Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
PLoS One ; 12(1): e0169855, 2017.
Article in English | MEDLINE | ID: mdl-28072869

ABSTRACT

Polycomb repressive complex 2 (PRC2), a histone H3 lysine 27 methyltransferase, plays a key role in gene regulation and is a known epigenetics drug target for cancer therapy. The WD40 domain-containing protein EED is the regulatory subunit of PRC2. It binds to the tri-methylated lysine 27 of the histone H3 (H3K27me3), and through which stimulates the activity of PRC2 allosterically. Recently, we disclosed a novel PRC2 inhibitor EED226 which binds to the K27me3-pocket on EED and showed strong antitumor activity in xenograft mice model. Here, we further report the identification and validation of four other EED binders along with EED162, the parental compound of EED226. The crystal structures for all these five compounds in complex with EED revealed a common deep pocket induced by the binding of this diverse set of compounds. This pocket was created after significant conformational rearrangement of the aromatic cage residues (Y365, Y148 and F97) in the H3K27me3 binding pocket of EED, the width of which was delineated by the side chains of these rearranged residues. In addition, all five compounds interact with the Arg367 at the bottom of the pocket. Each compound also displays unique features in its interaction with EED, suggesting the dynamics of the H3K27me3 pocket in accommodating the binding of different compounds. Our results provide structural insights for rational design of novel EED binder for the inhibition of PRC2 complex activity.


Subject(s)
Enzyme Inhibitors/pharmacology , Molecular Docking Simulation , Polycomb Repressive Complex 2/antagonists & inhibitors , Sulfones/pharmacology , Triazoles/pharmacology , Animals , Binding Sites , Drug Discovery , Enzyme Inhibitors/chemistry , High-Throughput Screening Assays , Mice , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism , Quantitative Structure-Activity Relationship , Sulfones/chemistry , Triazoles/chemistry
2.
ACS Chem Biol ; 11(12): 3338-3346, 2016 12 16.
Article in English | MEDLINE | ID: mdl-27712055

ABSTRACT

Protein kinases are known for their highly conserved adenosine triphosphate (ATP)-binding site, rendering the discovery of selective inhibitors a major challenge. In theory, allosteric inhibitors can achieve high selectivity by targeting less conserved regions of the kinases, often with an added benefit of retaining efficacy under high physiological ATP concentration. Although often overlooked in favor of ATP-site directed approaches, performing a screen at high ATP concentration or stringent hit triaging with high ATP concentration offers conceptually simple methods of identifying inhibitors that bind outside the ATP pocket. Here, we applied the latter approach to the With-No-Lysine (K) (WNK) kinases to discover lead molecules for a next-generation antihypertensive that requires a stringent safety profile. This strategy yielded several ATP noncompetitive WNK1-4 kinase inhibitors, the optimization of which enabled cocrystallization with WNK1, revealing an allosteric binding mode consistent with the observed exquisite specificity for WNK1-4 kinases. The optimized compound inhibited rubidium uptake by sodium chloride cotransporter 1 (NKCC1) in HT29 cells, consistent with the reported physiology of WNK kinases in renal electrolyte handling.


Subject(s)
Allosteric Regulation/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Drug Discovery , HEK293 Cells , HT29 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Minor Histocompatibility Antigens/chemistry , Minor Histocompatibility Antigens/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Solute Carrier Family 12, Member 2/metabolism , WNK Lysine-Deficient Protein Kinase 1
3.
J Biomol Screen ; 21(10): 1075-1089, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27601436

ABSTRACT

One of the central questions in the characterization of enzyme inhibitors is determining the mode of inhibition (MOI). Classically, this is done with a number of low-throughput methods in which inhibition models are fitted to the data. The ability to rapidly characterize the MOI for inhibitors arising from high-throughput screening in which hundreds to thousands of primary inhibitors may need to be characterized would greatly help in lead selection efforts. Here we describe a novel method for determining the MOI of a compound without the need for curve fitting of the enzyme inhibition data. We provide experimental data to demonstrate the utility of this new high-throughput MOI classification method based on nonparametric analysis of the activity derived from a small matrix of substrate and inhibitor concentrations (e.g., from a 4S × 4I matrix). Lists of inhibitors from four different enzyme assays are studied, and the results are compared with the previously described IC50-shift method for MOI classification. The MOI results from this method are in good agreement with the known MOI and compare favorably with those from the IC50-shift method. In addition, we discuss some advantages and limitations of the method and provide recommendations for utilization of this MOI classification method.


Subject(s)
Enzyme Inhibitors/chemistry , Enzymes/chemistry , High-Throughput Screening Assays/methods , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/classification , Humans
4.
Nature ; 535(7610): 148-52, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27362227

ABSTRACT

The non-receptor protein tyrosine phosphatase SHP2, encoded by PTPN11, has an important role in signal transduction downstream of growth factor receptor signalling and was the first reported oncogenic tyrosine phosphatase. Activating mutations of SHP2 have been associated with developmental pathologies such as Noonan syndrome and are found in multiple cancer types, including leukaemia, lung and breast cancer and neuroblastoma. SHP2 is ubiquitously expressed and regulates cell survival and proliferation primarily through activation of the RAS­ERK signalling pathway. It is also a key mediator of the programmed cell death 1 (PD-1) and B- and T-lymphocyte attenuator (BTLA) immune checkpoint pathways. Reduction of SHP2 activity suppresses tumour cell growth and is a potential target of cancer therapy. Here we report the discovery of a highly potent (IC50 = 0.071 µM), selective and orally bioavailable small-molecule SHP2 inhibitor, SHP099, that stabilizes SHP2 in an auto-inhibited conformation. SHP099 concurrently binds to the interface of the N-terminal SH2, C-terminal SH2, and protein tyrosine phosphatase domains, thus inhibiting SHP2 activity through an allosteric mechanism. SHP099 suppresses RAS­ERK signalling to inhibit the proliferation of receptor-tyrosine-kinase-driven human cancer cells in vitro and is efficacious in mouse tumour xenograft models. Together, these data demonstrate that pharmacological inhibition of SHP2 is a valid therapeutic approach for the treatment of cancers.


Subject(s)
Neoplasms/drug therapy , Neoplasms/enzymology , Piperidines/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Allosteric Regulation/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Inhibitory Concentration 50 , MAP Kinase Signaling System/drug effects , Mice , Mice, Nude , Models, Molecular , Neoplasms/pathology , Oncogene Protein p21(ras)/metabolism , Piperidines/chemistry , Piperidines/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Stability/drug effects , Protein Structure, Tertiary/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Reproducibility of Results , Xenograft Model Antitumor Assays
5.
J Med Chem ; 59(17): 7773-82, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27347692

ABSTRACT

SHP2 is a nonreceptor protein tyrosine phosphatase (PTP) encoded by the PTPN11 gene involved in cell growth and differentiation via the MAPK signaling pathway. SHP2 also purportedly plays an important role in the programmed cell death pathway (PD-1/PD-L1). Because it is an oncoprotein associated with multiple cancer-related diseases, as well as a potential immunomodulator, controlling SHP2 activity is of significant therapeutic interest. Recently in our laboratories, a small molecule inhibitor of SHP2 was identified as an allosteric modulator that stabilizes the autoinhibited conformation of SHP2. A high throughput screen was performed to identify progressable chemical matter, and X-ray crystallography revealed the location of binding in a previously undisclosed allosteric binding pocket. Structure-based drug design was employed to optimize for SHP2 inhibition, and several new protein-ligand interactions were characterized. These studies culminated in the discovery of 6-(4-amino-4-methylpiperidin-1-yl)-3-(2,3-dichlorophenyl)pyrazin-2-amine (SHP099, 1), a potent, selective, orally bioavailable, and efficacious SHP2 inhibitor.


Subject(s)
Antineoplastic Agents/chemistry , Piperidines/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Pyrazines/chemistry , Pyrimidines/chemistry , Administration, Oral , Allosteric Regulation , Allosteric Site , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , Female , Heterografts , High-Throughput Screening Assays , Humans , Male , Mice, Inbred C57BL , Mice, Nude , Models, Molecular , Neoplasm Transplantation , Piperidines/chemical synthesis , Piperidines/pharmacokinetics , Piperidines/pharmacology , Protein Conformation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Pyrazines/chemical synthesis , Pyrazines/pharmacokinetics , Pyrazines/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Structure-Activity Relationship
6.
J Biomol Screen ; 19(5): 651-60, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24246376

ABSTRACT

Pilot testing of an assay intended for high-throughput screening (HTS) with small compound sets is a necessary but often time-consuming step in the validation of an assay protocol. When the initial testing concentration is less than optimal, this can involve iterative testing at different concentrations to further evaluate the pilot outcome, which can be even more time-consuming. Quantitative HTS (qHTS) enables flexible and rapid collection of assay performance statistics, hits at different concentrations, and concentration-response curves in a single experiment. Here we describe the qHTS process for pilot testing in which eight-point concentration-response curves are produced using an interplate asymmetric dilution protocol in which the first four concentrations are used to represent the range of typical HTS screening concentrations and the last four concentrations are added for robust curve fitting to determine potency/efficacy values. We also describe how these data can be analyzed to predict the frequency of false-positives, false-negatives, hit rates, and confirmation rates for the HTS process as a function of screening concentration. By taking into account the compound pharmacology, this pilot-testing paradigm enables rapid assessment of the assay performance and choosing the optimal concentration for the large-scale HTS in one experiment.


Subject(s)
High-Throughput Screening Assays/methods , Biological Assay , Cell Line , Dose-Response Relationship, Drug , False Positive Reactions , Genes, Reporter , Humans , Pilot Projects , Reproducibility of Results , Software
7.
Proc Natl Acad Sci U S A ; 109(52): 21360-5, 2012 Dec 26.
Article in English | MEDLINE | ID: mdl-23236167

ABSTRACT

Ezh2 (Enhancer of zeste homolog 2) protein is the enzymatic component of the Polycomb repressive complex 2 (PRC2), which represses gene expression by methylating lysine 27 of histone H3 (H3K27) and regulates cell proliferation and differentiation during embryonic development. Recently, hot-spot mutations of Ezh2 were identified in diffused large B-cell lymphomas and follicular lymphomas. To investigate if tumor growth is dependent on the enzymatic activity of Ezh2, we developed a potent and selective small molecule inhibitor, EI1, which inhibits the enzymatic activity of Ezh2 through direct binding to the enzyme and competing with the methyl group donor S-Adenosyl methionine. EI1-treated cells exhibit genome-wide loss of H3K27 methylation and activation of PRC2 target genes. Furthermore, inhibition of Ezh2 by EI1 in diffused large B-cell lymphomas cells carrying the Y641 mutations results in decreased proliferation, cell cycle arrest, and apoptosis. These results provide strong validation of Ezh2 as a potential therapeutic target for the treatment of cancer.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/pathology , Polycomb Repressive Complex 2/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Embryo, Mammalian/cytology , Enhancer of Zeste Homolog 2 Protein , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockout Techniques , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Methylation/drug effects , Mice , Mutation/genetics , Phenotype , Polycomb Repressive Complex 2/metabolism , Small Molecule Libraries/chemistry , Tumor Stem Cell Assay , Up-Regulation/drug effects , Up-Regulation/genetics
8.
Assay Drug Dev Technol ; 10(2): 179-86, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22192306

ABSTRACT

Histone methylation is a regulated feature of nucleosomes that can have an impact on gene expression. The methylation state of histone residues has also been found in recent years to be associated with various disorders. Tools for detecting methylation state changes are very useful for dissecting the function of these epigenetic marks. In this work, a sensitive homogeneous assay for histone demethylase activity at the H3K4 site has been developed in a time-resolved fluorescent resonance energy transfer assay format. The assay is based on the detection of the unmethylated H3 peptide by a fluorescent europium-chelate labeled monoclonal antibody binding specifically to the H3K4 site. The assay was validated for histone lysine-specific demethylase 1 and was demonstrated to be a suitable assay for inhibitor profiling and high-throughput screening.


Subject(s)
High-Throughput Screening Assays/methods , Histone Deacetylase Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Antibodies, Monoclonal/chemistry , Binding Sites , Chelating Agents/chemistry , Data Interpretation, Statistical , Europium , Fluorescence Resonance Energy Transfer , Histone Demethylases/chemistry , Horseradish Peroxidase/chemistry , Humans , Hydrogen Peroxide/chemistry , Indicators and Reagents , Substrate Specificity
9.
Bioorg Med Chem Lett ; 21(5): 1447-51, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21300545

ABSTRACT

The synthesis and preliminary studies of the SAR of novel 3,5-diarylazole inhibitors of Protein Kinase D (PKD) are reported. Notably, optimized compounds in this class have been found to be active in cellular assays of phosphorylation-dependant HDAC5 nuclear export, orally bioavailable, and highly selective versus a panel of additional putative histone deacetylase (HDAC) kinases. Therefore these compounds could provide attractive tools for the further study of PKD/HDAC5 signaling.


Subject(s)
Azoles/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Administration, Oral , Animals , Azoles/chemical synthesis , Azoles/chemistry , Azoles/pharmacokinetics , Biological Availability , Histone Deacetylases/metabolism , Inhibitory Concentration 50 , Molecular Structure , Protein Kinase C/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Rats , Rats, Sprague-Dawley , Signal Transduction , Structure-Activity Relationship
10.
J Chem Inf Model ; 50(12): 2067-78, 2010 Dec 27.
Article in English | MEDLINE | ID: mdl-21073183

ABSTRACT

The main goal of high-throughput screening (HTS) is to identify active chemical series rather than just individual active compounds. In light of this goal, a new method (called compound set enrichment) to identify active chemical series from primary screening data is proposed. The method employs the scaffold tree compound classification in conjunction with the Kolmogorov-Smirnov statistic to assess the overall activity of a compound scaffold. The application of this method to seven PubChem data sets (containing between 9389 and 263679 molecules) is presented, and the ability of this method to identify compound classes with only weakly active compounds (potentially latent hits) is demonstrated. The analysis presented here shows how methods based on an activity cutoff can distort activity information, leading to the incorrect activity assignment of compound series. These results suggest that this method might have utility in the rational selection of active classes of compounds (and not just individual active compounds) for followup and validation.


Subject(s)
High-Throughput Screening Assays/methods , Biological Assay , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical
11.
J Med Chem ; 53(15): 5400-21, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20684591

ABSTRACT

A novel 2,6-naphthyridine was identified by high throughput screen (HTS) as a dual protein kinase C/D (PKC/PKD) inhibitor. PKD inhibition in the heart was proposed as a potential antihypertrophic mechanism with application as a heart failure therapy. As PKC was previously identified as the immediate upstream activator of PKD, PKD vs PKC selectivity was essential to understand the effect of PKD inhibition in models of cardiac hypertrophy and heart failure. The present study describes the modification of the HTS hit to a series of prototype pan-PKD inhibitors with routine 1000-fold PKD vs PKC selectivity. Example compounds inhibited PKD activity in vitro, in cells, and in vivo following oral administration. Their effects on heart morphology and function are discussed herein.


Subject(s)
Aminopyridines/chemical synthesis , Naphthyridines/chemical synthesis , Protein Kinase C/antagonists & inhibitors , Active Transport, Cell Nucleus , Administration, Oral , Aminopyridines/pharmacokinetics , Aminopyridines/pharmacology , Animals , Antihypertensive Agents/chemical synthesis , Antihypertensive Agents/chemistry , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Cell Nucleus/metabolism , Histone Deacetylases/metabolism , Isoenzymes/antagonists & inhibitors , Male , Models, Molecular , Muscle Cells/drug effects , Muscle Cells/metabolism , Muscle Cells/pathology , Myocardium/metabolism , Myocardium/pathology , Naphthyridines/pharmacokinetics , Naphthyridines/pharmacology , Phosphorylation , Protein Binding , Rats , Rats, Inbred Dahl , Rats, Sprague-Dawley , Structure-Activity Relationship
12.
J Biomol Screen ; 15(6): 695-702, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20484097

ABSTRACT

Many attractive targets for therapeutic intervention are enzymes that catalyze biological reactions involving small molecules such as lipids, fatty acids, amino acid derivatives, nucleic acid derivatives, and cofactors. Some of the reactions are difficult to detect by methods commonly used in high-throughput screening (HTS) without specific radioactive or fluorescent labeling of substrates. In addition, there are instances when labeling has a detrimental effect on the biological response. Generally, applicable assay methodologies for detection of such reactions are thus required. Mass spectrometry (MS), being a label-free detection tool, has been actively pursued for assay detection in HTS in the past several years. The authors have explored the use of multiparallel liquid chromatography coupled with tandem mass spectrometry (LC/MS/MS) for high-throughput detection of biochemical reactions. In this report, we describe in detail the assay development and screening with a LC/MS-based system for inhibitors of human diacylglycerol acyltransferase (DGAT1) with a chemical library of approximately 800,000 compounds. Several strategies and process improvements have been investigated to overcome technical challenges such as data variation and throughput. Results indicated that, through these innovative approaches, the LC/MS-based screening method is both feasible and suitable for high-throughput primary screening.


Subject(s)
Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Enzyme Assays/methods , Enzyme Inhibitors/analysis , Enzyme Inhibitors/pharmacology , Mass Spectrometry/methods , Chromatography, Liquid , Diacylglycerol O-Acyltransferase/metabolism , High-Throughput Screening Assays , Humans , Reference Standards , Reproducibility of Results , Solvents/chemistry , Time Factors , Titrimetry
13.
Anal Chem ; 79(21): 8207-13, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17902631

ABSTRACT

High-throughput screening (HTS) is an important tool for finding active compounds to initiate medicinal chemistry programs in pharmaceutical discovery research. Traditional HTS methods rely on fluorescent or radiolabeled reagents and/or coupling assays to permit quantitation of enzymatic target inhibition or activation. Mass spectrometry-based high-throughput screening (MS-HTS) is an alternative that is not susceptible to the limitations imposed by labeling and coupling enzymes. MS-HTS offers a selective and sensitive analytical method for unlabeled substrates and products. Furthermore, method development times are reduced without the need to incorporate labels or coupling assays. MS-HTS also permits screening of targets that are difficult or impossible to screen by other techniques. For example, enzymes that are challenging to purify can lead to the nonspecific detection of structurally similar components of the impure enzyme or matrix of membraneous enzymes. The high selectivity of tandem mass spectrometry (MS/MS) enables these screens to proceed with low levels of background noise to sensitively discover interesting hits even with relatively weak activity. In this article, we describe three techniques that we have adapted for large-scale (approximately 175,000 sample) compound library screening, including four-way parallel multiplexed electrospray liquid chromatography tandem mass spectrometry (MUX-LC/MS/MS), four-way parallel staggered gradient liquid chromatography tandem mass spectrometry (LC/MS/MS), and eight-way staggered flow injection MS/MS following 384-well plate solid-phase extraction (SPE). These methods are capable of analyzing a 384-well plate in 37 min, with typical analysis times of less than 2 h. The quality of the MS-HTS approach is demonstrated herein with screening data from two large-scale screens.


Subject(s)
Combinatorial Chemistry Techniques , Pharmaceutical Preparations/analysis , Tandem Mass Spectrometry/methods , Chromatography, High Pressure Liquid/instrumentation , Chromatography, High Pressure Liquid/methods , Drug Evaluation, Preclinical/instrumentation , Drug Evaluation, Preclinical/methods , Reproducibility of Results , Sensitivity and Specificity , Solid Phase Extraction/instrumentation , Solid Phase Extraction/methods , Tandem Mass Spectrometry/instrumentation
14.
J Biomol Screen ; 10(7): 695-704, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16129776

ABSTRACT

Despite a large body of references on assay development, assay optimization, strategies, and methodologies for high-throughput screening (HTS), there have been few reports on investigations of the efficiency of primary screening in a systematic and quantitative manner for a typical HTS process. Recently, the authors investigated the primary hit comparison and the effect of measurement variability by screening a library of approximately 25,000 random compounds in multiple replicate tests in a nuclear receptor recruitment assay with 2 different assay detection technologies. In this report, we utilized these sets of multiple replicate screening data from a different perspective and conducted a systematic data analysis in order to gain some insights into the hit-finding efficiency of a typical primary screening process. Specifically, hit confirmation, false-positive (declaration) rates, and false-negative rates at different hit cutoff limits were explored and calculated from the 2 different assay formats. Results and analyses provided some quantitative estimation regarding the reliability and efficiency of the primary screening process. For the 2 assay formats tested in this report, the confirmation rate (activity repeated at or above a certain hit limit) was found to be 65% or above. It was also suggested that, at least in this case, applying some hit-selection strategies, it is possible to decrease the number of false-negative or false-positive hits without significantly increasing the efforts in primary screening.


Subject(s)
Receptors, Cytoplasmic and Nuclear/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , False Negative Reactions , False Positive Reactions , Fluorescence Resonance Energy Transfer/methods , Ligands , Receptors, Cytoplasmic and Nuclear/analysis , Receptors, Cytoplasmic and Nuclear/chemistry , Transcription Factors/chemistry , Transcription Factors/metabolism
15.
J Biomol Screen ; 10(6): 615-23, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16103416

ABSTRACT

Just-in-time cell supply for cell-based high-throughput screening (HTS) is frequently problematic. In addition to scheduling and logistical issues, quality issues and variability due to passage effect, cell cycle, or confluency contribute to day-to-day signal variability in the course of cell-based HTS campaigns. Cell division-arrest and cryopreservation technologies permit the use of cells as assay-ready reagents for HTS and other cell-based profiling and structure-activity studies. In this report, the authors compare division-arrested and dividing cells in 2 assay types that are dependent on movement of proteins within or through cell membranes: a receptor tyrosine kinase assay involving A431 cells responsive to epidermal growth factor, and a secretion reporter assay, which measures secretion of a reporter gene, secreted alkaline phosphatase. In both assays, dividing and division-arrested cells yielded similar basal and maximal signals at a given cell density. Similar IC50s were obtained for reference inhibitors in each assay, type in both dividing and division-arrested cells. In addition, for the secretion reporter assay, when comparing IC50s obtained from 44 compounds randomly chosen from a primary screening hit list, the rank order of potency obtained from dividing cells and division-arrested cells was essentially identical. Furthermore, the results show that, under certain assay conditions, data generated using division-arrested cells are less variable than those generated using dividing cells. In summary, the results suggest that, in many cases, division-arrested cells can substitute for dividing cells and offer certain advantages for cell-based assays.


Subject(s)
Drug Evaluation, Preclinical/methods , Drug Industry/methods , Alkaline Phosphatase/metabolism , Cell Division , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Chemistry, Pharmaceutical/methods , Dimethyl Sulfoxide/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Genes, Reporter , Humans , Inhibitory Concentration 50 , Phosphorylation , Receptor Protein-Tyrosine Kinases/metabolism , Reproducibility of Results
16.
J Biomol Screen ; 10(6): 581-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16103421

ABSTRACT

High-throughput screening (HTS) has grown rapidly in the past decade, with many advances in new assay formats, detection technologies, and laboratory automation. Recently, several studies have shown that the choice of assay technology used for the screening process is particularly important and can yield quite different primary screening outcomes. However, because the screening assays in these previous studies were performed in a single-point determination, it is not clear to what extent the difference observed in the screening results between different assay technologies is attributable to inherent assay variability and day-to-day measurement variation. To address this question, a nuclear receptor coactivator recruitment assay was carried out in 2 different assay formats, namely, AlphaScreen and time-resolved fluorescence resonance energy transfer, which probed the same biochemical binding events but with different detection technologies. For each assay format, 4 independent screening runs in a typical HTS setting were completed to evaluate the run-to-run screening variability. These multiple tests with 2 assay formats allow an unambiguous comparison between the discrepancies of different assay formats and the effects of the variability of assay and screening measurements on the screening outcomes. The results provide further support that the choice of assay format or technology is a critical factor in HTS assay development.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Transcription Factors/antagonists & inhibitors , Animals , Automation , Biotin/chemistry , DNA-Binding Proteins/metabolism , Drug Industry/instrumentation , Drug Industry/methods , Fluorescence Resonance Energy Transfer , Ligands , Miniaturization , Protein Structure, Tertiary , Rats , Receptors, Cytoplasmic and Nuclear , Reproducibility of Results , Research Design , Spectroscopy, Fourier Transform Infrared , Transcription Factors/metabolism
18.
J Steroid Biochem Mol Biol ; 81(3): 217-25, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12163133

ABSTRACT

The mechanism by which ligands of nuclear receptors show differential effects on gene transcription is not fully understood, but is believed to result in part from the preferential recruitment and/or displacement of coactivators and corepressors. We have explored the interaction of several known ligands and the nuclear receptor (peroxisome proliferator activated receptor alpha, PPARalpha) using scintillation proximity assay (SPA) and the interaction of LXXLL containing peptides derived from three coactivators (SRC-1, CBP and PGC-1) with PPARalpha in the presence of PPARalpha agonist ligands using fluorescence resonance energy transfer (FRET). The EC(50)s of the individual ligands for recruitment showed the same rank order regardless of the coactivator peptide used, with GW2331

Subject(s)
Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Cell Line , Cell Nucleus/metabolism , DNA, Complementary/metabolism , Energy Transfer , Escherichia coli/metabolism , Histone Acetyltransferases , Humans , Kinetics , Ligands , Nuclear Receptor Coactivator 1 , Peptides/chemistry , Peptides/metabolism , Plasmids/metabolism , Protein Binding , Proto-Oncogene Proteins c-myc/metabolism , Spectrophotometry , Transfection
19.
Anal Biochem ; 304(1): 63-9, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11969190

ABSTRACT

The peroxisome proliferator-activated receptors (PPARs) are nuclear receptors activated by fatty acids and their metabolites. The PPARdelta subtype is believed to be involved in lipoprotein regulation and may have a role in reverse cholesterol transport. While the range of biological roles of PPARdelta still remains unclear, it is of therapeutic interest in cardiovascular diseases. Here we report a homogeneous in vitro assay for studying ligand activation of PPARdelta. We surveyed a panel of peptides containing the LXXLL motifs derived from coactivator protein sequences. Peptides with the best response were used to develop a sensitive and homogeneous recruitment assay for PPARdelta. The optimized assay has a signal-to-background ratio of about 8:1 and an assay quality parameter Z'-factor value of 0.8. The assay signal generated is stable for hours to even overnight. This simple recruitment assay can provide agonist and/or antagonist information that cannot be assessed by receptor-binding assay, and can be used for characterization and screening of ligands that modulate the activation of PPARdelta.


Subject(s)
Receptors, Cytoplasmic and Nuclear/analysis , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/analysis , Transcription Factors/metabolism , Amino Acid Motifs , Amino Acid Sequence , Energy Transfer , Humans , In Vitro Techniques , Ligands , Molecular Sequence Data , Peptides/chemistry , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/metabolism , Spectrometry, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL
...