et al., 2012), and intramolecular changes within β-arrestin (Charest et al., 2005). In general, two detection strategies are used: fluorescence or bioluminescence resonance energy transfer (FRET or BRET, respectively). The major difference between these two approaches is the light source used to monitor the sensor. In case of FRET probes, an external light source is required to excite the donor fluorophore, resulting in high-emission intensity that allows detection with high temporal and spatial resolution. In contrast, for BRET probes an enzymatic substrate conversion and hence chemical light generation achieves the excitation, yielding lower levels of energy but preventing direct excitation of the acceptor. When RET takes place, the excited state energy of the donor is transferred to the acceptor, which returns to the ground state by emission of fluorescence. RET can be optically detected as a decrease in donor emission, a decrease in excited state lifetime, and an increase in acceptor emission (Goedhart et al., 2014).

The most common optical readout for RET is the ratio of acceptor emission over donor emission. Both BRET and FRET measurements have been realized for GPCRs as well as heterotrimeric G-proteins (Lohse et al., 2012).

FRET and BRET probes come in different designs. Popular pairs are two genetically encoded fluorescent proteins in case of FRET (predominantly cyan and yellow variants derived from Aequorea victoria green fluorescent protein [GFP]) or one luminescent protein (frequently luciferase from Renilla reniformis) and one fluorescent protein (frequently variant forms of the GFP from Aequorea victoria) in case of BRET. An alternative combination has been realized by employing a small tetracysteine tag, to which a fluorescein derivative (i.e., FlAsH, fluorescein arsenical hairpin binder) can bind with high specificity as an acceptor (Hoffmann et al., 2010). Other tags (such as SNAP-, CLIP- and Halo-tags) are also available (Comps-Agrar et al., 2011). These genetically encoded tags are enzymes of about 20 kDa that catalyze the covalent attachment of small (fluorescent) molecules to their protein core (Hinner and Johnsson, 2010). Finally, it is also possible to use fluorescent ligands in combination with a GPCR (tagged with either a luminescent or fluorescent protein) in BRET and FRET applications to monitor ligand binding (e.g., Albizu et al., 2010; Stoddart et al., 2015).

Both luminescent proteins and the genetic tags that can covalently bind a fluorescent probe require addition of a cofactor or ligand. This allows flexibility with respect to the choice of the cofactor or fluorescent label. However, for in vivo applications there is the additional challenge of ensuring that substrate molecules (e.g., luciferin for BRET or conjugated dyes for FRET) can reach their desired target (e.g., crossing the blood-brain barrier).

These challenges also apply for the application of fluorescent ligands that are used to probe GPCR binding. Administration needs to be optimized, and the pharmacokinetic properties of the substrate or fluorescent ligand need to be considered in terms of distribution and excretion. For example, the lipophilicity of the fluorescent ligand will determine its local concentration in the immediate vicinity of the GPCR under study (Sykes et al., 2014). In addition, the ligand has to display the necessary receptor subtype selectivity to label the receptor of interest in a cell membrane that may contain other receptors for the same endogenous hormone or neurotransmitter. For example, most cells express multiple adenosine receptors, so it is important to develop highly selective fluorescent ligands for the specific adenosine receptor of interest (Vernall et al., 2013, 2014), especially in the context of an entire organism.

Pharmacokinetic considerations are also important for the use of tetracysteine tag specific FlAsH and its red-shifted variant ReAsH in vivo to prevent aspecific labeling. Although this technology appears to be applicable in tissue slices, it remains to be tested whether it is generally applicable in in vivo settings as well. FlAsH has been used in fly eyes (Venken et al., 2008), supporting the idea that it could become a useful tool for in vivo studies.

Importantly, RET-based sensors have successfully been used in cell culture experiments and have significantly extended our knowledge of agonist efficacy and GPCR function and engagement with intracellular signaling cascades (Lohse et al., 2012; Schann et al., 2013). However, to exploit their full potential and to enable in vivo studies on GPCR signaling, further improvements of RET sensors are required. The biosensors developed for the measurement of second messenger production or the activation of downstream effectors such as kinases detect amplified signals compared with GPCR or G-protein–activation sensors and thus are more sensitive, likely facilitating their use in vivo.

Optimization of RET Sensors for In Vivo Applications

Several RET probes have already been successfully applied in living organisms. They mostly represent intramolecular FRET reporters such as those sensing the activation of small GTPases, the production of the second messenger cGMP, and the protease activity of caspase-3 and calpain (Stockholm et al., 2005; Timpson et al., 2011b; Janssen et al., 2013; Thunemann et al., 2014). An intermolecular sensor has also been described for the interaction between Cdc42 and WASp (Sharifai et al., 2014). In general, these FRET-based biosensors incorporate cyan fluorescent protein (CFP) and yellow fluorescent protein (YFP) for historical reasons, namely, this pair was among the first that were successfully used in a FRET-based sensor in cells. Additionally, the CFP-YFP pair provides good contrast in ratiometric FRET imaging (Goedhart et al., 2014) due to the high quantum yield (QY) of the acceptor. Finally, the weak heterodimerization of this pair strengthens the FRET efficiency in the high-FRET state.

Unfortunately, the CFP-YFP pair has shortcomings that may limit in vivo imaging. The extinction coefficient of CFP is relatively low, and the blue excitation light (∼430 nm) excites autofluorescence. The photostability of YFP is rather poor, limiting the number of photons that can be detected, and the YFP fluorescence is sensitive to pH. As excitation of the donor is not necessary, BRET does not suffer from photobleaching, phototoxicity, or autofluorescence, thereby reducing background signals (Xu et al., 1999; Angers et al., 2000). Indeed, BRET sensors have been used in long-term intravital imaging of zebrafish and mice (Dragulescu-Andrasi et al., 2011; Bakayan et al., 2014). In vivo BRET detection was also possible using a polystyrene skin window and fiber optics to visualize the recruitment of β-arrestin fused to a blue-shifted GFP (GFP2) to V2-vasopressin receptor fused to Renilla luciferase (RLuc) in engineered cells transplanted into mouse kidney (Huang et al., 2007). Recruitment of β-arrestin to the β2-adrenergic receptor (β2AR) could also be detected by BRET in cells and tissue slices derived from transgenic mice expressing GFP2-β-arrestin and β2AR-Rluc (Audet et al., 2010). However, due to their relatively low light output, sensitive spectrometric detection is required and high-resolution subcellular imaging studies are rendered difficult. Yet improvements in BRET probes and microscopy equipment should soon enable BRET imaging at the subcellular level both ex vivo and in vivo. Subcellular resolution was already obtained in engineered human embryonic kidney 293 (HEK293) cells and primary cultures of hippocampal neurons (Coulon et al., 2008; Moutin et al., 2012; Takai et al., 2015).

However, in living organisms, subcellular resolution has, to the best of our knowledge, not yet been reached with respect to BRET sensors and optimized BRET partners (Kamal et al., 2009), and substrates will be needed. For instance, the use of brighter versions of luminescent enzymes such as RLuc8 (Loening et al., 2006), RLuc8-S257G (Takai et al., 2015), and variants thereof such as RLuc2 (Leduc et al., 2009; Breton et al., 2010) in combination with luciferase substrates such as methoxy e-coelenterazine (Me-O-e-CTZ also known as purple coelenterazine), which is 13 times brighter than the coelenterazine-400a (deep-blue coelenterazine), will certainly contribute to shortening the image acquisition time and provide better spatial resolution.

Another avenue to increase the light output for BRET imaging would be to use the GFP from Renilla reniformis (RGFP) rather than from Aequorea victoria as the energy acceptor. Indeed, the RLuc and RGFP combination provides a BRET pair with an optimized spatial relationship between the chromophores and a higher QY that results in much larger signals. Although the naturally occurring self-association between RLuc and RGFP may limit some of the pair’s uses, it proved not to be a problem when the background affinity of the reporter system is lower than that of the reported proteins under study (Molinari et al., 2008). Whether this BRET pair can be of general use and could provide higher resolution remains to be investigated.

Red and near-infrared light shows less scattering and penetrates deeper into tissue. Therefore, many of the fluorescent ligands developed to date incorporate red fluorophores, also to overcome interference by autofluorescence. In addition, fluorophores that are environmentally sensitive (e.g., have an increased QY in a lipid environment such as BODIPY 630/650) and therefore get brighter as ligand binds to a cell surface receptor may be required in vivo where washout of the fluorescent ligand is not possible. Importantly, also BRET and FRET will benefit from red-shifted emission in an in vivo setting. Hence, RET detection would be substantially improved if bright, high QY RFPs would become available. Given recent engineering efforts that resulted in novel bright fluorescent proteins (mTurquoise2, mNeonGreen), we are confident that bright (monomeric) RFPs (QY > 50%) will eventually see the light. Importantly, improving BRET sensors will require the development of brighter luminescence proteins in addition to those described above (e.g., Nanoluc; Hall et al., 2012; Stoddart et al., 2015), which are capable of energy transfer to red-shifted fluorophores. Recently, RFPs have been used in both BRET and FRET sensors in living organisms (Keese et al., 2010; Dragulescu-Andrasi et al., 2011; Bakayan et al., 2014), also for the study of a GPCR (Fruhwirth et al., 2011). A red-shifted unimolecular BRET sensor has also been successfully used to detect a cancer-associated antigen in vivo using an antibody attached to a far-red fluorescent indocyanine derivative fused to Cypridina luciferase via glycol chains (Wu et al., 2009), illustrating the advantages of red-shifted BRET emission for in vivo detection in tissue.

The above-described FRET sensors are commonly sampled based on intensity, which is complicated by the fact that, for example, the CFP signal is more scattered than the YFP signal in tissues. However, FRET-based biosensors can also be analyzed by fluorescence lifetime imaging (FLIM). A change in FRET is detected by a change in donor lifetime (Goedhart et al., 2014). This detection strategy requires a bright, photostable donor and an acceptor with good absorbance (full maturation), not necessarily high QY. However, most FRET-based biosensors show a poor dynamic range when analyzed by FLIM, making it difficult to measure subtle changes. Exceptions include protease sensors and a cAMP sensor that has been optimized for FLIM (Klarenbeek et al., 2015).

A dedicated effort to engineer biosensors with high fluorescence lifetime contrast is crucial to increase our chances to successfully apply biosensors in vivo, as exemplified by the development of a FLIM-based sensor that enables the detection of protein kinase A activity at the subcellular level upon endogenous GPCR activation in acute striatal slices (Chen et al., 2014). Several strategies have been reported that aim at increasing contrast (Hamers et al., 2014). Although such optimization is nowadays standard for ratiometric FRET sensors, similar strategies to improve sensors for FLIM are scarce.

Because the dynamic range of RET-based sensors can be low, alternative strategies for functional imaging of GPCR signaling with high contrast are briefly highlighted, including 1) protein complementation strategies, 2) homotypic fluorescent protein interactions, and 3) insertion of sensory modules in circularly permutated proteins. Protein complementation is a powerful strategy because of its contrast (light or no light), but the downside is that the complementation generally is irreversible, limiting its use in dynamic experiments. A recently reported reversible complementation system may represent a breakthrough in this area (Tchekanda et al., 2014).

Moreover, homotypic interactions between the β-barrels of two fluorescent proteins can lead to fluorescence changes. This was first observed between two GFP barrels and was denoted proximity imaging. A similar system based on RFP has been reported (Alford et al., 2012). A recent extension of this strategy may be employed to qualitatively detect protein-protein interactions or conformation changes (Ding et al., 2015). Interestingly, insertion of sensory modules near the chromophore of a permutated fluorescent protein has resulted in a new generation of high-contrast sensors that detect calcium in vivo (Ahrens et al., 2013). This approach can be generalized, and we expect that this sensor design will be used to engineer biosensors for other processes in the future.

Certainly, end-point-based approaches also provide valuable information and constitute a promising intermediate for looking at GPCR activation in vivo. For instance, S1P1 receptor activity can be detected at the cellular level in mouse tissues and embryos expressing both the S1P1 receptor fused to a transcription factor linked by a protease cleavage site and a β-arrestin/protease fusion. Upon activation of the receptor, β-arrestin is recruited, thereby leading to a release of the transcription factor that can then induce the expression of a GFP reporter gene in the nucleus (Kono et al., 2014).

Detecting RET Sensors in Living Organisms

In addition to the enhancement of probes and the optimization of model systems (e.g., the use of nonpigmented, hairless mice and transparent adult zebrafish), current developments in microscopy techniques and in vivo imaging could yield potentially exciting possibilities for efficient introduction of FRET and BRET probes in animal systems. Several of the previously mentioned FRET sensors have been imaged in living organisms using FLIM, mostly in combination with multiphoton (MP) microscopy.

In MP microscopy, the simultaneous absorption of two or more low-energy infrared photons excites an electron within a fluorophore. The chance for this event to occur is low and therefore only happens in the focal point of the objective. As such, MP microscopy shows reduced phototoxicity and photobleaching, reaching an imaging depth of about 1 mm (compared with 100 μm in confocal microscopy) and inherently leading to optical sectioning. Therefore, it does not require detection pinholes and enables so-called nondescanned detection because there are no photons coming from out-of-focus areas (Timpson et al., 2011a). Importantly, this facilitates the detection of scattered photons, as the detector can be placed as close to the objective as possible. Unfortunately, the generation of infrared photons with wavelengths between 650 and 1100 nm requires an expensive pulsed Ti:sapphire laser.

An important caveat with FLIM-based imaging is the loss of temporal resolution, which can be problematic for real-time measurements. Several recent advances in MP microscopy have provided improved acquisition speeds for FLIM-based approaches (Kirkpatrick et al., 2012; Rinnenthal et al., 2013; Poland et al., 2014). Additionally, commercially available enhancements to microscopy are now available, including gallium arsenide phosphide multidetector units, which have higher quantum efficiency and wavelength sensitivity while still enabling low-noise high-speed imaging.

By virtue of so-called imaging windows, certain organs and tissues are surgically exposed and become accessible to RET detection (Ellenbroek and van Rheenen, 2014). Moreover, in neuroscience, cranial windows are opened and gradient-index optics (GRIN lens) are implanted into deep tissue structures, such that single cells can be visualized in real time in awake-behaving mice. Other reports have used fiber optic photometry approaches to detect subtle changes in calcium responses (Cui et al., 2014; Warden et al., 2014). Optimizing gradient-index lens technology, coupled with advances in materials engineering have dramatically reduced the size of electronics, LEDs, and optics, while maintaining detection and resolution at cellular scales. Also, decreasing the effects of physiologic motion by, for instance, heartbeat-triggered scanning has improved image quality.

Moving deeper into a specimen brings with it more specimen-induced aberrations. Adaptive optics compensate for these aberrations by modulation of the excitation beam (Girkin et al., 2009).

Toward the Imaging of GPCR Signaling in Organs-on-a-Chip?

The approaches we have discussed should enable the introduction of an increasing number of RET-based biosensors into living model organisms for the study of GPCR signaling. In addition to model organisms, the design of optimized in vitro assays has to be considered because proper animal models are not at hand for each human physiologic setting (Langenhan et al., 2015). Also, experiments in living organisms are in general not amenable to extensive manipulation, even though recent breakthroughs allow for the on-demand activation of GPCRs by light (i.e., optoXRs; Masseck et al., 2014). Moreover, to perform high-throughput assays of, for example, drug efficiency, the development of more predictive in vitro assays using patient-derived cells is warranted.

Besides the pharmacologic benefits, these systems will also be very helpful in investigating the fundamental aspects of GPCR signaling. Especially in the field of regenerative medicine, multidisciplinary efforts have led to the development of tools that will be useful to GPCR-related research.

Classically, many signaling studies are performed using a transformed cell line cultured on two-dimensional substrates under atmospheric oxygen pressure, exogenously expressing the protein (sensor) of interest. However, the properties of a GPCR, and the signaling that results from its activation, will depend on native expression levels, cellular/membrane location (e.g., extent of cell polarity and the complexity of its membrane organization), and cell type (Corriden et al., 2013; Head et al., 2014). For this reason it is important to apply the sophisticated approaches described here to native cells. Viruses (e.g., lentivirus or adenovirus) offer approaches to deliver vectors encoding the proteins of interest to native cells. Intramolecular RET-sensors can be directed to the AAVS1 locus, a safe harbor in the human genome (Smith et al., 2014). GPCR sensors are encoded by a single gene, while for heterotrimeric G-protein sensors the α, β, and γ subunits all need to be expressed at similar levels to allow optimal function. The coexpression from a single promoter ensures stoichiometric coexpression of all three subunits and results in robust detection of G-protein activation by FRET (Goedhart et al., 2011). This design simplifies the development of transgenic organisms with multimeric RET sensors.

To obtain physiologically relevant expression levels, intermolecular RET sensors can replace their endogenous counterparts by means of genome editing techniques (i.e., TALEN, ZFN, or CRISPR-Cas; Gaj et al., 2013), provided that this leads to a detectable signal. Reassuringly, several knock-in mice expressing GPCRs fused to a fluorescent protein under the control of an endogenous promoter have been described (Scherrer et al., 2006; for a review, see Ceredig and Massotte, 2014).

Importantly, cells do not only respond to a plethora of soluble compounds because physical cues appear to be equally important in the regulation of cellular function. Both stiffness and dimensionality influence processes such as cell morphology and differentiation. Focal adhesions consisting of, for example, integrins connect the extracellular matrix (ECM) to the actin cytoskeleton inside the cell and enable stiffness sensing. As GPCRs are regulated by and involved in the regulation of focal adhesions (Luttrell and Luttrell, 2004), it is to be expected that GPCR-mediated signaling will also be influenced by the stiffness of the substratum the cells grow on. Indeed, several GPCRs have been reported to be mechanosensors themselves (Storch et al., 2012), and for some of them the functional consequences of their mechanotransduction mechanisms are known to be relevant in pathophysiology (Cook et al., 2014). Moreover, certain adhesion GPCRs have been described to functionally interact with ECM components such as laminin (Monk et al., 2015; Petersen et al., 2015; Scholz et al., 2015).

A straightforward protocol has been developed that enables high-resolution imaging of cells in three-dimensional extracellular matrices of which the stiffness can be controlled (Fischer et al., 2012). However, various other systems are available that, for instance, allow for cellular remodeling because the material used is sensitive to matrix metalloproteinases (Baker and Chen, 2012). The choice for a particular system clearly depends on the cell type(s) and the research question to be investigated.

Recent findings have indicated that the expression levels of certain cell adhesion molecules may change depending on the oxygen level. The same holds for several proteins involved in GPCR-mediated signaling, such as G-protein–coupled receptor kinase-2 and β-arrestin-1 (Lombardi et al., 2004), regulator of G-protein signaling 5 (RGS5) (Jin et al., 2009) and the GPCRs β2AR (Xie et al., 2009) and chemokine (C-X-C motif) receptor type 4 (CXCR4) (Romain et al., 2014). Thus, the use of physiologic oxygen levels, which range between 1 and 11%, may turn out to be important in improving cell-based assays (Carreau et al., 2011). To implement this, the microscope has to be placed in a so-called glove box, thereby providing an environment in which the oxygen level can be tuned.

As one begins to investigate the use of fluorescent ligands and RET-based biosensors in native cells (Audet et al., 2010) there is also the need to consider the impact of neighboring cells on the intracellular responses being measured and whether every cell (even of the same cell phenotype) expresses the same portfolio of GPCRs. For example, in small arteries and arterioles, endothelial cells are coupled to both each other and adjacent vascular smooth muscle cells via gap junctions (Dora and Garland, 2013). In pressurized rat mesenteric arteries, calcium signaling in endothelial cells is subject to regulation by surrounding smooth muscle cells via myoendothelial gap junctions (Kansui et al., 2008). Three-dimensional coculture systems have been developed in which cells cannot attach to the surface and thus must attach to each other. Pipetting together different cell types has been shown to result in spheroid formations consisting of mixed cell types. The recent advent of three-dimensional printing of matrices with cells, also enabling the spatial patterning of ECM components, will lead to even more flexibility in the production of tissue- and organ-like structures (Xu et al., 2013).

Certainly, so-called organoids are in need of oxygen and nutrients. Here, microfluidic devices come into play. The flow they provide not only mimics blood flow, thereby distributing oxygen and nutrients and removing waste products, but also simulates the shear stress experienced by many cell types, which has been shown to induce GPCR signaling (Storch et al., 2012). Moreover, microfluidics can be used to change agonist levels, create chemical gradients, or apply mixtures of soluble cues to investigate their effect on GPCR-mediated signaling (Torisawa et al., 2010).

Many of these devices are prepared from polydimethylsiloxane, which is flexible and, depending on the thickness, permeable to gas. Therefore, regulation of oxygen levels becomes feasible in such devices (Thomas et al., 2011). Polydimethylsiloxane can be coated with ECM components and is transparent, enabling microscopic analysis of FRET probes (Yu et al., 2013).

Using the techniques described here and combining them with patient-derived induced pluripotent stem cells (for a review, see Bellin et al., 2012) and differentiation protocols has led to the development of organs-on-a-chip, and current progress is being made in interconnecting the available organs to obtain a model for human (patho)physiology (van de Stolpe and den Toonder, 2013).

In conclusion, the impressive speed at which novel RET-based biosensors, optical acquisition and detection methods, and organs-on-a-chip techniques are being developed will enable exciting new avenues in both fundamental as well as applied research on GPCR signaling. Due to the multidisciplinary aspects, GPCR biologists will need to collaborate with researchers in the fields of optics, genetics, and materials design to advance our understanding of in vivo GPCR biology.

Authorship Contributions

Wrote or contributed to the writing of the manuscript: Adjobo-Hermans, Bouvier, Bruchas, Goedhart, Hill, Hoffmann, Rinken, Woolard, van Unen.

Footnotes

    • Received March 10, 2015.
    • Accepted May 13, 2015.
  • This work was supported by the National Institutes of Health National Institute on Drug Abuse [R01-DA033396] (to M.R.B.) and by the Estonian Ministry of Education and Research [IUT20-17] (to A.R.).

  • dx.doi.org/10.1124/mol.115.098897.

Abbreviations

β2AR
β2-adrenergic receptor
BRET
bioluminescence resonance energy transfer
CFP
cyan fluorescent protein
ECM
extracellular matrix
FlAsH
fluorescein arsenical hairpin
FLIM
fluorescence lifetime imaging
FRET
fluorescence resonance energy transfer
GFP
green fluorescent protein
GPCR
G-protein–coupled receptor
MP
multiphoton
QY
quantum yield
RET
resonance energy transfer
RGFP
green fluorescent protein from Renilla reniformis
RLuc
Renilla luciferase
YFP
yellow fluorescent protein
  • Copyright © 2015 by The American Society for Pharmacology and Experimental Therapeutics

References

  1. ↵
    1. Ahrens MB,
    2. Orger MB,
    3. Robson DN,
    4. Li JM, and
    5. Keller PJ
    (2013) Whole-brain functional imaging at cellular resolution using light-sheet microscopy. Nat Methods 10:413–420.
    OpenUrlCrossRefPubMed
  2. ↵
    1. Albizu L,
    2. Cottet M,
    3. Kralikova M,
    4. Stoev S,
    5. Seyer R,
    6. Brabet I,
    7. Roux T,
    8. Bazin H,
    9. Bourrier E,
    10. Lamarque L,
    11. et al.
    (2010) Time-resolved FRET between GPCR ligands reveals oligomers in native tissues. Nat Chem Biol 6:587–594.
    OpenUrlCrossRefPubMed
  3. ↵
    1. Alford SC,
    2. Abdelfattah AS,
    3. Ding Y, and
    4. Campbell RE
    (2012) A fluorogenic red fluorescent protein heterodimer. Chem Biol 19:353–360.
    OpenUrlCrossRefPubMed
  4. ↵
    1. Angers S,
    2. Salahpour A,
    3. Joly E,
    4. Hilairet S,
    5. Chelsky D,
    6. Dennis M, and
    7. Bouvier M
    (2000) Detection of beta 2-adrenergic receptor dimerization in living cells using bioluminescence resonance energy transfer (BRET). Proc Natl Acad Sci USA 97:3684–3689.
    OpenUrlAbstract/FREE Full Text
  5. ↵
    1. Audet M,
    2. Lagace M,
    3. Silversides DW, and
    4. Bouvier M
    (2010) Protein-protein interactions monitored in cells from transgenic mice using bioluminescence resonance energy transfer. FASEB J 24:2829–2838.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    1. Bakayan A,
    2. Domingo B,
    3. Miyawaki A, and
    4. Llopis J
    (2014) Imaging Ca2+ activity in mammalian cells and zebrafish with a novel red-emitting aequorin variant. Pflugers Arch DOI: 10.1007/s00424-014-1639-3 [published ahead of print].
  7. ↵
    1. Baker BM and
    2. Chen CS
    (2012) Deconstructing the third dimension: how 3D culture microenvironments alter cellular cues. J Cell Sci 125:3015–3024.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    1. Bellin M,
    2. Marchetto MC,
    3. Gage FH, and
    4. Mummery CL
    (2012) Induced pluripotent stem cells: the new patient? Nat Rev Mol Cell Biol 13:713–726.
    OpenUrlCrossRefPubMed
  9. ↵
    1. Breton B,
    2. Sauvageau É,
    3. Zhou J,
    4. Bonin H,
    5. Le Gouill C, and
    6. Bouvier M
    (2010) Multiplexing of multicolor bioluminescence resonance energy transfer. Biophys J 99:4037–4046.
    OpenUrlCrossRefPubMed
  10. ↵
    1. Carreau A,
    2. El Hafny-Rahbi B,
    3. Matejuk A,
    4. Grillon C, and
    5. Kieda C
    (2011) Why is the partial oxygen pressure of human tissues a crucial parameter? Small molecules and hypoxia. J Cell Mol Med 15:1239–1253.
    OpenUrlCrossRefPubMed
  11. ↵
    1. Ceredig RA and
    2. Massotte D
    (2014) Fluorescent knock-in mice to decipher the physiopathological role of G protein-coupled receptors. Front Pharmacol 5:289.
    OpenUrl
  12. ↵
    1. Charest PG,
    2. Terrillon S, and
    3. Bouvier M
    (2005) Monitoring agonist-promoted conformational changes of beta-arrestin in living cells by intramolecular BRET. EMBO Rep 6:334–340.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    1. Chen Y,
    2. Saulnier JL,
    3. Yellen G, and
    4. Sabatini BL
    (2014) A PKA activity sensor for quantitative analysis of endogenous GPCR signaling via 2-photon FRET-FLIM imaging. Front Pharmacol 5:56.
    OpenUrl
  14. ↵
    1. Clister T,
    2. Mehta S, and
    3. Zhang J
    (2015) Single-cell analysis of G-protein signal transduction. J Biol Chem 290:6681–6688.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    1. Comps-Agrar L,
    2. Maurel D,
    3. Rondard P,
    4. Pin JP,
    5. Trinquet E, and
    6. Prézeau L
    (2011) Cell-surface protein-protein interaction analysis with time-resolved FRET and snap-tag technologies: application to G protein-coupled receptor oligomerization. Methods Mol Biol 756:201–214.
    OpenUrlCrossRefPubMed
  16. ↵
    1. Cook JR,
    2. Carta L,
    3. Bénard L,
    4. Chemaly ER,
    5. Chiu E,
    6. Rao SK,
    7. Hampton TG,
    8. Yurchenco P,
    9. Costa KD,
    10. Hajjar RJ,
    11. et al., and
    12. GenTAC Registry Consortium
    (2014) Abnormal muscle mechanosignaling triggers cardiomyopathy in mice with Marfan syndrome. J Clin Invest 124:1329–1339.
    OpenUrlPubMed
  17. ↵
    1. Corriden R,
    2. Self T,
    3. Akong-Moore K,
    4. Nizet V,
    5. Kellam B,
    6. Briddon SJ, and
    7. Hill SJ
    (2013) Adenosine-A3 receptors in neutrophil microdomains promote the formation of bacteria-tethering cytonemes. EMBO Rep 14:726–732.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    1. Coulon V,
    2. Audet M,
    3. Homburger V,
    4. Bockaert J,
    5. Fagni L,
    6. Bouvier M, and
    7. Perroy J
    (2008) Subcellular imaging of dynamic protein interactions by bioluminescence resonance energy transfer. Biophys J 94:1001–1009.
    OpenUrlCrossRefPubMed
  19. ↵
    1. Cui G,
    2. Jun SB,
    3. Jin X,
    4. Luo G,
    5. Pham MD,
    6. Lovinger DM,
    7. Vogel SS, and
    8. Costa RM
    (2014) Deep brain optical measurements of cell type-specific neural activity in behaving mice. Nat Protoc 9:1213–1228.
    OpenUrlCrossRef
  20. ↵
    1. Ding Y,
    2. Li J,
    3. Enterina JR,
    4. Shen Y,
    5. Zhang I,
    6. Tewson PH,
    7. Mo GC,
    8. Zhang J,
    9. Quinn AM,
    10. Hughes TE,
    11. Maysinger D,
    12. Alford SC,
    13. Zhang Y, and
    14. Campbell RE
    (2015) Ratiometric biosensors based on dimerization-dependent fluorescent protein exchange. Nat Methods 12:195–198.
    OpenUrlCrossRef
  21. ↵
    1. Dora KA and
    2. Garland CJ
    (2013) Linking hyperpolarization to endothelial cell calcium events in arterioles. Microcirculation 20:248–256.
    OpenUrlCrossRefPubMed
  22. ↵
    1. Dragulescu-Andrasi A,
    2. Chan CT,
    3. De A,
    4. Massoud TF, and
    5. Gambhir SS
    (2011) Bioluminescence resonance energy transfer (BRET) imaging of protein-protein interactions within deep tissues of living subjects. Proc Natl Acad Sci USA 108:12060–12065.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    1. Ellenbroek SI and
    2. van Rheenen J
    (2014) Imaging hallmarks of cancer in living mice. Nat Rev Cancer 14:406–418.
    OpenUrlCrossRefPubMed
  24. ↵
    1. Fischer RS,
    2. Myers KA,
    3. Gardel ML, and
    4. Waterman CM
    (2012) Stiffness-controlled three-dimensional extracellular matrices for high-resolution imaging of cell behavior. Nat Protoc 7:2056–2066.
    OpenUrlCrossRefPubMed
  25. ↵
    1. Fruhwirth GO,
    2. Fernandes LP,
    3. Weitsman G,
    4. Patel G,
    5. Kelleher M,
    6. Lawler K,
    7. Brock A,
    8. Poland SP,
    9. Matthews DR,
    10. Kéri G,
    11. et al.
    (2011) How Förster resonance energy transfer imaging improves the understanding of protein interaction networks in cancer biology. Chemphyschem 12:442–461.
    OpenUrlCrossRefPubMed
  26. ↵
    1. Gaj T,
    2. Gersbach CA, and
    3. Barbas CF 3rd.
    (2013) ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol 31:397–405.
    OpenUrlCrossRefPubMed
  27. ↵
    1. Girkin JM,
    2. Poland S, and
    3. Wright AJ
    (2009) Adaptive optics for deeper imaging of biological samples. Curr Opin Biotechnol 20:106–110.
    OpenUrlCrossRefPubMed
  28. ↵
    1. Goedhart J,
    2. Hink MA, and
    3. Jalink K
    (2014) An introduction to fluorescence imaging techniques geared towards biosensor applications. Methods Mol Biol 1071:17–28.
    OpenUrlCrossRef
  29. ↵
    1. Goedhart J,
    2. van Weeren L,
    3. Adjobo-Hermans MJ,
    4. Elzenaar I,
    5. Hink MA, and
    6. Gadella TW Jr.
    (2011) Quantitative co-expression of proteins at the single cell level—application to a multimeric FRET sensor. PLoS ONE 6:e27321.
    OpenUrlCrossRefPubMed
  30. ↵
    1. Hall MP,
    2. Unch J,
    3. Binkowski BF,
    4. Valley MP,
    5. Butler BL,
    6. Wood MG,
    7. Otto P,
    8. Zimmerman K,
    9. Vidugiris G,
    10. Machleidt T,
    11. et al.
    (2012) Engineered luciferase reporter from a deep sea shrimp utilizing a novel imidazopyrazinone substrate. ACS Chem Biol 7:1848–1857.
    OpenUrlCrossRefPubMed
  31. ↵
    1. Hamers D,
    2. van Voorst Vader L,
    3. Borst JW, and
    4. Goedhart J
    (2014) Development of FRET biosensors for mammalian and plant systems. Protoplasma 251:333–347.
    OpenUrlCrossRef
  32. ↵
    1. Head BP,
    2. Patel HH, and
    3. Insel PA
    (2014) Interaction of membrane/lipid rafts with the cytoskeleton: impact on signaling and function: membrane/lipid rafts, mediators of cytoskeletal arrangement and cell signaling. Biochim Biophys Acta 1838:532–545.
    OpenUrlCrossRefPubMed
  33. ↵
    1. Hinner MJ and
    2. Johnsson K
    (2010) How to obtain labeled proteins and what to do with them. Curr Opin Biotechnol 21:766–776.
    OpenUrlCrossRefPubMed
  34. ↵
    1. Hoffmann C,
    2. Gaietta G,
    3. Zürn A,
    4. Adams SR,
    5. Terrillon S,
    6. Ellisman MH,
    7. Tsien RY, and
    8. Lohse MJ
    (2010) Fluorescent labeling of tetracysteine-tagged proteins in intact cells. Nat Protoc 5:1666–1677.
    OpenUrlCrossRefPubMed
  35. ↵
    1. Huang Q,
    2. Acha V,
    3. Yow R,
    4. Schneider E,
    5. Sardar DK, and
    6. Hornsby PJ
    (2007) Bioluminescence measurements in mice using a skin window. J Biomed Opt 12:054012.
    OpenUrlCrossRefPubMed
  36. ↵
    1. Janssen A,
    2. Beerling E,
    3. Medema R, and
    4. van Rheenen J
    (2013) Intravital FRET imaging of tumor cell viability and mitosis during chemotherapy. PLoS ONE 8:e64029.
    OpenUrlCrossRefPubMed
  37. ↵
    1. Jin Y,
    2. An X,
    3. Ye Z,
    4. Cully B,
    5. Wu J, and
    6. Li J
    (2009) RGS5, a hypoxia-inducible apoptotic stimulator in endothelial cells. J Biol Chem 284:23436–23443.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    1. Kamal M,
    2. Marquez M,
    3. Vauthier V,
    4. Leloire A,
    5. Froguel P,
    6. Jockers R, and
    7. Couturier C
    (2009) Improved donor/acceptor BRET couples for monitoring beta-arrestin recruitment to G protein-coupled receptors. Biotechnol J 4:1337–1344.
    OpenUrlCrossRefPubMed
  39. ↵
    1. Kansui Y,
    2. Garland CJ, and
    3. Dora KA
    (2008) Enhanced spontaneous Ca2+ events in endothelial cells reflect signalling through myoendothelial gap junctions in pressurized mesenteric arteries. Cell Calcium 44:135–146.
    OpenUrlCrossRefPubMed
  40. ↵
    1. Keese M,
    2. Yagublu V,
    3. Schwenke K,
    4. Post S, and
    5. Bastiaens P
    (2010) Fluorescence lifetime imaging microscopy of chemotherapy-induced apoptosis resistance in a syngenic mouse tumor model. Int J Cancer 126:104–113.
    OpenUrlCrossRefPubMed
  41. ↵
    1. Kirkpatrick ND,
    2. Chung E,
    3. Cook DC,
    4. Han X,
    5. Gruionu G,
    6. Liao S,
    7. Munn LL,
    8. Padera TP,
    9. Fukumura D, and
    10. Jain RK
    (2012) Video-rate resonant scanning multiphoton microscopy: an emerging technique for intravital imaging of the tumor microenvironment. Intravital 1:10.4161/intv.21557.
    OpenUrl
  42. ↵
    1. Klarenbeek J,
    2. Goedhart J,
    3. van Batenburg A,
    4. Groenewald D, and
    5. Jalink K
    (2015) Fourth-generation Epac-based FRET sensors for cAMP feature exceptional brightness, photostability and dynamic range: characterization of dedicated sensors for FLIM, for ratiometry and with high affinity. PLoS ONE 10:e0122513.
    OpenUrlCrossRef
  43. ↵
    1. Kono M,
    2. Tucker AE,
    3. Tran J,
    4. Bergner JB,
    5. Turner EM, and
    6. Proia RL
    (2014) Sphingosine-1-phosphate receptor 1 reporter mice reveal receptor activation sites in vivo. J Clin Invest 124:2076–2086.
    OpenUrlCrossRefPubMed
  44. ↵
    1. Langenhan T,
    2. Barr MM,
    3. Bruchas MR,
    4. Ewer J,
    5. Griffith LC,
    6. Maiellaro I,
    7. Taghert PH,
    8. White BH, and
    9. Monk KR
    (2015) Model organisms in GPCR research. Mol Pharmacol 88:596–603.
    OpenUrlAbstract/FREE Full Text
  45. ↵
    1. Leduc M,
    2. Breton B,
    3. Galés C,
    4. Le Gouill C,
    5. Bouvier M,
    6. Chemtob S, and
    7. Heveker N
    (2009) Functional selectivity of natural and synthetic prostaglandin EP4 receptor ligands. J Pharmacol Exp Ther 331:297–307.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    1. Loening AM,
    2. Fenn TD,
    3. Wu AM, and
    4. Gambhir SS
    (2006) Consensus guided mutagenesis of Renilla luciferase yields enhanced stability and light output. Protein Eng Des Sel 19:391–400.
    OpenUrlAbstract/FREE Full Text
  47. ↵
    1. Lohse MJ,
    2. Nuber S, and
    3. Hoffmann C
    (2012) Fluorescence/bioluminescence resonance energy transfer techniques to study G-protein-coupled receptor activation and signaling. Pharmacol Rev 64:299–336.
    OpenUrlAbstract/FREE Full Text
  48. ↵
    1. Lombardi MS,
    2. van den Tweel E,
    3. Kavelaars A,
    4. Groenendaal F,
    5. van Bel F, and
    6. Heijnen CJ
    (2004) Hypoxia/ischemia modulates G protein-coupled receptor kinase 2 and beta-arrestin-1 levels in the neonatal rat brain. Stroke 35:981–986.
    OpenUrlAbstract/FREE Full Text
  49. ↵
    1. Luttrell DK and
    2. Luttrell LM
    (2004) Not so strange bedfellows: G-protein-coupled receptors and Src family kinases. Oncogene 23:7969–7978.
    OpenUrlCrossRefPubMed
  50. ↵
    1. Ma Z,
    2. Du L, and
    3. Li M
    (2014) Toward fluorescent probes for G-protein-coupled receptors (GPCRs). J Med Chem 57:8187–8203.
    OpenUrlCrossRef
  51. ↵
    1. Marullo S and
    2. Bouvier M
    (2007) Resonance energy transfer approaches in molecular pharmacology and beyond. Trends Pharmacol Sci 28:362–365.
    OpenUrlCrossRefPubMed
  52. ↵
    1. Masseck OA,
    2. Spoida K,
    3. Dalkara D,
    4. Maejima T,
    5. Rubelowski JM,
    6. Wallhorn L,
    7. Deneris ES, and
    8. Herlitze S
    (2014) Vertebrate cone opsins enable sustained and highly sensitive rapid control of Gi/o signaling in anxiety circuitry. Neuron 81:1263–1273.
    OpenUrlCrossRefPubMed
  53. ↵
    1. Molinari P,
    2. Casella I, and
    3. Costa T
    (2008) Functional complementation of high-efficiency resonance energy transfer: a new tool for the study of protein binding interactions in living cells. Biochem J 409:251–261.
    OpenUrlAbstract/FREE Full Text
  54. ↵
    1. Monk KR,
    2. Hamann J,
    3. Langenhan T,
    4. Nijmeijer S,
    5. Schoneber T, and
    6. Liebscher I
    (2015) Adhesion GPCRs: from in vitro pharmacology to in vivo mechanisms. Mol Pharmacol 88:617–623.
    OpenUrlAbstract/FREE Full Text
  55. ↵
    1. Moutin E,
    2. Raynaud F,
    3. Fagni L, and
    4. Perroy J
    (2012) GKAP-DLC2 interaction organizes the postsynaptic scaffold complex to enhance synaptic NMDA receptor activity. J Cell Sci 125:2030–2040.
    OpenUrlAbstract/FREE Full Text
  56. ↵
    1. Petersen SC,
    2. Luo R,
    3. Liebscher I,
    4. Giera S,
    5. Jeong SJ,
    6. Mogha A,
    7. Ghidinelli M,
    8. Feltri ML,
    9. Schöneberg T,
    10. Piao X,
    11. et al.
    (2015) The adhesion GPCR GPR126 has distinct, domain-dependent functions in Schwann cell development mediated by interaction with laminin-211. Neuron 85:755–769.
    OpenUrlCrossRefPubMed
  57. ↵
    1. Poland SP,
    2. Krstajić N,
    3. Coelho S,
    4. Tyndall D,
    5. Walker RJ,
    6. Devauges V,
    7. Morton PE,
    8. Nicholas NS,
    9. Richardson J,
    10. Li DD,
    11. et al.
    (2014) Time-resolved multifocal multiphoton microscope for high speed FRET imaging in vivo. Opt Lett 39:6013–6016.
    OpenUrlCrossRef
  58. ↵
    1. Rinnenthal JL,
    2. Börnchen C,
    3. Radbruch H,
    4. Andresen V,
    5. Mossakowski A,
    6. Siffrin V,
    7. Seelemann T,
    8. Spiecker H,
    9. Moll I,
    10. Herz J,
    11. et al.
    (2013) Parallelized TCSPC for dynamic intravital fluorescence lifetime imaging: quantifying neuronal dysfunction in neuroinflammation. PLoS ONE 8:e60100.
    OpenUrlCrossRef
  59. ↵
    1. Romain B,
    2. Hachet-Haas M,
    3. Rohr S,
    4. Brigand C,
    5. Galzi JL,
    6. Gaub MP,
    7. Pencreach E, and
    8. Guenot D
    (2014) Hypoxia differentially regulated CXCR4 and CXCR7 signaling in colon cancer. Mol Cancer 13:58.
    OpenUrlCrossRef
  60. ↵
    1. Schann S,
    2. Bouvier M, and
    3. Neuville P
    (2013) Technology combination to address GPCR allosteric modulator drug-discovery pitfalls. Drug Discov Today Technol 10:e261–e267.
    OpenUrlCrossRef
  61. ↵
    1. Scherrer G,
    2. Tryoen-Tóth P,
    3. Filliol D,
    4. Matifas A,
    5. Laustriat D,
    6. Cao YQ,
    7. Basbaum AI,
    8. Dierich A,
    9. Vonesh JL,
    10. Gavériaux-Ruff C,
    11. et al.
    (2006) Knockin mice expressing fluorescent delta-opioid receptors uncover G protein-coupled receptor dynamics in vivo. Proc Natl Acad Sci USA 103:9691–9696.
    OpenUrlAbstract/FREE Full Text
  62. ↵
    1. Scholz N,
    2. Gehring J,
    3. Guan C,
    4. Ljaschenko D,
    5. Fischer R,
    6. Lakshmanan V,
    7. Kittel RJ, and
    8. Langenhan T
    (2015) The adhesion GPCR latrophilin/CIRL shapes mechanosensation. Cell Reports 11:866–874.
    OpenUrlCrossRef
  63. ↵
    1. Sharifai N,
    2. Samarajeewa H,
    3. Kamiyama D,
    4. Deng TC,
    5. Boulina M, and
    6. Chiba A
    (2014) Imaging dynamic molecular signaling by the Cdc42 GTPase within the developing CNS. PLoS ONE 9:e88870.
    OpenUrlCrossRef
  64. ↵
    1. Smith LJ,
    2. Ul-Hasan T,
    3. Carvaines SK,
    4. Van Vliet K,
    5. Yang E,
    6. Wong KK Jr.,
    7. Agbandje-McKenna M, and
    8. Chatterjee S
    (2014) Gene transfer properties and structural modeling of human stem cell-derived AAV. Mol Ther 22:1625–1634.
    OpenUrl
  65. ↵
    1. Stockholm D,
    2. Bartoli M,
    3. Sillon G,
    4. Bourg N,
    5. Davoust J, and
    6. Richard I
    (2005) Imaging calpain protease activity by multiphoton FRET in living mice. J Mol Biol 346:215–222.
    OpenUrlCrossRefPubMed
  66. ↵
    1. Stoddart LA,
    2. Johnstone EKM,
    3. Wheal AJ,
    4. Goulding J,
    5. Robers MB,
    6. Machleidt T,
    7. Wood KV,
    8. Hill SJ, and
    9. Pfleger KDG
    (2015) Application of BRET to monitor ligand binding to GPCRs. Nat Methods DOI: 10.1038/nmeth.3398.
  67. ↵
    1. Storch U,
    2. Mederos y Schnitzler M, and
    3. Gudermann T
    (2012) G protein-mediated stretch reception. Am J Physiol Heart Circ Physiol 302:H1241–H1249.
    OpenUrlAbstract/FREE Full Text
  68. ↵
    1. Sykes DA,
    2. Parry C,
    3. Reilly J,
    4. Wright P,
    5. Fairhurst RA, and
    6. Charlton SJ
    (2014) Observed drug-receptor association rates are governed by membrane affinity: the importance of establishing “micro-pharmacokinetic/pharmacodynamic relationships” at the β2-adrenoceptor. Mol Pharmacol 85:608–617.
    OpenUrlAbstract/FREE Full Text
  69. ↵
    1. Takai A,
    2. Nakano M,
    3. Saito K,
    4. Haruno R,
    5. Watanabe TM,
    6. Ohyanagi T,
    7. Jin T,
    8. Okada Y, and
    9. Nagai T
    (2015) Expanded palette of nano-lanterns for real-time multicolor luminescence imaging. Proc Natl Acad Sci USA 112:4352–4356.
    OpenUrlAbstract/FREE Full Text
  70. ↵
    1. Tchekanda E,
    2. Sivanesan D, and
    3. Michnick SW
    (2014) An infrared reporter to detect spatiotemporal dynamics of protein-protein interactions. Nat Methods 11:641–644.
    OpenUrlCrossRefPubMed
  71. ↵
    1. Thomas PC,
    2. Raghavan SR, and
    3. Forry SP
    (2011) Regulating oxygen levels in a microfluidic device. Anal Chem 83:8821–8824.
    OpenUrlCrossRefPubMed
  72. ↵
    1. Thunemann M,
    2. Schmidt K,
    3. de Wit C,
    4. Han X,
    5. Jain RK,
    6. Fukumura D, and
    7. Feil R
    (2014) Correlative intravital imaging of cGMP signals and vasodilation in mice. Front Physiol 5:394.
    OpenUrlPubMed
  73. ↵
    1. Timpson P,
    2. McGhee EJ, and
    3. Anderson KI
    (2011a) Imaging molecular dynamics in vivo—from cell biology to animal models. J Cell Sci 124:2877–2890.
    OpenUrlAbstract/FREE Full Text
  74. ↵
    1. Timpson P,
    2. McGhee EJ,
    3. Morton JP,
    4. von Kriegsheim A,
    5. Schwarz JP,
    6. Karim SA,
    7. Doyle B,
    8. Quinn JA,
    9. Carragher NO,
    10. Edward M,
    11. et al.
    (2011b) Spatial regulation of RhoA activity during pancreatic cancer cell invasion driven by mutant p53. Cancer Res 71:747–757.
    OpenUrlAbstract/FREE Full Text
  75. ↵
    1. Torisawa YS,
    2. Mosadegh B,
    3. Bersano-Begey T,
    4. Steele JM,
    5. Luker KE,
    6. Luker GD, and
    7. Takayama S
    (2010) Microfluidic platform for chemotaxis in gradients formed by CXCL12 source-sink cells. Integr Biol 2:680–686.
    OpenUrlCrossRef
  76. ↵
    1. van de Stolpe A and
    2. den Toonder J
    (2013) Workshop meeting report organs-on-chips: human disease models. Lab Chip 13:3449–3470.
    OpenUrlCrossRefPubMed
  77. ↵
    1. Venken KJ,
    2. Kasprowicz J,
    3. Kuenen S,
    4. Yan J,
    5. Hassan BA, and
    6. Verstreken P
    (2008) Recombineering-mediated tagging of Drosophila genomic constructs for in vivo localization and acute protein inactivation. Nucleic Acids Res 36:e114.
    OpenUrlAbstract/FREE Full Text
  78. ↵
    1. Vernall AJ,
    2. Hill SJ, and
    3. Kellam B
    (2014) The evolving small-molecule fluorescent-conjugate toolbox for class A GPCRs. Br J Pharmacol 171:1073–1084.
    OpenUrlCrossRef
  79. ↵
    1. Vernall AJ,
    2. Stoddart LA,
    3. Briddon SJ,
    4. Ng HW,
    5. Laughton CA,
    6. Doughty SW,
    7. Hill SJ, and
    8. Kellam B
    (2013) Conversion of a non-selective adenosine receptor antagonist into A3-selective high affinity fluorescent probes using peptide-based linkers. Org Biomol Chem 11:5673–5682.
    OpenUrlCrossRef
  80. ↵
    1. Warden MR,
    2. Cardin JA, and
    3. Deisseroth K
    (2014) Optical neural interfaces. Annu Rev Biomed Eng 16:103–129.
    OpenUrlCrossRefPubMed
  81. ↵
    1. Wu C,
    2. Mino K,
    3. Akimoto H,
    4. Kawabata M,
    5. Nakamura K,
    6. Ozaki M, and
    7. Ohmiya Y
    (2009) In vivo far-red luminescence imaging of a biomarker based on BRET from Cypridina bioluminescence to an organic dye. Proc Natl Acad Sci USA 106:15599–15603.
    OpenUrlAbstract/FREE Full Text
  82. ↵
    1. Xie L,
    2. Xiao K,
    3. Whalen EJ,
    4. Forrester MT,
    5. Freeman RS,
    6. Fong G,
    7. Gygi SP,
    8. Lefkowitz RJ, and
    9. Stamler JS
    (2009) Oxygen-regulated β2-adrenergic receptor hydroxylation by EGLN3 and ubiquitylation by pVHL. Sci Signal 2:ra33.
    OpenUrlAbstract/FREE Full Text
  83. ↵
    1. Xu T,
    2. Zhao W,
    3. Zhu JM,
    4. Albanna MZ,
    5. Yoo JJ, and
    6. Atala A
    (2013) Complex heterogeneous tissue constructs containing multiple cell types prepared by inkjet printing technology. Biomaterials 34:130–139.
    OpenUrlCrossRefPubMed
  84. ↵
    1. Xu Y,
    2. Piston DW, and
    3. Johnson CH
    (1999) A bioluminescence resonance energy transfer (BRET) system: application to interacting circadian clock proteins. Proc Natl Acad Sci USA 96:151–156.
    OpenUrlAbstract/FREE Full Text
  85. ↵
    1. Yu JQ,
    2. Liu XF,
    3. Chin LK,
    4. Liu AQ, and
    5. Luo KQ
    (2013) Study of endothelial cell apoptosis using fluorescence resonance energy transfer (FRET) biosensor cell line with hemodynamic microfluidic chip system. Lab Chip 13:2693–2700.
    OpenUrlCrossRef
PreviousNext
Back to top

In this issue

Molecular Pharmacology: 88 (3)
Molecular Pharmacology
Vol. 88, Issue 3
1 Sep 2015
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Editorial Board (PDF)
  • Front Matter (PDF)
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Molecular Pharmacology article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
A Perspective on Studying G-Protein–Coupled Receptor Signaling with Resonance Energy Transfer Biosensors in Living Organisms
(Your Name) has forwarded a page to you from Molecular Pharmacology
(Your Name) thought you would be interested in this article in Molecular Pharmacology.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Rapid CommunicationMinireview—Exploring the biology of GPCRs: from in vitro to in vivo

RET Biosensors to Study GPCR Signaling in Living Organisms

Jakobus van Unen, Jeanette Woolard, Ago Rinken, Carsten Hoffmann, Stephen J. Hill, Joachim Goedhart, Michael R. Bruchas, Michel Bouvier and Merel J. W. Adjobo-Hermans
Molecular Pharmacology September 1, 2015, 88 (3) 589-595; DOI: https://doi.org/10.1124/mol.115.098897

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Rapid CommunicationMinireview—Exploring the biology of GPCRs: from in vitro to in vivo

RET Biosensors to Study GPCR Signaling in Living Organisms

Jakobus van Unen, Jeanette Woolard, Ago Rinken, Carsten Hoffmann, Stephen J. Hill, Joachim Goedhart, Michael R. Bruchas, Michel Bouvier and Merel J. W. Adjobo-Hermans
Molecular Pharmacology September 1, 2015, 88 (3) 589-595; DOI: https://doi.org/10.1124/mol.115.098897
del.icio.us logo Digg logo Reddit logo Twitter logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Visual Overview
    • Abstract
    • Introduction
    • State of the Art: RET Sensors
    • Optimization of RET Sensors for In Vivo Applications
    • Detecting RET Sensors in Living Organisms
    • Toward the Imaging of GPCR Signaling in Organs-on-a-Chip?
    • Authorship Contributions
    • Footnotes
    • Abbreviations
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Spatial and Temporal Aspects of Signaling by GPCRs
  • Residence Time at GPCRs
  • Model Organisms in GPCR Research
Show more Minireview—Exploring the biology of GPCRs: from in vitro to in vivo

Similar Articles

Advertisement
  • Home
  • Alerts
Facebook   Twitter   LinkedIn   RSS

Navigate

  • Current Issue
  • Fast Forward by date
  • Fast Forward by section
  • Latest Articles
  • Archive
  • Search for Articles
  • Feedback
  • ASPET

More Information

  • About Molecular Pharmacology
  • Editorial Board
  • Instructions to Authors
  • Submit a Manuscript
  • Customized Alerts
  • RSS Feeds
  • Subscriptions
  • Permissions
  • Terms & Conditions of Use

ASPET's Other Journals

  • Drug Metabolism and Disposition
  • Journal of Pharmacology and Experimental Therapeutics
  • Pharmacological Reviews
  • Pharmacology Research & Perspectives
ISSN 1521-0111 (Online)

Copyright © 2022 by the American Society for Pharmacology and Experimental Therapeutics