Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
ACS Pharmacol Transl Sci ; 5(8): 668-678, 2022 Aug 12.
Article in English | MEDLINE | ID: mdl-35983281

ABSTRACT

The two human melatonin receptors MT1 and MT2, which belong to the G protein-coupled receptor (GPCR) family, are important drug targets with approved indications for circadian rhythm- and sleep-related disorders and major depression. Currently, most of the pharmacological studies were performed using [3H]melatonin and 2-[125I]iodomelatonin (2-[125I]-MLT) radioligands. Recently, NanoLuc-based bioluminescence resonance energy transfer (NanoBRET) monitoring competitive binding between fluorescent tracers and unmodified test compounds has emerged as a sensitive, nonradioactive alternative for quantifying GPCR ligand engagement on the surface of living cells in equilibrium and real time. However, developing such assays for the two melatonin receptors depends on the availability of fluorescent tracers, which has been challenging predominantly owing to their narrow ligand entry channel and small ligand binding pocket. Here, we generated a set of melatonergic fluorescent tracers and used NanoBRET to evaluate their engagement with MT1 and MT2 receptors that are genetically fused to an N-terminal luminogenic HiBiT-peptide. We identified several nonselective and subtype-selective tracers. Among the selective tracers, PBI-8238 exhibited high nanomolar affinity to MT1, and PBI-8192 exhibited low nanomolar affinity to MT2. The pharmacological profiles of both tracers were in good agreement with those obtained with the current standard 2-[125I]-MLT radioligand. Molecular docking and mutagenesis studies suggested the binding mode of PBI-8192 in MT2 and its selectivity over MT1. In conclusion, we describe the development of the first nonradioactive, real-time binding assays for melatonin receptors expressed at the cell surface of living cells that are likely to accelerate drug discovery for melatonin receptors.

2.
Bio Protoc ; 10(24): e3861, 2020 Dec 20.
Article in English | MEDLINE | ID: mdl-33659503

ABSTRACT

G-protein coupled receptors (GPCRs) remain at the forefront of drug discovery efforts. Detailed assessment of features contributing to GPCR ligand engagement in a physiologically relevant environment is imperative to the development of new therapeutics with improved efficacy. Traditionally, binding properties such as affinity and kinetics were obtained using biochemical radioligand binding assays. More recently, the high specificity of resonance energy transfer has been leveraged toward the development of homogeneous cell-based proximity assays with capacity for real-time kinetic measurements. This suite of ligand binding protocols couples the specificity of bioluminescent resonance energy transfer (BRET) with the sensitivity afforded by the luminescent HiBiT peptide. The BRET format is used to quantify dynamic interactions between ligands and their cognate HiBiT-tagged GPCRs through competitive binding with fluorescent Tracers. At the same time, high affinity complementation of HiBiT with the cell impermeable LgBiT limits the bright bioluminescence donor signal to the cell surface and eliminates luminescence background from unoccupied receptors present in intracellular compartments.

3.
Cell Chem Biol ; 26(6): 830-841.e9, 2019 06 20.
Article in English | MEDLINE | ID: mdl-30956148

ABSTRACT

Vascular endothelial growth factor (VEGF) is an important mediator of endothelial cell proliferation and angiogenesis via its receptor VEGFR2. A common tumor associated with elevated VEGFR2 signaling is infantile hemangioma that is caused by a rapid proliferation of vascular endothelial cells. The current first-line treatment for infantile hemangioma is the ß-adrenoceptor antagonist, propranolol, although its mechanism of action is not understood. Here we have used bioluminescence resonance energy transfer and VEGFR2 genetically tagged with NanoLuc luciferase to demonstrate that oligomeric complexes involving VEGFR2 and the ß2-adrenoceptor can be generated in both cell membranes and intracellular endosomes. These complexes are induced by agonist treatment and retain their ability to couple to intracellular signaling proteins. Furthermore, coupling of ß2-adrenoceptor to ß-arrestin2 is prolonged by VEGFR2 activation. These data suggest that protein-protein interactions between VEGFR2, the ß2-adrenoceptor, and ß-arrestin2 may provide insight into their roles in health and disease.


Subject(s)
Receptors, Adrenergic, beta-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Bioluminescence Resonance Energy Transfer Techniques , Cells, Cultured , Fluorescent Dyes/chemistry , HEK293 Cells , Humans , Ligands , Luciferases/chemistry , Luciferases/metabolism , Protein Binding , Receptors, Adrenergic, beta-2/chemistry , Vascular Endothelial Growth Factor Receptor-2/chemistry , Vascular Endothelial Growth Factor Receptor-2/genetics
4.
Methods Mol Biol ; 1888: 45-71, 2019.
Article in English | MEDLINE | ID: mdl-30519940

ABSTRACT

Intracellular target affinity and residence time are fundamental aspects of pharmacological mechanism (Lu and Tonge, Curr Opin Chem Biol 14:467-474, 2010). Although various robust biochemical approaches exist to measure these binding characteristics, analysis of compound binding with isolated targets may not accurately reflect engagement in the milieu of living cells. To realize the influence of cellular context, methods are needed that are capable of quantifying affinity and residence time in the presence of the intracellular factors that may impact target engagement. Bioluminescence resonance energy transfer (BRET) offers a solution for intracellular target engagement when quantitative metrics or kinetic analyses are required.


Subject(s)
Drug Discovery/methods , Fluorescence Resonance Energy Transfer , Luminescent Measurements , Cell Culture Techniques , Cell Line , Fluorescence Resonance Energy Transfer/methods , High-Throughput Screening Assays , Humans , Luminescent Measurements/methods , Molecular Probes/chemistry , Molecular Probes/metabolism , Permeability , Reproducibility of Results
5.
Cell Chem Biol ; 25(2): 206-214.e11, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29174542

ABSTRACT

For kinase inhibitors, intracellular target selectivity is fundamental to pharmacological mechanism. Although a number of acellular techniques have been developed to measure kinase binding or enzymatic inhibition, such approaches can fail to accurately predict engagement in cells. Here we report the application of an energy transfer technique that enabled the first broad-spectrum, equilibrium-based approach to quantitatively profile target occupancy and compound affinity in live cells. Using this method, we performed a selectivity profiling for clinically relevant kinase inhibitors against 178 full-length kinases, and a mechanistic interrogation of the potency offsets observed between cellular and biochemical analysis. For the multikinase inhibitor crizotinib, our approach accurately predicted cellular potency and revealed improved target selectivity compared with biochemical measurements. Due to cellular ATP, a number of putative crizotinib targets are unexpectedly disengaged in live cells at a clinically relevant drug dose.


Subject(s)
Adenosine Triphosphate/metabolism , Phosphotransferases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Cell Survival , Dose-Response Relationship, Drug , Energy Transfer , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Humans , Mass Spectrometry , Molecular Structure , Phosphotransferases/metabolism , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
6.
Nat Commun ; 6: 10091, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26631872

ABSTRACT

The therapeutic action of drugs is predicated on their physical engagement with cellular targets. Here we describe a broadly applicable method using bioluminescence resonance energy transfer (BRET) to reveal the binding characteristics of a drug with selected targets within intact cells. Cell-permeable fluorescent tracers are used in a competitive binding format to quantify drug engagement with the target proteins fused to Nanoluc luciferase. The approach enabled us to profile isozyme-specific engagement and binding kinetics for a panel of histone deacetylase (HDAC) inhibitors. Our analysis was directed particularly to the clinically approved prodrug FK228 (Istodax/Romidepsin) because of its unique and largely unexplained mechanism of sustained intracellular action. Analysis of the binding kinetics by BRET revealed remarkably long intracellular residence times for FK228 at HDAC1, explaining the protracted intracellular behaviour of this prodrug. Our results demonstrate a novel application of BRET for assessing target engagement within the complex milieu of the intracellular environment.


Subject(s)
Cells/drug effects , Fluorescence Resonance Energy Transfer/methods , Histone Deacetylase Inhibitors/chemistry , Pharmaceutical Preparations/chemistry , Cell Proliferation , Cells/chemistry , Cells/cytology , HeLa Cells , Histone Deacetylase 1/chemistry , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Humans , Luciferases/chemistry , Luciferases/genetics , Luciferases/metabolism , Luminescence
7.
ACS Chem Biol ; 10(10): 2316-24, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26162280

ABSTRACT

Phenotypic screening of compound libraries is a significant trend in drug discovery, yet success can be hindered by difficulties in identifying the underlying cellular targets. Current approaches rely on tethering bioactive compounds to a capture tag or surface to allow selective enrichment of interacting proteins for subsequent identification by mass spectrometry. Such methods are often constrained by ineffective capture of low affinity and low abundance targets. In addition, these methods are often not compatible with living cells and therefore cannot be used to verify the pharmacological activity of the tethered compounds. We have developed a novel chloroalkane capture tag that minimally affects compound potency in cultured cells, allowing binding interactions with the targets to occur under conditions relevant to the desired cellular phenotype. Subsequent isolation of the interacting targets is achieved through rapid lysis and capture onto immobilized HaloTag protein. Exchanging the chloroalkane tag for a fluorophore, the putative targets identified by mass spectrometry can be verified for direct binding to the compound through resonance energy transfer. Using the interaction between histone deacetylases (HDACs) and the inhibitor, Vorinostat (SAHA), as a model system, we were able to identify and verify all the known HDAC targets of SAHA as well as two previously undescribed targets, ADO and CPPED1. The discovery of ADO as a target may provide mechanistic insight into a reported connection between SAHA and Huntington's disease.


Subject(s)
Alkanes/chemistry , Chemistry Techniques, Analytical/methods , Chlorine/chemistry , Drug Discovery , Chromatography, Liquid , Drug Delivery Systems , HEK293 Cells , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Protein Binding/drug effects , Vorinostat
8.
Curr Protoc Mol Biol ; 110: 10.31.1-10.31.15, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25827085

ABSTRACT

Cultured mammalian cells provide an environment ideal for producing functional recombinant mammalian proteins. However, low expression levels of recombinant proteins present a challenge for their detection and purification. This unit will focus on HaloTag, a protein fusion tag designed to bind selectively and covalently to a chloroalkane ligand that may be attached to a variety of functional groups, allowing both protein detection and immobilization. Detection of HaloTag-fusion protein is achieved through binding to a fluorescent chloroalkane ligand, enabling rapid optimization of expression levels. HaloTag-based purification uses covalent capture of the HaloTag fusion onto HaloLink resin coupled with proteolytic cleavage to release the protein of interest from the resin. Covalent binding provides efficient protein capture regardless of expression level and eliminates protein loss during washes of the resin and as a result, offers significant improvements in protein recovery and purity over traditional non-covalent approaches.


Subject(s)
Chromatography, Affinity/methods , Recombinant Fusion Proteins/isolation & purification , Staining and Labeling/methods , Animals , Cell Line , Humans , Mammals , Recombinant Fusion Proteins/genetics
9.
Protein Expr Purif ; 76(2): 154-64, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21129486

ABSTRACT

Although cultured mammalian cells are preferred for producing functional mammalian proteins with appropriate post-translational modifications, purification of recombinant proteins is frequently hampered by low expression. We have addressed this by creating a new method configured specifically for mammalian cell culture that provides rapid detection and efficient purification. This approach is based on HaloTag, a protein fusion tag designed to bind rapidly, selectively and covalently to a series of synthetic ligands that can carry a variety of functional groups, including fluorescent dyes for detection or solid supports for purification. Since the binding of HaloTag to the HaloLink resin is essentially irreversible, it overcomes the equilibrium-based binding limitations associated with affinity tags and enables efficient capture and purification of target protein, even at low expression levels. The target protein is released from the HaloLink resin by specific cleavage using a TEV protease fused to HaloTag (HaloTEV), leaving both HaloTag and HaloTEV permanently attached to the resin and highly pure, tag-free protein in solution. HaloTag fluorescent ligands enable fluorescent labeling of HaloTag fusion proteins, providing a convenient way to monitor expression, and thus facilitate the identification of optimal transient transfection conditions as well as the selection of high expression stable cell lines. The capabilities of this method have been demonstrated by the efficient purification of five functional human kinases from HEK293T cells. In addition, when purifications using FLAG, 3xFLAG, His(6)Tag and HaloTag were performed in parallel, HaloTag was shown to provide significantly higher yields, purity and overall recovery of the expressed proteins.


Subject(s)
Chromatography, Affinity/methods , Cloning, Molecular/methods , Protein Kinases/isolation & purification , Recombinant Fusion Proteins/isolation & purification , Blotting, Western , Cell Culture Techniques , Electrophoresis, Polyacrylamide Gel , HEK293 Cells , Humans , Immobilized Proteins/metabolism , Protein Kinases/analysis , Protein Kinases/metabolism , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/metabolism , Time Factors
10.
Protein Expr Purif ; 68(1): 110-20, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19464373

ABSTRACT

Over-expression and purification of soluble and functional proteins remain critical challenges for many aspects of biomolecular research. To address this, we have developed a novel protein tag, HaloTag7, engineered to enhance expression and solubility of recombinant proteins and to provide efficient protein purification coupled with tag removal. HaloTag7 was designed to bind rapidly and covalently with a unique synthetic linker to achieve an essentially irreversible attachment. The synthetic linker may be attached to a variety of entities such as fluorescent dyes and solid supports, permitting labeling of fusion proteins in cell lysates for expression screening, and efficient capture of fusion proteins onto a purification resin. The combination of covalent capture with rapid binding kinetics overcomes the equilibrium-based limitations associated with traditional affinity tags and enables efficient capture even at low expression levels. Following immobilization on the resin, the protein of interest is released by cleavage at an optimized TEV protease recognition site, leaving HaloTag7 bound to the resin and pure protein in solution. Evaluation of HaloTag7 for expression of 23 human proteins in Escherichia coli relative to MBP, GST and His(6)Tag revealed that 74% of the proteins were produced in soluble form when fused to HaloTag7 compared to 52%, 39% and 22%, respectively, for the other tags. Using a subset of the test panel, more proteins fused to HaloTag7 were successfully purified than with the other tags, and these proteins were of higher yield and purity.


Subject(s)
Protein Engineering/methods , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Chromatography, Gel , Electrophoresis, Polyacrylamide Gel , Escherichia coli/metabolism , Humans , Pilot Projects , Recombinant Fusion Proteins/genetics , Solubility
11.
ACS Chem Biol ; 3(6): 373-82, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18533659

ABSTRACT

We have designed a modular protein tagging system that allows different functionalities to be linked onto a single genetic fusion, either in solution, in living cells, or in chemically fixed cells. The protein tag (HaloTag) is a modified haloalkane dehalogenase designed to covalently bind to synthetic ligands (HaloTag ligands). The synthetic ligands comprise a chloroalkane linker attached to a variety of useful molecules, such as fluorescent dyes, affinity handles, or solid surfaces. Covalent bond formation between the protein tag and the chloroalkane linker is highly specific, occurs rapidly under physiological conditions, and is essentially irreversible. We demonstrate the utility of this system for cellular imaging and protein immobilization by analyzing multiple molecular processes associated with NF-kappaB-mediated cellular physiology, including imaging of subcellular protein translocation and capture of protein--protein and protein--DNA complexes.


Subject(s)
Biosensing Techniques/methods , Cells/cytology , Fluorescent Dyes/chemistry , Luminescent Measurements/methods , Luminescent Proteins/chemistry , Staining and Labeling , Animals , Binding Sites , Cells/metabolism , DNA/analysis , DNA/chemistry , DNA/metabolism , Enzymes, Immobilized , Humans , Hydrocarbons, Chlorinated/chemistry , NF-kappa B/analysis , NF-kappa B/metabolism , Proteins/analysis , Proteins/chemistry , Proteins/metabolism , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL