Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 31
Filtrer
1.
J Med Chem ; 67(15): 12618-12631, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39044606

RÉSUMÉ

The delta opioid receptor (δOR or DOR) is a G protein-coupled receptor (GPCR) showing a promising profile as a drug target for nociception and analgesia. Herein, we design and synthesize new fluorescent antagonist probes with high δOR selectivity that are ideally suited for single-molecule microscopy (SMM) applications in unmodified, untagged receptors. Using our new probes, we investigated wild-type δOR localization and mobility at low physiological receptor densities for the first time. Furthermore, we investigate the potential formation of δOR homodimers, as such a receptor organization might exhibit distinct pharmacological activity, potentially paving the way for innovative pharmacological therapies. Our findings indicate that the majority of δORs labeled with these probes exist as freely diffusing monomers on the cell surface in a simple cell model. This discovery advances our understanding of OR behavior and offers potential implications for future therapeutic research.


Sujet(s)
Conception de médicament , Colorants fluorescents , Récepteur delta , Récepteur delta/métabolisme , Récepteur delta/antagonistes et inhibiteurs , Colorants fluorescents/composition chimique , Colorants fluorescents/synthèse chimique , Humains , Imagerie de molécules uniques/méthodes , Cellules HEK293 , Animaux , Microscopie de fluorescence
2.
AIDS ; 38(10): 1449-1459, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38770825

RÉSUMÉ

OBJECTIVE: CCR5, a G protein-coupled receptor (GPCR), is used by most HIV strains as a coreceptor. In this study, we looked for other GPCR able to modify HIV-1 infection. DESIGN: We analyzed the effects of one GPCR coexpressed with CCR5, EBI2, on HIV-1 replicative cycle. METHODS: We identified GPCR expressed in primary CD4 + CCR5 + T cells by multi-RT-qPCR. We studied GPCR dimerization by FRET technology. Cell lines expressing EBI2 were established by transduction with HIV vectors. HIV-1 entry was quantified with virions harboring ß-lactamase fused to the viral protein vpr, early and late HIV-1 transcriptions by qPCR, NFkB nuclear activation by immunofluorescence and transfection, and viral production by measuring p24 concentration in culture supernatant by ELISA. RESULTS: We showed that EBI2 is naturally expressed in primary CD4 + CCR5 + T cells, and that CCR5 and EBI2 heterodimerize. We observed that this coexpression reduced viral entry by 50%. The amount of HIV reverse transcripts was similar in cells expressing or not EBI2. Finally, the presence of EBI2 induced the translocation of NFkB and activated HIV-1 genome expression. Globally, the result was a drastic HIV-1 R5, but not X4, overproduction in EBI2 -transduced cells. CONCLUSION: EBI2 expression in CD4 + CCR5 + cells boosts HIV-1 R5 productive infection. As the natural ligand for EBI2 is present in blood and lymphoid tissues, the constant EBI2 activation might increase HIV replication in CD4 + T cells. It might be of interest to test the effect of EBI2 antagonists on the residual viral production persisting in patients aviremic under treatment.


Sujet(s)
Lymphocytes T CD4+ , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Récepteurs CCR5 , Récepteurs couplés aux protéines G , Réplication virale , Humains , Récepteurs CCR5/métabolisme , Lymphocytes T CD4+/virologie , Lymphocytes T CD4+/métabolisme , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Pénétration virale , Cellules cultivées , Infections à VIH/virologie , Infections à VIH/métabolisme , Multimérisation de protéines , Expression des gènes
3.
ACS Chem Neurosci ; 15(3): 645-655, 2024 02 07.
Article de Anglais | MEDLINE | ID: mdl-38275568

RÉSUMÉ

In recent years, there has been growing interest in the potential therapeutic use of inhibitors of adenosine A2A receptors (A2AR) for the treatment of neurodegenerative diseases and cancer. Nevertheless, the widespread expression of A2AR throughout the body emphasizes the importance of temporally and spatially selective ligands. Photopharmacology is an emerging strategy that utilizes photosensitive ligands to attain high spatiotemporal precision and regulate the function of biomolecules using light. In this study, we combined photochemistry and cellular and in vivo photopharmacology to investigate the light sensitivity of the FDA-approved antagonist istradefylline and its potential use as an A2AR photopharmacological tool. Our findings reveal that istradefylline exhibits rapid trans-to-cis isomerization under near-UV light, and prolonged exposure results in the formation of photocycloaddition products. We demonstrate that exposure to UV light triggers a time-dependent decrease in the antagonistic activity of istradefylline in A2AR-expressing cells and enables real-time optical control of A2AR signaling in living cells and zebrafish. Together, these data demonstrate that istradefylline is a photoinactivatable A2AR antagonist and that this property can be utilized to perform photopharmacological experiments in living cells and animals.


Sujet(s)
Récepteur A2A à l'adénosine , Danio zébré , Animaux , Récepteur A2A à l'adénosine/métabolisme , Danio zébré/métabolisme , Purines/pharmacologie , Transduction du signal , Antagonistes des récepteurs A2 à l'adénosine/usage thérapeutique
4.
ACS Chem Biol ; 17(10): 2744-2752, 2022 10 21.
Article de Anglais | MEDLINE | ID: mdl-36149353

RÉSUMÉ

Recently determined structures of class C G protein-coupled receptors (GPCRs) revealed the location of allosteric binding sites and opened new opportunities for the discovery of novel modulators. In this work, molecular docking screens for allosteric modulators targeting the metabotropic glutamate receptor 5 (mGlu5) were performed. The mGlu5 receptor is activated by the main excitatory neurotransmitter of the nervous central system, L-glutamate, and mGlu5 receptor activity can be allosterically modulated by negative or positive allosteric modulators. The mGlu5 receptor is a promising target for the treatment of psychiatric and neurodegenerative diseases, and several allosteric modulators of this GPCR have been evaluated in clinical trials. Chemical libraries containing fragment- (1.6 million molecules) and lead-like (4.6 million molecules) compounds were docked to an allosteric binding site of mGlu5 identified in X-ray crystal structures. Among the top-ranked compounds, 59 fragments and 59 lead-like compounds were selected for experimental evaluation. Of these, four fragment- and seven lead-like compounds were confirmed to bind to the allosteric site with affinities ranging from 0.43 to 8.6 µM, corresponding to a hit rate of 9%. The four compounds with the highest affinities were demonstrated to be negative allosteric modulators of mGlu5 signaling in functional assays. The results demonstrate that virtual screens of fragment- and lead-like chemical libraries have complementary advantages and illustrate how access to high-resolution structures of GPCRs in complex with allosteric modulators can accelerate lead discovery.


Sujet(s)
Récepteur-5 métabotropique du glutamate , Bibliothèques de petites molécules , Récepteur-5 métabotropique du glutamate/métabolisme , Régulation allostérique , Simulation de docking moléculaire , Bibliothèques de petites molécules/pharmacologie , Ligands , Acide glutamique , Site allostérique , Récepteurs couplés aux protéines G
5.
Angew Chem Int Ed Engl ; 61(2): e202109967, 2022 01 10.
Article de Anglais | MEDLINE | ID: mdl-34668624

RÉSUMÉ

Sphingolipid metabolism is tightly controlled by enzymes to regulate essential processes in human physiology. The central metabolite is ceramide, a pro-apoptotic lipid catabolized by ceramidase enzymes to produce pro-proliferative sphingosine-1-phosphate. Alkaline ceramidases are transmembrane enzymes that recently attracted attention for drug development in fatty liver diseases. However, due to their hydrophobic nature, no specific small molecule inhibitors have been reported. We present the discovery and mechanism of action of the first drug-like inhibitors of alkaline ceramidase 3 (ACER3). In particular, we chemically engineered novel fluorescent ceramide substrates enabling screening of large compound libraries and characterized enzyme:inhibitor interactions using mass spectrometry and MD simulations. In addition to revealing a new paradigm for inhibition of lipid metabolising enzymes with non-lipidic small molecules, our data lay the ground for targeting ACER3 in drug discovery efforts.


Sujet(s)
Ceramidases
6.
Mol Cell ; 81(20): 4165-4175.e6, 2021 10 21.
Article de Anglais | MEDLINE | ID: mdl-34433090

RÉSUMÉ

GPCR functional selectivity opens new opportunities for the design of safer drugs. Ligands orchestrate GPCR signaling cascades by modulating the receptor conformational landscape. Our study provides insights into the dynamic mechanism enabling opioid ligands to preferentially activate the G protein over the ß-arrestin pathways through the µ-opioid receptor (µOR). We combine functional assays in living cells, solution NMR spectroscopy, and enhanced-sampling molecular dynamic simulations to identify the specific µOR conformations induced by G protein-biased agonists. In particular, we describe the dynamic and allosteric communications between the ligand-binding pocket and the receptor intracellular domains, through conserved motifs in class A GPCRs. Most strikingly, the biased agonists trigger µOR conformational changes in the intracellular loop 1 and helix 8 domains, which may impair ß-arrestin binding or signaling. The findings may apply to other GPCR families and provide key molecular information that could facilitate the design of biased ligands.


Sujet(s)
Analgésiques morphiniques/pharmacologie , Spectroscopie par résonance magnétique , Simulation de dynamique moléculaire , Transduction du signal/effets des médicaments et des substances chimiques , Analgésiques morphiniques/composition chimique , Animaux , Sites de fixation , Conception assistée par ordinateur , Conception de médicament , Agonisme partiel des médicaments , Cellules HEK293 , Humains , Ligands , Souris , Liaison aux protéines , Motifs et domaines d'intéraction protéique , Stabilité protéique , Récepteur mu/agonistes , Récepteur mu/génétique , Récepteur mu/métabolisme , Cellules Sf9 , Relation structure-activité , bêta-Arrestines/génétique , bêta-Arrestines/métabolisme
7.
Angew Chem Int Ed Engl ; 59(15): 5958-5964, 2020 04 06.
Article de Anglais | MEDLINE | ID: mdl-31808251

RÉSUMÉ

µ-Opioid receptors (µ-ORs) play a critical role in the modulation of pain and mediate the effects of the most powerful analgesic drugs. Despite extensive efforts, it remains insufficiently understood how µ-ORs produce specific effects in living cells. We developed new fluorescent ligands based on the µ-OR antagonist E-p-nitrocinnamoylamino-dihydrocodeinone (CACO), that display high affinity, long residence time and pronounced selectivity. Using these ligands, we achieved single-molecule imaging of µ-ORs on the surface of living cells at physiological expression levels. Our results reveal a high heterogeneity in the diffusion of µ-ORs, with a relevant immobile fraction. Using a pair of fluorescent ligands of different color, we provide evidence that µ-ORs interact with each other to form short-lived homodimers on the plasma membrane. This approach provides a new strategy to investigate µ-OR pharmacology at single-molecule level.


Sujet(s)
Colorants fluorescents/composition chimique , Hydrocodone/composition chimique , Multimérisation de protéines , Récepteur mu/composition chimique , Imagerie de molécules uniques/méthodes , Diffusion , Colorants fluorescents/pharmacologie , Hydrocodone/pharmacologie , Ligands , Structure quaternaire des protéines , Récepteur mu/antagonistes et inhibiteurs , Récepteur mu/métabolisme
9.
AIDS ; 31(18): 2443-2454, 2017 11 28.
Article de Anglais | MEDLINE | ID: mdl-28926402

RÉSUMÉ

OBJECTIVE: In this study, we looked for a new family of latency reversing agents. DESIGN: We searched for G-protein-coupled receptors (GPCR) coexpressed with the C-C chemokine receptor type 5 (CCR5) in primary CD4 T cells that activate infected cells and boost HIV production. METHODS: GPCR coexpression was unveiled by reverse transcriptase-PCR. We used fluorescence resonance energy transfer to analyze the dimerization with CCR5 of the expressed GPCR. Viral entry was measured by flow cytometry, reverse transcription by quantitative PCR, nuclear factor-kappa B translocation by immunofluorescence, long terminal repeat activation using a gene reporter assay and viral production by p24 quantification. RESULTS: Gαi-coupled sphingosine-1-phophate receptor 1 (S1P1) is highly coexpressed with CCR5 on primary CD4 T cells and dimerizes with it. The presence of S1P1 had major effects neither on viral entry nor on reverse transcription. Yet, S1P1 signaling induced NFκB activation, boosting the expression of the HIV LTR. Consequently, in culture medium containing sphingosine-1-phophate, the presence of S1P1 enhanced the replication of a CCR5-, but also of a CXCR4-using HIV-1 strain. The S1P1 ligand FTY720, a drug used in multiple sclerosis treatment, inhibited HIV-1 productive infection of monocyte-derived dendritic cells and of severe combined immunodeficiency mice engrafted with human peripheral blood mononuclear cells. Conversely, S1P1 agonists were able to force latently infected peripheral blood mononuclear cells and lymph node cells to produce virions in vitro. CONCLUSION: Altogether these data indicate that the presence of S1P1 facilitates HIV-1 replicative cycle by boosting viral genome transcription, S1P1 antagonists have anti-HIV effects and S1P1 agonists are HIV latency reversing agents.


Sujet(s)
Lymphocytes T CD4+/virologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Récepteurs aux lysosphingolipides/agonistes , Récepteurs aux lysosphingolipides/antagonistes et inhibiteurs , Transduction du signal , Latence virale/effets des médicaments et des substances chimiques , Réplication virale/effets des médicaments et des substances chimiques , Animaux , Cellules cultivées , Analyse de profil d'expression de gènes , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/croissance et développement , Humains , Souris SCID , Récepteurs CCR5/biosynthèse , Récepteurs aux lysosphingolipides/biosynthèse
10.
Elife ; 62017 06 29.
Article de Anglais | MEDLINE | ID: mdl-28661401

RÉSUMÉ

Metabotropic glutamate receptors (mGluRs) are mandatory dimers playing important roles in regulating CNS function. Although assumed to form exclusive homodimers, 16 possible heterodimeric mGluRs have been proposed but their existence in native cells remains elusive. Here, we set up two assays to specifically identify the pharmacological properties of rat mGlu heterodimers composed of mGlu2 and 4 subunits. We used either a heterodimer-specific conformational LRET-based biosensor or a system that guarantees the cell surface targeting of the heterodimer only. We identified mGlu2-4 specific pharmacological fingerprints that were also observed in a neuronal cell line and in lateral perforant path terminals naturally expressing mGlu2 and mGlu4. These results bring strong evidence for the existence of mGlu2-4 heterodimers in native cells. In addition to reporting a general approach to characterize heterodimeric mGluRs, our study opens new avenues to understanding the pathophysiological roles of mGlu heterodimers.


Sujet(s)
Composés bicycliques pontés/pharmacologie , Embryon de mammifère/métabolisme , Hippocampe/métabolisme , Neurones/métabolisme , Multimérisation de protéines/effets des médicaments et des substances chimiques , Récepteurs métabotropes au glutamate/composition chimique , Animaux , Cellules cultivées , Embryon de mammifère/cytologie , Embryon de mammifère/effets des médicaments et des substances chimiques , Agonistes des acides aminés excitateurs/pharmacologie , Cellules HEK293 , Hippocampe/cytologie , Hippocampe/effets des médicaments et des substances chimiques , Humains , Neurones/cytologie , Neurones/effets des médicaments et des substances chimiques , Rats , Rat Sprague-Dawley , Récepteurs métabotropes au glutamate/métabolisme
11.
Sci Rep ; 6: 30797, 2016 08 05.
Article de Anglais | MEDLINE | ID: mdl-27492592

RÉSUMÉ

If activation of recombinant G protein-coupled receptors in host cells (by drugs or other ligands) has predictive value, similar data must be obtained with native receptors naturally expressed in tissues. Using mouse and human recombinant κ opioid receptors transfected into a host cell, two selectively-acting compounds (ICI204448, asimadoline) equi-effectively activated both receptors, assessed by measuring two different cell signalling pathways which were equally affected without evidence of bias. In mouse intestine, naturally expressing κ receptors within its nervous system, both compounds also equi-effectively activated the receptor, inhibiting nerve-mediated muscle contraction. However, whereas ICI204448 acted similarly in human intestine, where κ receptors are again expressed within its nervous system, asimadoline was inhibitory only at very high concentrations; instead, low concentrations of asimadoline reduced the activity of ICI204448. This demonstration of species-dependence in activation of native, not recombinant κ receptors may be explained by different mouse/human receptor structures affecting receptor expression and/or interactions with intracellular signalling pathways in native environments, to reveal differences in intrinsic efficacy between receptor agonists. These results have profound implications in drug design for κ and perhaps other receptors, in terms of recombinant-to-native receptor translation, species-dependency and possibly, a need to use human, therapeutically-relevant, not surrogate tissues.


Sujet(s)
Muqueuse intestinale/métabolisme , Récepteur kappa/métabolisme , Protéines recombinantes/métabolisme , Acétamides/pharmacologie , Animaux , Conception de médicament , Cellules HEK293 , Humains , Souris , Pyrrolidines/pharmacologie , Transduction du signal , Spécificité d'espèce
12.
Article de Anglais | MEDLINE | ID: mdl-26617570

RÉSUMÉ

Although G protein-coupled receptor (GPCR) internalization has long been considered as a major aspect of the desensitization process that tunes ligand responsiveness, internalization is also involved in receptor resensitization and signaling, as well as the ligand scavenging function of some atypical receptors. Internalization thus contributes to the diversity of GPCR-dependent signaling, and its dynamics and quantification in living cells has generated considerable interest. We developed a robust and sensitive assay to follow and quantify ligand-induced and constitutive-induced GPCR internalization but also receptor recycling in living cells. This assay is based on diffusion-enhanced resonance energy transfer (DERET) between cell surface GPCRs labeled with a luminescent terbium cryptate donor and a fluorescein acceptor present in the culture medium. GPCR internalization results in a quantifiable reduction of energy transfer. This method yields a high signal-to-noise ratio due to time-resolved measurements. For various GPCRs belonging to different classes, we demonstrated that constitutive and ligand-induced internalization could be monitored as a function of time and ligand concentration, thus allowing accurate quantitative determination of kinetics of receptor internalization but also half-maximal effective or inhibitory concentrations of compounds. In addition to its selectivity and sensitivity, we provided evidence that DERET-based internalization assay is particularly suitable for characterizing biased ligands. Furthermore, the determination of a Z'-factor value of 0.45 indicates the quality and suitability of DERET-based internalization assay for high-throughput screening (HTS) of compounds that may modulate GPCRs internalization.

13.
J Am Chem Soc ; 134(46): 19026-34, 2012 Nov 21.
Article de Anglais | MEDLINE | ID: mdl-23095089

RÉSUMÉ

While γ-aminobutyric acid (GABA) is the main inhibitory neurotransmitter, suitable tools to measure its concentration in living cells with high spatiotemporal resolution are missing. Herein, we describe the first ratiometric fluorescent sensor for GABA, dubbed GABA-Snifit, which senses GABA with high specificity and spatiotemporal resolution on the surface of living mammalian cells. GABA-Snifit is a semisynthetic fusion protein containing the GABA(B) receptor, SNAP- and CLIP-tag, a synthetic fluorophore and a fluorescent GABA(B) receptor antagonist. When assembled on cell surfaces, GABA-Snifit displays a GABA-dependent fluorescence emission spectrum in the range of 500-700 nm that permits sensing micromolar to millimolar GABA concentrations. The ratiometric change of the sensor on living cells is 1.8. Furthermore, GABA-Snifit can be utilized to quantify the relative binding affinities of GABA(B) receptor agonists, antagonists and the effect of allosteric modulators. These properties make GABA-Snifit a valuable tool to investigate the role of GABA and GABA(B) in biological systems.


Sujet(s)
Colorants fluorescents/composition chimique , Récepteurs GABA-B/composition chimique , Acide gamma-amino-butyrique/composition chimique , Régulation allostérique , Électrophorèse sur gel de polyacrylamide , Transfert d'énergie par résonance de fluorescence , Cellules HEK293 , Humains , Ligands
14.
Article de Anglais | MEDLINE | ID: mdl-22837753

RÉSUMÉ

The concept of oligomerization of G protein-coupled receptor (GPCR) opens new perspectives regarding physiological function regulation. The capacity of one GPCR to modify its binding and coupling properties by interacting with a second one can be at the origin of regulations unsuspected two decades ago. Although the concept is interesting, its validation at a physiological level is challenging and probably explains why receptor oligomerization is still controversial. Demonstrating direct interactions between two proteins is not trivial since few techniques present a spatial resolution allowing this precision. Resonance energy transfer (RET) strategies are actually the most convenient ones. During the last two decades, bioluminescent resonance energy transfer and time-resolved fluorescence resonance energy transfer (TR-FRET) have been widely used since they exhibit high signal-to-noise ratio. Most of the experiments based on GPCR labeling have been performed in cell lines and it has been shown that all GPCRs have the propensity to form homo- or hetero-oligomers. However, whether these data can be extrapolated to GPCRs expressed in native tissues and explain receptor functioning in real life, remains an open question. Native tissues impose different constraints since GPCR sequences cannot be modified. Recently, a fluorescent ligand-based GPCR labeling strategy combined to a TR-FRET approach has been successfully used to prove the existence of GPCR oligomerization in native tissues. Although the RET-based strategies are generally quite simple to implement, precautions have to be taken before concluding to the absence or the existence of specific interactions between receptors. For example, one should exclude the possibility of collision of receptors diffusing throughout the membrane leading to a specific FRET signal. The advantages and the limits of different approaches will be reviewed and the consequent perspectives discussed.

15.
ACS Chem Biol ; 7(2): 289-93, 2012 Feb 17.
Article de Anglais | MEDLINE | ID: mdl-22026407

RÉSUMÉ

A caged rhodamine 110 derivative for the specific labeling of SNAP-tag fusion proteins is introduced. The caged rhodamine 110 derivative permits the labeling of cell surface proteins in living cells and of intracellular proteins in fixed cells. The probe requires only a single caging group to maintain the fluorophore in a non-fluorescent state and becomes highly fluorescent after uncaging. The high contrast ratio is confirmed both in bulk and at the single molecule level. This property, together with its high photon yield makes it an excellent dye for photoactivated localization microscopy (PALM), as we demonstrate here.


Sujet(s)
Colorants fluorescents/analyse , Protéines/analyse , Rhodamines/analyse , Lignée cellulaire tumorale , Cellules HEK293 , Humains , Microscopie de fluorescence , Protéines de fusion recombinantes/analyse
16.
Methods Mol Biol ; 756: 201-14, 2011.
Article de Anglais | MEDLINE | ID: mdl-21870227

RÉSUMÉ

G protein-coupled receptors (GPCRs) are key players in cell-cell communication, the dysregulation of which has often deleterious effects leading to pathologies such as psychiatric and neurological diseases. Consequently, GPCRs represent excellent drug targets, and as such are the object of intense research in drug discovery for therapeutic application. Recently, the GPCR field has been revolutionized by the demonstration that GPCRs are part of large protein complexes that control their pharmacology, activity, and signaling. Moreover, in these complexes, one GPCR can either associate with itself, forming homodimers or homooligomers, or with other receptor types, forming heterodimeric or heterooligomeric receptor entities that display new receptor features. These features include alterations in ligand cooperativity and selectivity, the activation of novel signaling pathways, and novel processes of desensitization. Thus, it has become necessary to identify GPCR-associated protein complexes of interest at the cell surface, and to determine the state of oligomerization of these receptors and their interactions with their partner proteins. This is essential to understand the function of GPCRs in their native environment, as well as ways to either modulate or control receptor activity with appropriate pharmacological tools, and to develop new therapeutic strategies. This requires the development of technologies to precisely address protein-protein interactions between oligomers at the cell surface. In collaboration with Cisbio Bioassay, we have developed such a technology, which combines TR-FRET detection with a new labeling method called SnapTag. This technology has allowed us to address the oligomeric state of many GPCRs.


Sujet(s)
Transfert d'énergie par résonance de fluorescence/méthodes , Cartographie d'interactions entre protéines/méthodes , Récepteurs couplés aux protéines G/métabolisme , Animaux , Lignée cellulaire , Humains , Multimérisation de protéines , Récepteurs couplés aux protéines G/composition chimique , Récepteurs GABA-B/composition chimique , Récepteurs GABA-B/métabolisme
17.
Chembiochem ; 12(14): 2217-26, 2011 Sep 19.
Article de Anglais | MEDLINE | ID: mdl-21793150

RÉSUMÉ

The ability to specifically attach chemical probes to individual proteins represents a powerful approach to the study and manipulation of protein function in living cells. It provides a simple, robust and versatile approach to the imaging of fusion proteins in a wide range of experimental settings. However, a potential drawback of detection using chemical probes is the fluorescence background from unreacted or nonspecifically bound probes. In this report we present the design and application of novel fluorogenic probes for labeling SNAP-tag fusion proteins in living cells. SNAP-tag is an engineered variant of the human repair protein O(6)-alkylguanine-DNA alkyltransferase (hAGT) that covalently reacts with benzylguanine derivatives. Reporter groups attached to the benzyl moiety become covalently attached to the SNAP tag while the guanine acts as a leaving group. Incorporation of a quencher on the guanine group ensures that the benzylguanine probe becomes highly fluorescent only upon labeling of the SNAP-tag protein. We describe the use of intramolecularly quenched probes for wash-free labeling of cell surface-localized epidermal growth factor receptor (EGFR) fused to SNAP-tag and for direct quantification of SNAP-tagged ß-tubulin in cell lysates. In addition, we have characterized a fast-labeling variant of SNAP-tag, termed SNAP(f), which displays up to a tenfold increase in its reactivity towards benzylguanine substrates. The presented data demonstrate that the combination of SNAP(f) and the fluorogenic substrates greatly reduces the background fluorescence for labeling and imaging applications. This approach enables highly sensitive spatiotemporal investigation of protein dynamics in living cells.


Sujet(s)
Conception de médicament , Colorants fluorescents/composition chimique , Colorants fluorescents/synthèse chimique , Imagerie moléculaire/méthodes , O(6)-methylguanine-DNA methyltransferase/composition chimique , Protéines de fusion recombinantes/composition chimique , Extrait cellulaire , Membrane cellulaire/métabolisme , Survie cellulaire , Récepteurs ErbB/métabolisme , Colorants fluorescents/métabolisme , Guanidine/composition chimique , Cellules HEK293 , Humains , Cinétique , O(6)-methylguanine-DNA methyltransferase/génétique , O(6)-methylguanine-DNA methyltransferase/métabolisme , Transport des protéines , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme
18.
EMBO J ; 30(12): 2336-49, 2011 May 06.
Article de Anglais | MEDLINE | ID: mdl-21552208

RÉSUMÉ

G protein-coupled receptors (GPCRs) have key roles in cell-cell communication. Recent data suggest that these receptors can form large complexes, a possibility expected to expand the complexity of this regulatory system. Among the brain GPCRs, the heterodimeric GABA(B) receptor is one of the most abundant, being distributed in most brain regions, on either pre- or post-synaptic elements. Here, using specific antibodies labelled with time-resolved FRET compatible fluorophores, we provide evidence that the heterodimeric GABA(B) receptor can form higher-ordered oligomers in the brain, as suggested by the close proximity of the GABA(B1) subunits. Destabilizing the oligomers using a competitor or a GABA(B1) mutant revealed different G protein coupling efficiencies depending on the oligomeric state of the receptor. By examining, in heterologous system, the G protein coupling properties of such GABA(B) receptor oligomers composed of a wild-type and a non-functional mutant heterodimer, we provide evidence for a negative functional cooperativity between the GABA(B) heterodimers.


Sujet(s)
Récepteurs GABA-B/composition chimique , Transduction du signal/physiologie , Régulation allostérique/génétique , Animaux , Cellules COS , Chlorocebus aethiops , Cellules HEK293 , Humains , Souris , Souris de lignée BALB C , Souris knockout , Mutagenèse dirigée , Isoformes de protéines/composition chimique , Isoformes de protéines/déficit , Isoformes de protéines/génétique , Multimérisation de protéines/génétique , Stabilité protéique , Récepteurs GABA-B/déficit , Récepteurs GABA-B/génétique , Transduction du signal/génétique
19.
Chimia (Aarau) ; 65(11): 868-71, 2011.
Article de Anglais | MEDLINE | ID: mdl-22289374

RÉSUMÉ

The development of molecular probes to visualize cellular processes is an important challenge in chemical biology. One possibility to create such cellular indicators is based on the selective labeling of proteins with synthetic probes in living cells. Over the last years, our laboratory has developed different labeling approaches for monitoring protein activity and for localizing synthetic probes inside living cells. In this article, we review two of these labeling approaches, the SNAP-tag and CLIP-tag technologies, and their use for studying cellular processes.


Sujet(s)
Protéines/métabolisme , Calcium/métabolisme , Colorants fluorescents/métabolisme
20.
ACS Chem Biol ; 5(5): 507-16, 2010 May 21.
Article de Anglais | MEDLINE | ID: mdl-20218675

RÉSUMÉ

Photosensitive probes are powerful tools to study cellular processes with high temporal and spatial resolution. However, most synthetic fluorophores suited for biomolecular imaging have not been converted yet to appropriate photosensitive analogues. Here we describe a generally applicable strategy for the generation of photoactivatable and photoconvertible fluorescent probes that can be selectively coupled to SNAP-tag fusion proteins in living cells. Photoactivatable versions of fluorescein and Cy3 as well as a photoconvertible Cy5-Cy3 probe were prepared and coupled to selected proteins on the cell surface, in the cytosol, and in the nucleus of cells. In proof-of-principle experiments, the photoactivatable Cy3 probe was used to characterize the mobility of a lipid-anchored cell surface protein and of a G protein coupled receptor (GPCR). This work establishes a generally applicable strategy for the generation of a large variety of different photosensitive fluorophores with tailor-made properties for biomolecular imaging.


Sujet(s)
Colorants fluorescents , Photochimie , Protéines de fusion recombinantes , Spectrométrie de fluorescence/méthodes , Carbocyanines/synthèse chimique , Carbocyanines/composition chimique , Membrane cellulaire/métabolisme , Fluorescéine/synthèse chimique , Fluorescéine/composition chimique , Colorants fluorescents/synthèse chimique , Colorants fluorescents/composition chimique , Microscopie confocale , Microscopie de fluorescence , Récepteurs couplés aux protéines G/métabolisme , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE