Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
2.
Cell Chem Biol ; 29(2): 287-299.e8, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34520747

ABSTRACT

Contemporary drug discovery typically quantifies the effect of a molecule on a biological target using the equilibrium-derived measurements of IC50, EC50, or KD. Kinetic descriptors of drug binding are frequently linked with the effectiveness of a molecule in modulating a disease phenotype; however, these parameters are yet to be fully adopted in early drug discovery. Nanoluciferase bioluminescence resonance energy transfer (NanoBRET) can be used to measure interactions between fluorophore-conjugated probes and luciferase fused target proteins. Here, we describe an intracellular NanoBRET competition assay that can be used to quantify cellular kinetic rates of compound binding to nanoluciferase-fused bromodomain and extra-terminal (BET) proteins. Comparative rates are generated using a cell-free NanoBRET assay and by utilizing orthogonal recombinant protein-based methodologies. A screen of known pan-BET inhibitors is used to demonstrate the value of this approach in the investigation of kinetic selectivity between closely related proteins.


Subject(s)
Luciferases/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/metabolism , Binding Sites , Bioluminescence Resonance Energy Transfer Techniques , Cells, Cultured , Female , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , HEK293 Cells , Humans , Kinetics , Luciferases/chemistry , Nerve Tissue Proteins/chemistry , Receptors, Cell Surface/chemistry
3.
SLAS Discov ; 26(5): 663-675, 2021 06.
Article in English | MEDLINE | ID: mdl-33783261

ABSTRACT

The predominant assay detection methodologies used for enzyme inhibitor identification during early-stage drug discovery are fluorescence-based. Each fluorophore has a characteristic fluorescence decay, known as the fluorescence lifetime, that occurs throughout a nanosecond-to-millisecond timescale. The measurement of fluorescence lifetime as a reporter for biological activity is less common than fluorescence intensity, even though the latter has numerous issues that can lead to false-positive readouts. The confirmation of hit compounds as true inhibitors requires additional assays, cost, and time to progress from hit identification to lead drug-candidate optimization. To explore whether the use of fluorescence lifetime technology (FLT) can offer comparable benefits to label-free-based approaches such as RapidFire mass spectroscopy (RF-MS) and a superior readout compared to time-resolved fluorescence resonance energy transfer (TR-FRET), three equivalent assays were developed against the clinically validated tyrosine kinase 2 (TYK2) and screened against annotated compound sets. FLT provided a marked decrease in the number of false-positive hits when compared to TR-FRET. Further cellular screening confirmed that a number of potential inhibitors directly interacted with TYK2 and inhibited the downstream phosphorylation of the signal transducer and activator of transcription 4 protein (STAT4).


Subject(s)
Drug Discovery/methods , Drug Discovery/standards , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/standards , Fluorescent Dyes , TYK2 Kinase/antagonists & inhibitors , TYK2 Kinase/chemistry , Fluorescence Resonance Energy Transfer , High-Throughput Screening Assays , Mass Spectrometry , Reproducibility of Results , Sensitivity and Specificity
4.
SLAS Discov ; 25(2): 163-175, 2020 02.
Article in English | MEDLINE | ID: mdl-31875412

ABSTRACT

Malfunctions in the basic epigenetic mechanisms such as histone modifications, DNA methylation, and chromatin remodeling are implicated in a number of cancers and immunological and neurodegenerative conditions. Within GlaxoSmithKline (GSK) we have utilized a number of variations of the NanoBRET technology for the direct measurement of compound-target engagement within native cellular environments to drive high-throughput, routine structure-activity relationship (SAR) profiling across differing epigenetic targets. NanoBRET is a variation of the bioluminescence resonance energy transfer (BRET) methodology utilizing proteins of interest fused to either NanoLuc, a small, high-emission-intensity luciferase, or HaloTag, a modified dehalogenase enzyme that can be selectively labeled with a fluorophore. The combination of these two technologies has enabled the application of NanoBRET to biological systems such as epigenetic protein-protein interactions, which have previously been challenging. By synergizing target engagement assays with more complex primary cell phenotypic assays, we have been able to demonstrate compound-target selectivity profiles to enhance cellular potency and offset potential liability risks. Additionally, we have shown that in the absence of a robust, cell phenotypic assay, it is possible to utilize NanoBRET target engagement assays to aid chemistry in progressing at a higher scale than would have otherwise been achievable. The NanoBRET target engagement assays utilized have further shown an excellent correlation with more reductionist biochemical and biophysical assay systems, clearly demonstrating the possibility of using such assay systems at scale, in tandem with, or in preference to, lower-throughput cell phenotypic approaches.


Subject(s)
Biological Assay , Epigenesis, Genetic/genetics , Structure-Activity Relationship , Chromatin Assembly and Disassembly/genetics , DNA Methylation/genetics , Fluorescence Resonance Energy Transfer , Histone Code/genetics , Humans , Luciferases/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...