Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Med Chem ; 67(14): 12099-12117, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-38994645

RESUMEN

The study of protein function and dynamics in their native cellular environment is essential for progressing fundamental science. To overcome the requirement of genetic modification of the protein or the limitations of dissociable fluorescent ligands, ligand-directed (LD) chemistry has most recently emerged as a complementary, bioorthogonal approach for labeling native proteins. Here, we describe the rational design, development, and application of the first ligand-directed chemistry approach for labeling the A1AR in living cells. We pharmacologically demonstrate covalent labeling of A1AR expressed in living cells while the orthosteric binding site remains available. The probes were imaged using confocal microscopy and fluorescence correlation spectroscopy to study A1AR localization and dynamics in living cells. Additionally, the probes allowed visualization of the specific localization of A1ARs endogenously expressed in dorsal root ganglion (DRG) neurons. LD probes developed here hold promise for illuminating ligand-binding, receptor signaling, and trafficking of the A1AR in more physiologically relevant environments.


Asunto(s)
Colorantes Fluorescentes , Receptor de Adenosina A1 , Ligandos , Receptor de Adenosina A1/metabolismo , Receptor de Adenosina A1/química , Humanos , Colorantes Fluorescentes/química , Animales , Ganglios Espinales/metabolismo , Ganglios Espinales/citología , Células HEK293 , Neuronas/metabolismo
2.
Arch Biochem Biophys ; 753: 109915, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38307314

RESUMEN

The human ATP-binding cassette (ABC) transporter, ABCG2, is responsible for multidrug resistance in some tumours. Detailed knowledge of its activity is crucial for understanding drug transport and resistance in cancer, and has implications for wider pharmacokinetics. The binding of substrates and inhibitors is a key stage in the transport cycle of ABCG2. Here, we describe a novel binding assay using a high affinity fluorescent inhibitor based on Ko143 and time-resolved Förster resonance energy transfer (TR-FRET) to measure saturation binding to ABCG2. This binding is displaced by Ko143 and other known ABCG2 ligands, and is sensitive to the addition of AMP-PNP, a non-hydrolysable ATP analogue. This assay complements the arsenal of methods for determining drug:ABCG2 interactions and has the possibility of being adaptable for other multidrug pumps.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia , Neoplasias , Humanos , Resistencia a Antineoplásicos , Transportadoras de Casetes de Unión a ATP/metabolismo , Resistencia a Múltiples Medicamentos , Adenosina Trifosfato , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Proteínas de Neoplasias/metabolismo
3.
ACS Med Chem Lett ; 15(1): 143-148, 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38229752

RESUMEN

The atypical chemokine receptor 3 (ACKR3) is a receptor that induces cancer progression and metastasis in multiple cell types. Therefore, new chemical tools are required to study the role of ACKR3 in cancer and other diseases. In this study, fluorescent probes, based on a series of small molecule ACKR3 agonists, were synthesized. Three fluorescent probes, which showed specific binding to ACKR3 through a luminescence-based NanoBRET binding assay (pKd ranging from 6.8 to 7.8) are disclosed. Due to their high affinity at the ACKR3, we have shown their application in both competition binding experiments and confocal microscopy studies showing the cellular distribution of this receptor.

4.
J Med Chem ; 66(7): 5208-5222, 2023 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-36944083

RESUMEN

The C-X-C chemokine receptor type 4, or CXCR4, is a chemokine receptor found to promote cancer progression and metastasis of various cancer cell types. To investigate the pharmacology of this receptor, and to further elucidate its role in cancer, novel chemical tools are a necessity. In the present study, using classic medicinal chemistry approaches, small-molecule-based fluorescent probes were designed and synthesized based on previously reported small-molecule antagonists. Here, we report the development of three distinct chemical classes of fluorescent probes that show specific binding to the CXCR4 receptor in a novel fluorescence-based NanoBRET binding assay (pKD ranging 6.6-7.1). Due to their retained affinity at CXCR4, we furthermore report their use in competition binding experiments and confocal microscopy to investigate the pharmacology and cellular distribution of this receptor.


Asunto(s)
Colorantes Fluorescentes , Receptores CXCR4 , Receptores CXCR4/metabolismo , Ligandos , Colorantes Fluorescentes/química , Unión Proteica , Quimiocinas/metabolismo , Quimiocina CXCL12/metabolismo
5.
FASEB J ; 35(4): e21211, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33710641

RESUMEN

Organization of G protein-coupled receptors at the plasma membrane has been the focus of much recent attention. Advanced microscopy techniques have shown that these receptors can be localized to discrete microdomains and reorganization upon ligand activation is crucial in orchestrating their signaling. Here, we have compared the membrane organization and downstream signaling of a mutant (R108A, R3.50A) of the adenosine A3 receptor (A3 AR) to that of the wild-type receptor. Fluorescence Correlation Spectroscopy (FCS) studies with a fluorescent agonist (ABEA-X-BY630) demonstrated that both wild-type and mutant receptors bind agonist with high affinity but in subsequent downstream signaling assays the R108A mutation abolished agonist-mediated inhibition of cAMP production and ERK phosphorylation. In further FCS studies, both A3 AR and A3 AR R108A underwent similar agonist-induced increases in receptor density and molecular brightness which were accompanied by a decrease in membrane diffusion after agonist treatment. Using bimolecular fluorescence complementation, experiments showed that the R108A mutant retained the ability to recruit ß-arrestin and these receptor/arrestin complexes displayed similar membrane diffusion and organization to that observed with wild-type receptors. These data demonstrate that effective G protein signaling is not a prerequisite for agonist-stimulated ß-arrestin recruitment and membrane reorganization of the A3 AR.


Asunto(s)
Agonistas del Receptor de Adenosina A3/farmacología , Adenosina/análogos & derivados , Compuestos de Boro/farmacología , Proteínas de Unión al GTP/metabolismo , Receptor de Adenosina A3/metabolismo , Adenosina/farmacología , Animales , Arrestina/metabolismo , Células CHO , Cricetulus , Regulación de la Expresión Génica/efectos de los fármacos , Mutación , Unión Proteica , Receptor de Adenosina A3/genética
6.
Commun Biol ; 3(1): 722, 2020 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-33247190

RESUMEN

To study the localisation of G protein-coupled receptors (GPCR) in their native cellular environment requires their visualisation through fluorescent labelling. To overcome the requirement for genetic modification of the receptor or the limitations of dissociable fluorescent ligands, here we describe rational design of a compound that covalently and selectively labels a GPCR in living cells with a fluorescent moiety. We designed a fluorescent antagonist, in which the linker incorporated between pharmacophore (ZM241385) and fluorophore (sulfo-cyanine5) is able to facilitate covalent linking of the fluorophore to the adenosine A2A receptor. We pharmacologically and biochemically demonstrate irreversible fluorescent labelling without impeding access to the orthosteric binding site and demonstrate its use in endogenously expressing systems. This offers a non-invasive and selective approach to study function and localisation of native GPCRs.


Asunto(s)
Colorantes Fluorescentes , Receptores Acoplados a Proteínas G/metabolismo , Triazinas , Triazoles , Marcadores de Afinidad , Diseño de Fármacos , Células HEK293 , Humanos , Ligandos , Receptor de Adenosina A2A/metabolismo
7.
Nanoscale ; 12(21): 11518-11525, 2020 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-32428052

RESUMEN

The fundamental importance of membrane proteins in cellular processes has driven a marked increase in the use of membrane mimetic approaches for studying and exploiting these proteins. Nano-encapsulation strategies which preserve the native lipid bilayer environment are particularly attractive. Consequently, the use of poly(styrene co-maleic acid) (SMA) has been widely adopted to solubilise proteins directly from cell membranes by spontaneously forming "SMA Lipid Particles" (SMALPs). G-protein-coupled receptors (GPCRs) are ubiquitous "chemical switches", are central to cell signalling throughout the evolutionary tree, form the largest family of membrane proteins in humans and are a major drug discovery target. GPCR-SMALPs that retain binding capability would be a versatile platform for a wide range of down-stream applications. Here, using the adenosine A2A receptor (A2AR) as an archetypical GPCR, we show for the first time the utility of fluorescence correlation spectroscopy (FCS) to characterise the binding capability of GPCRs following nano-encapsulation. Unbound fluorescent ligand CA200645 exhibited a monophasic autocorrelation curve (dwell time, τD = 68 ± 2 µs; diffusion coefficient, D = 287 ± 15 µm2 s-1). In the presence of A2AR-SMALP, bound ligand was also evident (τD = 625 ± 23 µs; D = 30 ± 4 µm2 s-1). Using a non-receptor control (ZipA-SMALP) plus competition binding confirmed that this slower component represented binding to the encapsulated A2AR. Consequently, the combination of GPCR-SMALP and FCS is an effective platform for the quantitative real-time characterisation of nano-encapsulated receptors, with single molecule sensitivity, that will have widespread utility for future exploitation of GPCR-SMALPs in general.


Asunto(s)
Ligandos , Maleatos/química , Receptores Acoplados a Proteínas G/metabolismo , Estireno/química , Materiales Biomiméticos , Fluorescencia , Humanos , Lípidos de la Membrana/química , Lípidos de la Membrana/metabolismo , Unión Proteica , Receptor de Adenosina A2A/química , Receptor de Adenosina A2A/metabolismo , Receptores Acoplados a Proteínas G/química , Imagen Individual de Molécula , Espectrometría de Fluorescencia
8.
J Biol Chem ; 295(36): 12822-12839, 2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32111735

RESUMEN

A disintegrin and metalloprotease 10 (ADAM10) is a transmembrane protein essential for embryonic development, and its dysregulation underlies disorders such as cancer, Alzheimer's disease, and inflammation. ADAM10 is a "molecular scissor" that proteolytically cleaves the extracellular region from >100 substrates, including Notch, amyloid precursor protein, cadherins, growth factors, and chemokines. ADAM10 has been recently proposed to function as six distinct scissors with different substrates, depending on its association with one of six regulatory tetraspanins, termed TspanC8s. However, it remains unclear to what degree ADAM10 function critically depends on a TspanC8 partner, and a lack of monoclonal antibodies specific for most TspanC8s has hindered investigation of this question. To address this knowledge gap, here we designed an immunogen to generate the first monoclonal antibodies targeting Tspan15, a model TspanC8. The immunogen was created in an ADAM10-knockout mouse cell line stably overexpressing human Tspan15, because we hypothesized that expression in this cell line would expose epitopes that are normally blocked by ADAM10. Following immunization of mice, this immunogen strategy generated four Tspan15 antibodies. Using these antibodies, we show that endogenous Tspan15 and ADAM10 co-localize on the cell surface, that ADAM10 is the principal Tspan15-interacting protein, that endogenous Tspan15 expression requires ADAM10 in cell lines and primary cells, and that a synthetic ADAM10/Tspan15 fusion protein is a functional scissor. Furthermore, two of the four antibodies impaired ADAM10/Tspan15 activity. These findings suggest that Tspan15 directly interacts with ADAM10 in a functional scissor complex.


Asunto(s)
Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Proteínas de la Membrana/metabolismo , Complejos Multiproteicos/metabolismo , Tetraspaninas/metabolismo , Células A549 , Proteína ADAM10/genética , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Células HEK293 , Humanos , Células Jurkat , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Complejos Multiproteicos/genética , Tetraspaninas/genética
9.
SLAS Discov ; 25(2): 186-194, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31583945

RESUMEN

Receptor internalization in response to prolonged agonist treatment is an important regulator of G protein-coupled receptor (GPCR) function. The adenosine A1 receptor (A1AR) is one of the adenosine receptor family of GPCRs, and evidence for its agonist-induced internalization is equivocal. The recently developed NanoBiT technology uses split NanoLuc Luciferase to monitor changes in protein interactions. We have modified the human A1AR on the N-terminus with the small high-affinity HiBiT tag. In the presence of the large NanoLuc subunit (LgBiT), complementation occurs, reconstituting a full-length functional NanoLuc Luciferase. Here, we have used complemented luminescence to monitor the internalization of the A1AR in living HEK293 cells. Agonist treatment resulted in a robust decrease in cell-surface luminescence, indicating an increase in A1AR internalization. These responses were inhibited by the A1AR-selective antagonist 1,3-dipropyl-8-cyclopentylxanthine (DPCPX), with an antagonist affinity that closely matched that measured using ligand binding with a fluorescent A1 receptor antagonist (CA200645). The agonist potencies for inducing A1AR internalization were very similar to the affinities previously determined by ligand binding, suggesting little or no amplification of the internalization response. By complementing the HiBiT tag to exogenous purified LgBiT, it was also possible to perform NanoBRET ligand-binding experiments using HiBiT-A1AR. This study demonstrates the use of NanoBiT technology to monitor internalization of the A1AR and offers the potential to combine these experiments with NanoBRET ligand-binding assays.


Asunto(s)
Adenosina/genética , Receptor de Adenosina A1/genética , Receptores Acoplados a Proteínas G/genética , Adenosina/química , Agonistas del Receptor de Adenosina A1/farmacología , Células HEK293 , Humanos , Cinética , Ligandos , Mapas de Interacción de Proteínas/efectos de los fármacos , Mapas de Interacción de Proteínas/genética , Receptor de Adenosina A1/efectos de los fármacos , Receptores Acoplados a Proteínas G/agonistas , Xantinas/farmacología
10.
Mol Pharmacol ; 96(6): 778-793, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31092552

RESUMEN

G protein-coupled receptors (GPCRs) are regulated by complex molecular mechanisms, both in physiologic and pathologic conditions, and their signaling can be intricate. Many factors influence their signaling behavior, including the type of ligand that activates the GPCR, the presence of interacting partners, the kinetics involved, or their location. The two CXC-type chemokine receptors, CXC chemokine receptor 4 (CXCR4) and atypical chemokine receptor 3 (ACKR3), both members of the GPCR superfamily, are important and established therapeutic targets in relation to cancer, human immunodeficiency virus infection, and inflammatory diseases. Therefore, it is crucial to understand how the signaling of these receptors works to be able to specifically target them. In this review, we discuss how the signaling pathways activated by CXCR4 and ACKR3 can vary in different situations. G protein signaling of CXCR4 depends on the cellular context, and discrepancies exist depending on the cell lines used. ACKR3, as an atypical chemokine receptor, is generally reported to not activate G proteins but can broaden its signaling spectrum upon heteromerization with other receptors, such as CXCR4, endothelial growth factor receptor, or the α 1-adrenergic receptor (α 1-AR). Also, CXCR4 forms heteromers with CC chemokine receptor (CCR) 2, CCR5, the Na+/H+ exchanger regulatory factor 1, CXCR3, α 1-AR, and the opioid receptors, which results in differential signaling from that of the monomeric subunits. In addition, CXCR4 is present on membrane rafts but can go into the nucleus during cancer progression, probably acquiring different signaling properties. In this review, we also provide an overview of the currently known critical amino acids involved in CXCR4 and ACKR3 signaling.


Asunto(s)
Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Receptores CXCR/genética , Receptores CXCR/metabolismo , Transducción de Señal/fisiología , Secuencia de Aminoácidos , Animales , Humanos
11.
Purinergic Signal ; 15(2): 139-153, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30919204

RESUMEN

There is a growing interest in understanding the binding kinetics of compounds that bind to G protein-coupled receptors prior to progressing a lead compound into clinical trials. The widely expressed adenosine A3 receptor (A3AR) has been implicated in a range of diseases including immune conditions, and compounds that aim to selectively target this receptor are currently under development for arthritis. Kinetic studies at the A3AR have been performed using a radiolabelled antagonist, but due to the kinetics of this probe, they have been carried out at 10 °C in membrane preparations. In this study, we have developed a live cell NanoBRET ligand binding assay using fluorescent A3AR antagonists to measure kinetic parameters of labelled and unlabelled compounds at the A3AR at physiological temperatures. The kinetic profiles of four fluorescent antagonists were determined in kinetic association assays, and it was found that XAC-ser-tyr-X-BY630 had the longest residence time (RT = 288 ± 62 min) at the A3AR. The association and dissociation rate constants of three antagonists PSB-11, compound 5, and LUF7565 were also determined using two fluorescent ligands (XAC-ser-tyr-X-BY630 or AV039, RT = 6.8 ± 0.8 min) as the labelled probe and compared to those obtained using a radiolabelled antagonist ([3H]PSB-11, RT = 44.6 ± 3.9 min). There was close agreement in the kinetic parameters measured with AV039 and [3H]PSB-11 but significant differences to those obtained using XAC-S-ser-S-tyr-X-BY630. These data indicate that selecting a probe with the appropriate kinetics is important to accurately determine the kinetics of unlabelled ligands with markedly different kinetic profiles.


Asunto(s)
Antagonistas del Receptor de Adenosina A3/farmacocinética , Transferencia Resonante de Energía de Fluorescencia/métodos , Mediciones Luminiscentes , Receptor de Adenosina A3/metabolismo , Células HEK293 , Humanos , Cinética
12.
Biochem Pharmacol ; 136: 62-75, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28392095

RESUMEN

Vascular endothelial growth factor (VEGF) is an important mediator of angiogenesis. Here we have used a novel stoichiometric protein-labeling method to generate a fluorescent variant of VEGF (VEGF165a-TMR) labeled on a single cysteine within each protomer of the antiparallel VEGF homodimer. VEGF165a-TMR has then been used in conjunction with full length VEGFR2, tagged with the bioluminescent protein NanoLuc, to undertake a real time quantitative evaluation of VEGFR2 binding characteristics in living cells using bioluminescence resonance energy transfer (BRET). This provided quantitative information on VEGF-VEGFR2 interactions. At longer incubation times, VEGFR2 is internalized by VEGF165a-TMR into intracellular endosomes. This internalization can be prevented by the receptor tyrosine kinase inhibitors (RTKIs) cediranib, sorafenib, pazopanib or vandetanib. In the absence of RTKIs, the BRET signal is decreased over time as a consequence of the dissociation of agonist from the receptor in intracellular endosomes and recycling of VEGFR2 back to the plasma membrane.


Asunto(s)
Sistemas de Computación , Endocitosis/fisiología , Colorantes Fluorescentes/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Secuencia de Aminoácidos , Relación Dosis-Respuesta a Droga , Endocitosis/efectos de los fármacos , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
13.
Front Pharmacol ; 8: 908, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29321740

RESUMEN

Recent advances in fluorescent ligand technology have enabled the study of G protein-coupled receptors in their native environment without the need for genetic modification such as addition of N-terminal fluorescent or bioluminescent tags. Here, we have used a non-imaging plate reader (PHERAstar FS) to monitor the binding of fluorescent ligands to the human adenosine-A3 receptor (A3AR; CA200645 and AV039), stably expressed in CHO-K1 cells. To verify that this method was suitable for the study of other GPCRs, assays at the human adenosine-A1 receptor, and ß1 and ß2 adrenoceptors (ß1AR and ß2AR; BODIPY-TMR-CGP-12177) were also carried out. Affinity values determined for the binding of the fluorescent ligands CA200645 and AV039 to A3AR for a range of classical adenosine receptor antagonists were consistent with A3AR pharmacology and correlated well (R2 = 0.94) with equivalent data obtained using a confocal imaging plate reader (ImageXpress Ultra). The binding of BODIPY-TMR-CGP-12177 to the ß1AR was potently inhibited by low concentrations of the ß1-selective antagonist CGP 20712A (pKi 9.68) but not by the ß2-selective antagonist ICI 118551(pKi 7.40). Furthermore, in experiments conducted in CHO K1 cells expressing the ß2AR this affinity order was reversed with ICI 118551 showing the highest affinity (pKi 8.73) and CGP20712A (pKi 5.68) the lowest affinity. To determine whether the faster data acquisition of the non-imaging plate reader (~3 min per 96-well plate) was suitable for high throughput screening (HTS), we screened the LOPAC library for inhibitors of the binding of CA200645 to the A3AR. From the initial 1,263 compounds evaluated, 67 hits (defined as those that inhibited the total binding of 25 nM CA200645 by ≥40%) were identified. All compounds within the library that had medium to high affinity for the A3AR (pKi ≥6) were successfully identified. We found three novel compounds in the library that displayed unexpected sub-micromolar affinity for the A3AR. These were K114 (pKi 6.43), retinoic acid p-hydroxyanilide (pKi 6.13) and SU 6556 (pKi 6.17). Molecular docking of these latter three LOPAC library members provided a plausible set of binding poses within the vicinity of the established orthosteric A3AR binding pocket. A plate reader based library screening using an untagged receptor is therefore possible using fluorescent ligand opening the possibility of its use in compound screening at natively expressed receptors.

14.
Biochim Biophys Acta ; 1863(1): 19-29, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26453803

RESUMEN

ABCG2 is one of three human ATP binding cassette (ABC) transporters involved in the export from cells of a chemically and structurally diverse range of compounds. This multidrug efflux capability, together with a broad tissue distribution in the body, means that ABCG2 exerts a range of effects on normal physiology such as kidney urate transport, as well as contributing towards the pharmacokinetic profiles of many exogenous drugs. The primary sequence of ABCG2 contains only half the number of domains required for a functioning ABC transporter and so it must oligomerise in order to function, yet its oligomeric state in intact cell membranes remains uncharacterized. We have analysed ABCG2 in living cell membranes using a combination of fluorescence correlation spectroscopy, photon counting histogram analysis, and stepwise photobleaching to demonstrate a predominantly tetrameric structure for ABCG2 in the presence or absence of transport substrates. These results provide the essential basis for exploring pharmacological manipulation of oligomeric state as a strategy to modulate ABCG2 activity in future selective therapeutics.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Membrana Celular/metabolismo , Riñón/metabolismo , Imagen Molecular , Proteínas de Neoplasias/metabolismo , Multimerización de Proteína/fisiología , Ácido Úrico/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Transporte Biológico Activo/fisiología , Membrana Celular/genética , Células HEK293 , Humanos , Riñón/citología , Proteínas de Neoplasias/genética , Estructura Cuaternaria de Proteína
15.
Neuropharmacology ; 98: 68-77, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25937210

RESUMEN

Fluorescence based probes provide a novel way to study the dynamic internalization process of G protein-coupled receptors (GPCRs). Recent advances in the rational design of fluorescent ligands for GPCRs have been used here to generate new fluorescent agonists containing tripeptide linkers for the adenosine A3 receptor. The fluorescent agonist BY630-X-(D)-A-(D)-A-G-ABEA was found to be a highly potent agonist at the adenosine A3 receptor in both reporter gene (pEC50 = 8.48 ± 0.09) and internalization assays (pEC50 = 7.47 ± 0.11). Confocal imaging studies showed that BY630-X-(D)-A-(D)-A-G-ABEA was internalized with A3 linked to yellow fluorescent protein, which was blocked by the competitive antagonist MRS1220. Internalization of untagged adenosine A3 could also be visualized with BY630-X-(D)-A-(D)-A-G-ABEA treatment. Further, BY630-X-(D)-A-(D)-A-G-ABEA stimulated the formation of receptor-arrestin3 complexes and was found to localize with these intracellular complexes. This highly potent agonist with excellent imaging properties should be a valuable tool to study receptor internalization. This article is part of the Special Issue entitled 'Fluorescent Tools in Neuropharmacology'.


Asunto(s)
Arrestinas/metabolismo , Colorantes Fluorescentes/química , Receptor de Adenosina A3/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacología , Agonistas del Receptor de Adenosina A3/farmacología , Adenosina-5'-(N-etilcarboxamida)/farmacología , Animales , Arrestinas/genética , Compuestos de Boro/farmacología , Células CHO , Colforsina/farmacología , Cricetulus , Relación Dosis-Respuesta a Droga , Colorantes Fluorescentes/farmacocinética , Expresión Génica/efectos de los fármacos , Ligandos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía Confocal , Estructura Molecular , Transporte de Proteínas/fisiología , Receptor de Adenosina A3/genética , Factores de Tiempo , Transfección
16.
FASEB J ; 28(10): 4211-22, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24970394

RESUMEN

In our previous work, using a fluorescent adenosine-A3 receptor (A3AR) agonist and fluorescence correlation spectroscopy (FCS), we demonstrated high-affinity labeling of the active receptor (R*) conformation. In the current study, we used a fluorescent A3AR antagonist (CA200645) to study the binding characteristics of antagonist-occupied inactive receptor (R) conformations in membrane microdomains of individual cells. FCS analysis of CA200645-occupied A3ARs revealed 2 species, τD2 and τD3, that diffused at 2.29 ± 0.35 and 0.09 ± 0.03 µm(2)/s, respectively. FCS analysis of a green fluorescent protein (GFP)-tagged A3AR exhibited a single diffusing species (0.105 µm(2)/s). The binding of CA200645 to τD3 was antagonized by nanomolar concentrations of the A3 antagonist MRS 1220, but not by the agonist NECA (up to 300 nM), consistent with labeling of R. CA200645 normally dissociated slowly from the A3AR, but inclusion of xanthine amine congener (XAC) or VUF 5455 during washout markedly accelerated the reduction in the number of particles exhibiting τD3 characteristics. It is notable that this effect was accompanied by a significant increase in the number of particles with τD2 diffusion. These data show that FCS analysis of ligand-occupied receptors provides a unique means of monitoring ligand A3AR residence times that are significantly reduced as a consequence of allosteric interaction across the dimer interface


Asunto(s)
Microdominios de Membrana/metabolismo , Receptor de Adenosina A3/metabolismo , Antagonistas del Receptor de Adenosina A3/farmacología , Regulación Alostérica , Animales , Células CHO , Cricetinae , Cricetulus , Humanos , Cinética , Unión Proteica , Multimerización de Proteína
17.
Br J Pharmacol ; 171(16): 3827-44, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24750014

RESUMEN

BACKGROUND AND PURPOSE: The highly conserved tryptophan (W6.48) in transmembrane domain 6 of GPCRs has been shown to play a central role in forming an active conformation in response to agonist binding. We set out to characterize the effect of this mutation on the efficacy of two agonists at multiple signalling pathways downstream of the adenosine A3 receptor. EXPERIMENTAL APPROACH: Residue W6.48 in the human adenosine A3 receptor fused to yellow fluorescent protein was mutated to phenylalanine and expressed in CHO-K1 cells containing a cAMP response element reporter gene. The effects on agonist-mediated receptor internalization were monitored by automated confocal microscopy and image analysis. Further experiments were carried out to investigate agonist-mediated ERK1/2 phosphorylation, inhibition of [(3)H]-cAMP accumulation and ß-arrestin2 binding. KEY RESULTS: NECA was able to stimulate agonist-mediated internalization of the W6.48F mutant receptor, while the agonist HEMADO was inactive. Investigation of other downstream signalling pathways indicated that G-protein coupling was impaired for both agonists tested. Mutation of W6.48F therefore resulted in differential effects on agonist efficacy, and introduced signalling pathway bias for HEMADO at the adenosine A3 receptor. CONCLUSIONS AND IMPLICATIONS: Investigation of the pharmacology of the W6.48F mutant of the adenosine A3 receptor confirms that this region is important in forming the active conformation of the receptor for stimulating a number of different signalling pathways and that mutations in this residue can lead to changes in agonist efficacy and signalling bias.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Agonistas del Receptor Purinérgico P1/farmacología , Receptor de Adenosina A3/genética , Receptor de Adenosina A3/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacología , Adenosina-5'-(N-etilcarboxamida)/farmacología , Animales , Arrestinas/metabolismo , Células CHO , Cricetulus , AMP Cíclico/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mutación , Estructura Terciaria de Proteína , Transducción de Señal , beta-Arrestinas
18.
EMBO Rep ; 14(8): 726-32, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23817552

RESUMEN

The A3-adenosine receptor (A3AR) has recently emerged as a key regulator of neutrophil behaviour. Using a fluorescent A3AR ligand, we show that A3ARs aggregate in highly polarized immunomodulatory microdomains on human neutrophil membranes. In addition to regulating chemotaxis, A3ARs promote the formation of filipodia-like projections (cytonemes) that can extend up to 100 µm to tether and 'reel in' pathogens. Exposure to bacteria or an A3AR agonist stimulates the formation of these projections and bacterial phagocytosis, whereas an A3AR-selective antagonist inhibits cytoneme formation. Our results shed new light on the behaviour of neutrophils and identify the A3AR as a potential target for modulating their function.


Asunto(s)
Estructuras de la Membrana Celular/ultraestructura , Neutrófilos/ultraestructura , Receptor de Adenosina A3/metabolismo , Estructuras de la Membrana Celular/efectos de los fármacos , Estructuras de la Membrana Celular/metabolismo , Estructuras de la Membrana Celular/microbiología , Escherichia coli/efectos de los fármacos , Escherichia coli/fisiología , Células HL-60 , Interacciones Huésped-Patógeno , Humanos , Ligandos , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Neutrófilos/microbiología , Fagocitosis/efectos de los fármacos , Unión Proteica , Antagonistas de Receptores Purinérgicos P1/farmacología
19.
Org Biomol Chem ; 11(34): 5673-82, 2013 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-23881285

RESUMEN

Advances in fluorescence-based imaging technologies have helped propel the study of real-time biological readouts and analysis across many different areas. In particular the use of fluorescent ligands as chemical tools to study proteins such as G protein-coupled receptors (GPCRs) has received ongoing interest. Methods to improve the efficient chemical synthesis of fluorescent ligands remain of paramount importance to ensure this area of bioanalysis continues to advance. Here we report conversion of the non-selective GPCR adenosine receptor antagonist Xanthine Amine Congener into higher affinity and more receptor subtype-selective fluorescent antagonists. This was achieved through insertion and optimisation of a dipeptide linker between the adenosine receptor pharmacophore and the fluorophore. Fluorescent probe 27 containing BODIPY 630/650 (pK(D) = 9.12 ± 0.05 [hA3AR]), and BODIPY FL-containing 28 (pK(D) = 7.96 ± 0.09 [hA3AR]) demonstrated clear, displaceable membrane binding using fluorescent confocal microscopy. From in silico analysis of the docked ligand-receptor complexes of 27, we suggest regions of molecular interaction that could account for the observed selectivity of these peptide-linker based fluorescent conjugates. This general approach of converting a non-selective ligand to a selective biological tool could be applied to other ligands of interest.


Asunto(s)
Colorantes Fluorescentes/química , Péptidos/química , Antagonistas de Receptores Purinérgicos P1/química , Animales , Células CHO , Cricetulus , Colorantes Fluorescentes/síntesis química , Humanos , Modelos Moleculares , Estructura Molecular
20.
Chem Biol ; 19(9): 1105-15, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22999879

RESUMEN

G protein-coupled receptors (GPCRs) comprise the largest family of transmembrane proteins. For GPCR drug discovery, it is important that ligand affinity is determined in the correct cellular environment and preferably using an unmodified receptor. We developed a live cell high-content screening assay that uses a fluorescent antagonist, CA200645, to determine binding affinity constants of competing ligands at human adenosine-A(1) and -A(3) receptors. This method was validated as a tool to screen a library of low molecular weight fragments, and identified a hit with submicromolar binding affinity (K(D)). This fragment was structurally unrelated to substructures of known adenosine receptor antagonists and was optimized to show selectivity for the adenosine-A(3) receptor. This technology represents a significant advance that will allow the determination of ligand and fragment affinities at receptors in their native membrane environment.


Asunto(s)
Fluorescencia , Mediciones Luminiscentes/métodos , Receptor de Adenosina A3/metabolismo , Bibliotecas de Moléculas Pequeñas/análisis , Bibliotecas de Moléculas Pequeñas/metabolismo , Animales , Células CHO , Supervivencia Celular , Células Cultivadas , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Humanos , Ligandos , Microscopía Confocal , Estructura Molecular , Unión Proteica , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA