Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Curr Protoc ; 3(10): e890, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37787634

ABSTRACT

G protein-coupled receptors (GPCRs) represent ∼30% of current drug targets. Ligand binding to these receptors activates G proteins and arrestins, which function in different signaling pathways. Given that functionally selective or biased ligands preferentially activate one of these two groups of pathways, they may be superior medications for certain disease states. The identification of such ligands requires robust drug screening assays for both G protein and arrestin activity. This unit describes protocols for assays that monitor reversible arrestin recruitment to GPCRs in living cells using either bioluminescence resonance energy transfer (BRET) or nanoluciferase complementation (NanoLuc). Two types of assays can be used: one configuration directly measures arrestin recruitment to a GPCR fused to a protein tag at its intracellular C-terminus, whereas the other configuration detects arrestin translocation to the plasma membrane in response to activation of an unmodified GPCR. Together, these assays are powerful tools for studying dynamic interactions between GPCRs and arrestins. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Receptor-arrestin BRET assay to measure ligand-induced recruitment of arrestin to receptors Basic Protocol 2: Receptor-arrestin NANOBIT assay to measure ligand-induced recruitment of arrestin to receptors Alternative Protocol 1: BRET assay to measure ligand-induced recruitment of arrestin to the plasma membrane Alternative Protocol 2: NANOBIT assay to measure ligand-induced recruitment of arrestin to the plasma membrane Support Protocol 1: Optimization of polyethylenimine (PEI) concentration for transfection.


Subject(s)
Arrestin , Arrestins , Ligands , Research Design , Cell Membrane
2.
J Am Chem Soc ; 145(34): 18778-18788, 2023 08 30.
Article in English | MEDLINE | ID: mdl-37586061

ABSTRACT

Dopamine D2-like receptors (D2R, D3R, and D4R) control diverse physiological and behavioral functions and are important targets for the treatment of a variety of neuropsychiatric disorders. Their complex distribution and activation kinetics in the brain make it difficult to target specific receptor populations with sufficient precision. We describe a new toolkit of light-activatable, fast-relaxing, covalently taggable chemical photoswitches that fully activate, partially activate, or block D2-like receptors. This technology combines the spatiotemporal precision of a photoswitchable ligand (P) with cell type and spatial specificity of a genetically encoded membrane anchoring protein (M) to which the P tethers. These tools set the stage for targeting endogenous D2-like receptor signaling with molecular, cellular, and spatiotemporal precision using only one wavelength of light.


Subject(s)
Dopamine , Receptors, Dopamine D2 , Dopamine/metabolism , Receptors, Dopamine D2/metabolism , Brain/metabolism
3.
Curr Opin Pharmacol ; 65: 102259, 2022 08.
Article in English | MEDLINE | ID: mdl-35749908

ABSTRACT

Blinding diseases that are caused by degeneration of rod and cone photoreceptor cells often spare the rest of the retinal circuit, from bipolar cells, which are directly innervated by photoreceptor cells, to the output ganglion cells that project axons to the brain. A strategy for restoring vision is to introduce light sensitivity to the surviving cells of the retina. One approach is optogenetics, in which surviving cells are virally transfected with a gene encoding a signaling protein that becomes sensitive to light by binding to the biologically available chromophore retinal, the same chromophore that is used by the opsin photo-detectors of rods and cones. A second approach uses photopharmacology, in which a synthetic photoswitch associates with a native or engineered ion channel or receptor. We review these approaches and look ahead to the next generation of advances that could reconstitute core aspects of natural vision.


Subject(s)
Retina , Retinal Cone Photoreceptor Cells , Humans , Optogenetics , Retinal Cone Photoreceptor Cells/metabolism , Rod Opsins/genetics , Rod Opsins/metabolism
4.
Nat Commun ; 12(1): 4775, 2021 08 06.
Article in English | MEDLINE | ID: mdl-34362914

ABSTRACT

Dopamine controls diverse behaviors and their dysregulation contributes to many disorders. Our ability to understand and manipulate the function of dopamine is limited by the heterogenous nature of dopaminergic projections, the diversity of neurons that are regulated by dopamine, the varying distribution of the five dopamine receptors (DARs), and the complex dynamics of dopamine release. In order to improve our ability to specifically modulate distinct DARs, here we develop a photo-pharmacological strategy using a Membrane anchored Photoswitchable orthogonal remotely tethered agonist for the Dopamine receptor (MP-D). Our design selectively targets D1R/D5R receptor subtypes, most potently D1R (MP-D1ago), as shown in HEK293T cells. In vivo, we targeted dorsal striatal medium spiny neurons where the photo-activation of MP-D1ago increased movement initiation, although further work is required to assess the effects of MP-D1ago on neuronal function. Our method combines ligand and cell type-specificity with temporally precise and reversible activation of D1R to control specific aspects of movement. Our results provide a template for analyzing dopamine receptors.


Subject(s)
Dopamine/metabolism , Neurons/metabolism , Receptors, Dopamine/chemistry , Receptors, Dopamine/metabolism , Animals , Brain/metabolism , Corpus Striatum/metabolism , Dopamine/chemistry , Dopamine Agonists/pharmacology , Female , HEK293 Cells , Humans , Ligands , Male , Mice , Receptors, Dopamine D1/chemistry , Receptors, Dopamine D1/metabolism , Synaptic Transmission/physiology
5.
J Am Chem Soc ; 143(24): 8951-8956, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34115935

ABSTRACT

G protein-coupled receptors (GPCRs) are the most common targets of drug discovery. However, the similarity between related GPCRs combined with the complex spatiotemporal dynamics of receptor activation in vivo has hindered drug development. Photopharmacology offers the possibility of using light to control the location and timing of drug action by incorporating a photoisomerizable azobenzene into a GPCR ligand, enabling rapid and reversible switching between an inactive and active configuration. Recent advances in this area include (i) photoagonists and photoantagonists that directly control receptor activity but are nonselective because they bind conserved sites, and (ii) photoallosteric modulators that bind selectively to nonconserved sites but indirectly control receptor activity by modulating the response to endogenous ligand. In this study, we designed a photoswitchable allosteric agonist that targets a nonconserved allosteric site for selectivity and activates the receptor on its own to provide direct control. This work culminated in the development of aBINA, a photoswitchable allosteric agonist that selectively activates the Gi/o-coupled metabotropic glutamate receptor 2 (mGluR2). aBINA is the first example of a new class of precision drugs for GPCRs and other clinically important signaling proteins.


Subject(s)
Benzene Derivatives/pharmacology , Receptors, G-Protein-Coupled/agonists , Allosteric Regulation/drug effects , Benzene Derivatives/chemical synthesis , Benzene Derivatives/chemistry , Humans , Ligands , Photochemical Processes
6.
J Am Chem Soc ; 142(24): 10612-10616, 2020 06 17.
Article in English | MEDLINE | ID: mdl-32469525

ABSTRACT

Lysophosphatidic acid (LPA) is a phospholipid that acts as an extracellular signaling molecule and activates the family of lysophosphatidic acid receptors (LPA1-6). These G protein-coupled receptors (GPCRs) are broadly expressed and are particularly important in development as well as in the nervous, cardiovascular, reproductive, gastrointestinal, and pulmonary systems. Here, we report on a photoswitchable analogue of LPA, termed AzoLPA, which contains an azobenzene photoswitch embedded in the acyl chain. AzoLPA enables optical control of LPA receptor activation, shown through its ability to rapidly control LPA-evoked increases in intracellular Ca2+ levels. AzoLPA shows greater activation of LPA receptors in its light-induced cis-form than its dark-adapted (or 460 nm light-induced) trans-form. AzoLPA enabled the optical control of neurite retraction through its activation of the LPA2 receptor.


Subject(s)
Lysophospholipids/metabolism , Humans , Lysophospholipids/chemistry , Photochemical Processes , Receptors, Lysophosphatidic Acid/chemistry , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction
7.
Mol Psychiatry ; 25(9): 2070-2085, 2020 09.
Article in English | MEDLINE | ID: mdl-30626912

ABSTRACT

Although long-studied in the central nervous system, there is increasing evidence that dopamine (DA) has important roles in the periphery including in metabolic regulation. Insulin-secreting pancreatic ß-cells express the machinery for DA synthesis and catabolism, as well as all five DA receptors. In these cells, DA functions as a negative regulator of glucose-stimulated insulin secretion (GSIS), which is mediated by DA D2-like receptors including D2 (D2R) and D3 (D3R) receptors. However, the fundamental mechanisms of DA synthesis, storage, release, and signaling in pancreatic ß-cells and their functional relevance in vivo remain poorly understood. Here, we assessed the roles of the DA precursor L-DOPA in ß-cell DA synthesis and release in conjunction with the signaling mechanisms underlying DA's inhibition of GSIS. Our results show that the uptake of L-DOPA is essential for establishing intracellular DA stores in ß-cells. Glucose stimulation significantly enhances L-DOPA uptake, leading to increased DA release and GSIS reduction in an autocrine/paracrine manner. Furthermore, D2R and D3R act in combination to mediate dopaminergic inhibition of GSIS. Transgenic knockout mice in which ß-cell D2R or D3R expression is eliminated exhibit diminished DA secretion during glucose stimulation, suggesting a new mechanism where D2-like receptors modify DA release to modulate GSIS. Lastly, ß-cell-selective D2R knockout mice exhibit marked postprandial hyperinsulinemia in vivo. These results reveal that peripheral D2R and D3R receptors play important roles in metabolism through their inhibitory effects on GSIS. This opens the possibility that blockade of peripheral D2-like receptors by drugs including antipsychotic medications may significantly contribute to the metabolic disturbances observed clinically.


Subject(s)
Dopamine , Insulin-Secreting Cells , Animals , Dopamine/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/genetics , Receptors, Dopamine D3/metabolism
8.
Mol Psychiatry ; 25(9): 2086-2100, 2020 09.
Article in English | MEDLINE | ID: mdl-30120413

ABSTRACT

The dopamine (DA) D2 receptor (D2R) is an important target for the treatment of neuropsychiatric disorders such as schizophrenia and Parkinson's disease. However, the development of improved therapeutic strategies has been hampered by our incomplete understanding of this receptor's downstream signaling processes in vivo and how these relate to the desired and undesired effects of drugs. D2R is a G protein-coupled receptor (GPCR) that activates G protein-dependent as well as non-canonical arrestin-dependent signaling pathways. Whether these effector pathways act alone or in concert to facilitate specific D2R-dependent behaviors is unclear. Here, we report on the development of a D2R mutant that recruits arrestin but is devoid of G protein activity. When expressed virally in "indirect pathway" medium spiny neurons (iMSNs) in the ventral striatum of D2R knockout mice, this mutant restored basal locomotor activity and cocaine-induced locomotor activity in a manner indistinguishable from wild-type D2R, indicating that arrestin recruitment can drive locomotion in the absence of D2R-mediated G protein signaling. In contrast, incentive motivation was enhanced only by wild-type D2R, signifying a dissociation in the mechanisms that underlie distinct D2R-dependent behaviors, and opening the door to more targeted therapeutics.


Subject(s)
Arrestin , Locomotion , Motivation , Receptors, Dopamine D2 , Animals , Cocaine , Corpus Striatum/metabolism , Mice , Mice, Knockout , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism
9.
J Am Chem Soc ; 141(29): 11522-11530, 2019 07 24.
Article in English | MEDLINE | ID: mdl-31291105

ABSTRACT

G protein-coupled receptors (GPCRs) are membrane proteins that play important roles in biology. However, our understanding of their function in complex living systems is limited because we lack tools that can target individual receptors with sufficient precision. State-of-the-art approaches, including DREADDs, optoXRs, and PORTL gated-receptors, control GPCR signaling with molecular, cell type, and temporal specificity. Nonetheless, these tools are based on engineered non-native proteins that may (i) express at nonphysiological levels, (ii) localize and turnover incorrectly, and/or (iii) fail to interact with endogenous partners. Alternatively, membrane-anchored ligands (t-toxins, DARTs) target endogenous receptors with molecular and cell type specificity but cannot be turned on and off. In this study, we used a combination of chemistry, biology, and light to control endogenous metabotropic glutamate receptor 2 (mGluR2), a Family C GPCR, in primary cortical neurons. mGluR2 was rapidly, reversibly, and selectively activated with photoswitchable glutamate tethered to a genetically targeted-plasma membrane anchor (membrane anchored Photoswitchable Orthogonal Remotely Tethered Ligand; maPORTL). Photoactivation was tuned by adjusting the length of the PORTL as well as the expression level and geometry of the membrane anchor. Our findings provide a template for controlling endogenous GPCRs with cell type specificity and high spatiotemporal precision.


Subject(s)
Molecular Biology/methods , Receptors, Metabotropic Glutamate/genetics , Amino Acids/pharmacology , Animals , Azo Compounds/chemistry , Cell Membrane/metabolism , Glutamic Acid/chemistry , HEK293 Cells , Humans , Ligands , Light , Neurons/metabolism , Photochemical Processes , Protein Engineering/methods , Rats , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Xanthenes/pharmacology
10.
Nat Chem Biol ; 15(6): 623-631, 2019 06.
Article in English | MEDLINE | ID: mdl-31036923

ABSTRACT

Sphingosine-1-phosphate (S1P) plays important roles as a signaling lipid in a variety of physiological and pathophysiological processes. S1P signals via a family of G-protein-coupled receptors (GPCRs) (S1P1-5) and intracellular targets. Here, we report on photoswitchable analogs of S1P and its precursor sphingosine, respectively termed PhotoS1P and PhotoSph. PhotoS1P enables optical control of S1P1-3, shown through electrophysiology and Ca2+ mobilization assays. We evaluated PhotoS1P in vivo, where it reversibly controlled S1P3-dependent pain hypersensitivity in mice. The hypersensitivity induced by PhotoS1P is comparable to that induced by S1P. PhotoS1P is uniquely suited for the study of S1P biology in cultured cells and in vivo because it exhibits prolonged metabolic stability compared to the rapidly metabolized S1P. Using lipid mass spectrometry analysis, we constructed a metabolic map of PhotoS1P and PhotoSph. The formation of these photoswitchable lipids was found to be light dependent, providing a novel approach to optically probe sphingolipid biology.


Subject(s)
Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Animals , Lysophospholipids/chemistry , Mice , Models, Molecular , Molecular Structure , Optical Imaging , Photochemical Processes , Sphingosine/chemistry , Sphingosine/metabolism
11.
Neuron ; 98(3): 575-587.e4, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29656874

ABSTRACT

Dopamine input to the dorsal and ventral striatum originates from separate populations of midbrain neurons. Despite differences in afferent inputs and behavioral output, little is known about how dopamine release is encoded by dopamine receptors on medium spiny neurons (MSNs) across striatal subregions. Here we examined the activation of D2 receptors following the synaptic release of dopamine in the dorsal striatum (DStr) and nucleus accumbens (NAc) shell. We found that D2 receptor-mediated synaptic currents were slower in the NAc and this difference occurred at the level of D2-receptor signaling. As a result of preferential coupling to Gαo, we also found that D2 receptors in MSNs demonstrated higher sensitivity for dopamine in the NAc. The higher sensitivity in the NAc was eliminated following cocaine exposure. These results identify differences in the sensitivity and timing of D2-receptor signaling across the striatum that influence how nigrostriatal and mesolimbic signals are encoded across these circuits.


Subject(s)
Corpus Striatum/metabolism , Nucleus Accumbens/metabolism , Receptors, Dopamine D2/metabolism , Signal Transduction/physiology , Animals , Corpus Striatum/drug effects , Dopamine/metabolism , Dopamine/pharmacology , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Mice , Mice, Knockout , Mice, Transgenic , Nucleus Accumbens/drug effects , Receptors, Dopamine D2/agonists , Signal Transduction/drug effects
12.
J Am Chem Soc ; 139(51): 18522-18535, 2017 12 27.
Article in English | MEDLINE | ID: mdl-29166564

ABSTRACT

Family A G protein-coupled receptors (GPCRs) control diverse biological processes and are of great clinical relevance. Their archetype rhodopsin becomes naturally light sensitive by binding covalently to the photoswitchable tethered ligand (PTL) retinal. Other GPCRs, however, neither bind covalently to ligands nor are light sensitive. We sought to impart the logic of rhodopsin to light-insensitive Family A GPCRs in order to enable their remote control in a receptor-specific, cell-type-specific, and spatiotemporally precise manner. Dopamine receptors (DARs) are of particular interest for their roles in motor coordination, appetitive, and aversive behavior, as well as neuropsychiatric disorders such as Parkinson's disease, schizophrenia, mood disorders, and addiction. Using an azobenzene derivative of the well-known DAR ligand 2-(N-phenethyl-N-propyl)amino-5-hydroxytetralin (PPHT), we were able to rapidly, reversibly, and selectively block dopamine D1 and D2 receptors (D1R and D2R) when the PTL was conjugated to an engineered cysteine near the dopamine binding site. Depending on the site of tethering, the ligand behaved as either a photoswitchable tethered neutral antagonist or inverse agonist. Our results indicate that DARs can be chemically engineered for selective remote control by light and provide a template for precision control of Family A GPCRs.


Subject(s)
Dopamine D2 Receptor Antagonists/pharmacology , Dopamine D2 Receptor Antagonists/radiation effects , Drug Inverse Agonism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D1/radiation effects , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D2/radiation effects , Binding Sites , Cysteine/chemistry , Dopamine/metabolism , Humans , Ligands , Receptors, Dopamine D1/antagonists & inhibitors
13.
J Med Chem ; 60(2): 580-593, 2017 01 26.
Article in English | MEDLINE | ID: mdl-27983845

ABSTRACT

Both dopamine D3 receptor (D3R) partial agonists and antagonists have been implicated as potential medications for substance use disorders. In contrast to antagonists, partial agonists may cause fewer side effects since they maintain some dopaminergic tone and may be less disruptive to normal neuronal functions. Here, we report three sets of 4-phenylpiperazine stereoisomers that differ considerably in efficacy: the (R)-enantiomers are antagonists/weak partial agonists, whereas the (S)-enantiomers are much more efficacious. To investigate the structural basis of partial agonism, we performed comparative microsecond-scale molecular dynamics simulations starting from the inactive state of D3R in complex with these enantiomers. Analysis of the simulation results reveals common structural rearrangements near the ligand binding site induced by the bound (S)-enantiomers, but not by the (R)-enantiomers, that are features of partially activated receptor conformations. These receptor models bound with partial agonists may be useful for structure-based design of compounds with tailored efficacy profiles.


Subject(s)
Piperazines/pharmacology , Receptors, Dopamine D3/agonists , Receptors, Dopamine D3/chemistry , Binding Sites , Drug Partial Agonism , HEK293 Cells , Humans , Molecular Dynamics Simulation , Piperazines/chemical synthesis , Piperazines/chemistry , Protein Structure, Tertiary , Stereoisomerism
14.
J Med Chem ; 59(7): 2973-88, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27035329

ABSTRACT

Novel 1-, 5-, and 8-substituted analogues of sumanirole (1), a dopamine D2/D3 receptor (D2R/D3R) agonist, were synthesized. Binding affinities at both D2R and D3R were higher when determined in competition with the agonist radioligand [(3)H]7-hydroxy-N,N-dipropyl-2-aminotetralin (7-OH-DPAT) than with the antagonist radioligand [(3)H]N-methylspiperone. Although 1 was confirmed as a D2R-preferential agonist, its selectivity in binding and functional studies was lower than previously reported. All analogues were determined to be D2R/D3R agonists in both GoBRET and mitogenesis functional assays. Loss of efficacy was detected for the N-1-substituted analogues at D3R. In contrast, the N-5-alkyl-substituted analogues, and notably the n-butyl-arylamides (22b and 22c), all showed improved affinity at D2R over 1 with neither a loss of efficacy nor an increase in selectivity. Computational modeling provided a structural basis for the D2R selectivity of 1, illustrating how subtle differences in the highly homologous orthosteric binding site (OBS) differentially affect D2R/D3R affinity and functional efficacy.


Subject(s)
Benzimidazoles/chemistry , Receptors, Dopamine D2/agonists , Receptors, Dopamine D3/agonists , Structure-Activity Relationship , Animals , Binding Sites , CHO Cells , Chemistry Techniques, Synthetic , Cricetulus , Humans , Ligands , Models, Molecular , Molecular Dynamics Simulation , Radioligand Assay , Receptors, Dopamine D2/genetics , Receptors, Dopamine D3/genetics
15.
Nat Commun ; 7: 10842, 2016 Feb 24.
Article in English | MEDLINE | ID: mdl-26905976

ABSTRACT

Biased agonism describes the ability of ligands to stabilize different conformations of a GPCR linked to distinct functional outcomes and offers the prospect of designing pathway-specific drugs that avoid on-target side effects. This mechanism is usually inferred from pharmacological data with the assumption that the confounding influences of observational (that is, assay dependent) and system (that is, cell background dependent) bias are excluded by experimental design and analysis. Here we reveal that 'kinetic context', as determined by ligand-binding kinetics and the temporal pattern of receptor-signalling processes, can have a profound influence on the apparent bias of a series of agonists for the dopamine D2 receptor and can even lead to reversals in the direction of bias. We propose that kinetic context must be acknowledged in the design and interpretation of studies of biased agonism.


Subject(s)
Dopamine Agonists/pharmacokinetics , Receptors, Dopamine D2/agonists , Animals , Aripiprazole/pharmacokinetics , CHO Cells , Cricetulus , Dopamine/pharmacokinetics , Indoles/pharmacokinetics , Kinetics , Ligands , Piperazines/pharmacokinetics , Piperidines/pharmacokinetics , Principal Component Analysis , Protein Stability , Receptors, Dopamine D2/metabolism
16.
PLoS One ; 11(2): e0148684, 2016.
Article in English | MEDLINE | ID: mdl-26849707

ABSTRACT

Direct measurement of insulin is critical for basic and clinical studies of insulin secretion. However, current methods are expensive and time-consuming. We developed an insulin assay based on homogenous time-resolved fluorescence that is significantly more rapid and cost-effective than current commonly used approaches. This assay was applied effectively to an insulin secreting cell line, INS-1E cells, as well as pancreatic islets, allowing us to validate the assay by elucidating mechanisms by which dopamine regulates insulin release. We found that dopamine functioned as a significant negative modulator of glucose-stimulated insulin secretion. Further, we showed that bromocriptine, a known dopamine D2/D3 receptor agonist and newly approved drug used for treatment of type II diabetes mellitus, also decreased glucose-stimulated insulin secretion in islets to levels comparable to those caused by dopamine treatment.


Subject(s)
Fluorescence , Insulin-Secreting Cells/metabolism , Insulin/analysis , Animals , Bromocriptine/chemistry , Bromocriptine/pharmacology , Cell Line , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Mice , Rats , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/agonists , Receptors, Dopamine D3/metabolism
17.
Curr Protoc Pharmacol ; 70: 2.14.1-2.14.14, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26331887

ABSTRACT

G protein-coupled receptors (GPCRs) represent ∼25% of current drug targets. Ligand binding to these receptors activates G proteins and arrestins, which are involved in differential signaling pathways. Because functionally selective or biased ligands activate one of these two pathways, they may be superior medications for certain diseases states. The identification of such ligands requires robust drug screening assays for both G protein and arrestin activity. This unit describes protocols for two bioluminescence resonance energy transfer (BRET)-based assays used to monitor arrestin recruitment to GPCRs. One assay requires modification of GPCRs by fusion to a BRET donor or acceptor moiety, whereas the other can detect arrestin recruitment to unmodified GPCRs.


Subject(s)
Arrestin/agonists , Arrestin/metabolism , Luminescent Measurements/methods , Receptors, G-Protein-Coupled/drug effects , Arrestin/antagonists & inhibitors , Cell Membrane/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Ligands , Polyethyleneimine/pharmacology , Translocation, Genetic
18.
J Med Chem ; 58(15): 6195-213, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26203768

ABSTRACT

The dopamine D3 receptor (D3R) is a promising target for the development of pharmacotherapeutics to treat substance use disorders. Several D3R-selective antagonists are effective in animal models of drug abuse, especially in models of relapse. Nevertheless, poor bioavailability, metabolic instability, and/or predicted toxicity have impeded success in translating these drug candidates to clinical use. Herein, we report a series of D3R-selective 4-phenylpiperazines with improved metabolic stability. A subset of these compounds was evaluated for D3R functional efficacy and off-target binding at selected 5-HT receptor subtypes, where significant overlap in SAR with D3R has been observed. Several high affinity D3R antagonists, including compounds 16 (Ki = 0.12 nM) and 32 (Ki = 0.35 nM), showed improved metabolic stability compared to the parent compound, PG648 (6). Notably, 16 and the classic D3R antagonist SB277011A (2) were effective in reducing self-administration of heroin in wild-type but not D3R knockout mice.


Subject(s)
Dopamine Antagonists/pharmacology , Heroin/administration & dosage , Receptors, Dopamine D3/antagonists & inhibitors , Self Administration , Animals , Dopamine Antagonists/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Radioligand Assay , Receptors, Dopamine D3/genetics
19.
Pharmacol Rev ; 67(1): 198-213, 2015.
Article in English | MEDLINE | ID: mdl-25527701

ABSTRACT

G protein-coupled receptors (GPCRs) are integral membrane proteins that represent an important class of drug targets. In particular, aminergic GPCRs interact with a significant portion of drugs currently on the market. However, most drugs that target these receptors are associated with undesirable side effects, which are due in part to promiscuous interactions with close homologs of the intended target receptors. Here, based on a systematic analysis of all 37 of the currently available high-resolution crystal structures of aminergic GPCRs, we review structural elements that contribute to and can be exploited for designing subtype-selective compounds. We describe the roles of secondary binding pockets (SBPs), as well as differences in ligand entry pathways to the orthosteric binding site, in determining selectivity. In addition, using the available crystal structures, we have identified conformational changes in the SBPs that are associated with receptor activation and explore the implications of these changes for the rational development of selective ligands with tailored efficacy.


Subject(s)
Amines/chemistry , Drug Design , Molecular Targeted Therapy , Receptors, G-Protein-Coupled/chemistry , Amines/metabolism , Amino Acid Sequence , Animals , Binding Sites , Crystallography , Humans , Ligands , Molecular Sequence Data , Protein Conformation , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship
20.
J Biol Chem ; 289(48): 33663-75, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25336643

ABSTRACT

Arrestins mediate desensitization and internalization of G protein-coupled receptors and also direct receptor signaling toward heterotrimeric G protein-independent signaling pathways. We previously identified a four-residue segment (residues 212-215) of the dopamine D2 receptor that is necessary for arrestin binding in an in vitro heterologous expression system but that also impairs receptor expression. We now describe the characterization of additional mutations at that arrestin binding site in the third intracellular loop. Mutating two (residues 214 and 215) or three (residues 213-215) of the four residues to alanine partially decreased agonist-induced recruitment of arrestin3 without altering activation of a G protein. Arrestin-dependent receptor internalization, which requires arrestin binding to ß2-adaptin (the ß2 subunit of the clathrin-associated adaptor protein AP2) and clathrin, was disproportionately affected by the three-residue mutation, with no agonist-induced internalization observed even in the presence of overexpressed arrestin or G protein-coupled receptor kinase 2. The disjunction between arrestin recruitment and internalization could not be explained by alterations in the time course of the receptor-arrestin interaction, the recruitment of G protein-coupled receptor kinase 2, or the receptor-induced interaction between arrestin and ß2-adaptin, suggesting that the mutation impairs a property of the internalization complex that has not yet been identified.


Subject(s)
Mutation , Receptors, Dopamine D2/metabolism , Adaptor Protein Complex beta Subunits/genetics , Adaptor Protein Complex beta Subunits/metabolism , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 2/metabolism , HEK293 Cells , Humans , Protein Structure, Secondary , Protein Transport/physiology , Receptors, Dopamine D2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...