Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters











Publication year range
1.
Toxicol In Vitro ; 28(2): 171-81, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24189122

ABSTRACT

Drug-induced liver injury (DILI) is a major cause of failed drug development, withdrawal and restricted usage. Therefore screening assays which aid selection of candidate drugs with reduced propensity to cause DILI are required. We have investigated the toxicity of 144 drugs, 108 of which caused DILI, using assays identified in the literature as having some predictivity for hepatotoxicity. The validated assays utilised either HepG2 cells, HepG2 cells in the presence of rat S9 fraction or isolated human hepatocytes. All parameters were quantified by multiplexed and automated high content fluorescence microscopy, at appropriate time points after compound administration (4, 24 or 48h). The individual endpoint which identified drugs that caused DILI with greatest precision was maximal fold induction in CM-H2DFFDA staining in hepatocytes after 24h (41% sensitivity, 86% specificity). However, hierarchical clustering analysis of all endpoints provided the most sensitive identification of drugs which caused DILI (58% sensitivity, 75% specificity). We conclude that multi-parametric high content cell toxicity assays can enable in vitro detection of drugs that have high propensity to cause DILI in vivo but that many DILI compounds exhibit few in vitro signals when evaluated using these assays.


Subject(s)
Chemical and Drug Induced Liver Injury/pathology , Hepatocytes/pathology , Algorithms , Animals , Caspase 3/metabolism , Cell Count , Cell Line , Cell Survival , Cluster Analysis , Cryopreservation , Cytological Techniques/instrumentation , HSP70 Heat-Shock Proteins/metabolism , HSP72 Heat-Shock Proteins/metabolism , Hep G2 Cells , Histones/metabolism , Humans , Image Processing, Computer-Assisted , Lipid Metabolism , Phospholipids/metabolism , Primary Cell Culture , Rats , Subcellular Fractions/chemistry , Subcellular Fractions/metabolism
2.
J Biol Chem ; 286(51): 43951-43958, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22020937

ABSTRACT

Eukaryotic elongation factor-2 kinase (eEF2K) relays growth and stress signals to protein synthesis through phosphorylation and inactivation of eukaryotic elongation factor 2 (eEF2). 1-Benzyl-3-cetyl-2-methylimidazolium iodide (NH125) is a widely accepted inhibitor of mammalian eEF2K and an efficacious anti-proliferation agent against different cancer cells. It implied that eEF2K could be an efficacious anticancer target. However, eEF2K siRNA was ineffective against cancer cells including those sensitive to NH125. To test if pharmacological intervention differs from siRNA interference, we identified a highly selective small molecule eEF2K inhibitor A-484954. Like siRNA, A-484954 had little effect on cancer cell growth. We carefully examined the effect of NH125 and A-484954 on phosphorylation of eEF2, the known cellular substrate of eEF2K. Surprisingly, NH125 increased eEF2 phosphorylation, whereas A-484954 inhibited the phosphorylation as expected for an eEF2K inhibitor. Both A-484954 and eEF2K siRNA inhibited eEF2K and reduced eEF2 phosphorylation with little effect on cancer cell growth. These data demonstrated clearly that the anticancer activity of NH125 was more correlated with induction of eEF2 phosphorylation than inhibition of eEF2K. Actually, induction of eEF2 phosphorylation was reported to correlate with inhibition of cancer cell growth. We compared several known inducers of eEF2 phosphorylation including AMPK activators and an mTOR inhibitor. Interestingly, stronger induction of eEF2 phosphorylation correlated with more effective growth inhibition. We also explored signal transduction pathways leading to NH125-induced eEF2 phosphorylation. Preliminary data suggested that NH125-induced eEF2 phosphorylation was likely mediated through multiple pathways. These observations identified an opportunity for a new multipathway approach to anticancer therapies.


Subject(s)
Gene Expression Regulation, Neoplastic , Imidazoles/pharmacology , Peptide Elongation Factor 2/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation , Drug Design , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Phosphorylation , RNA, Small Interfering/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , eIF-2 Kinase/metabolism
3.
Assay Drug Dev Technol ; 9(6): 635-45, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21309712

ABSTRACT

α7 nicotinic acetylcholine receptors (nAChRs) are characterized by relatively low ACh sensitivity, rapid activation, and fast desensitization kinetics. ACh/agonist evoked currents at the α7 nAChR are transient, and, typically, calcium flux responses are difficult to detect using conventional fluorometric assay techniques. One approach to study interactions of agonists with the α7 nAChR is by utilizing positive allosteric modulators (PAMs). In this study, we demonstrate that inclusion of type II PAMs such as PNU-120596, but not type I, can enable detection of endogenous α7 nAChR-mediated calcium responses in human neuroblastoma (IMR-32) cells. Using this approach, we characterized the pharmacological profile of nicotine, epibatidine, choline, and other nAChR agonists such as PNU-282987, SSR-180711, GTS-21, OH-GTS21, tropisetron, NS6784, and A-582941. The rank order potency of agonists well correlated with α7 nAChR binding affinities measured in brain membranes. Inhibition of calcium response by methyllycaconitine in the presence of increasing concentrations of PNU-282987 or PNU-120596 revealed that the IC(50) value of methyllycaconitine was sensitive to varying concentrations of the agonist, but not that of the PAM. This format demonstrated the feasibility of this approach for high-throughput screening to identify small molecule, PAMs, which were further confirmed in electrophysiological assays of human α7 nAChR expressed in oocytes.


Subject(s)
High-Throughput Screening Assays/methods , Nicotinic Agonists/chemistry , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/chemistry , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/physiology , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Animals, Newborn , Cell Line, Tumor , Cells, Cultured , Dose-Response Relationship, Drug , Female , Humans , Neuroblastoma , Rats , Xenopus laevis , alpha7 Nicotinic Acetylcholine Receptor
4.
J Med Chem ; 51(20): 6571-80, 2008 Oct 23.
Article in English | MEDLINE | ID: mdl-18811133

ABSTRACT

A series of 2-aminopyrimidines was synthesized as ligands of the histamine H4 receptor (H4R). Working in part from a pyrimidine hit that was identified in an HTS campaign, SAR studies were carried out to optimize the potency, which led to compound 3, 4- tert-butyl-6-(4-methylpiperazin-1-yl)pyrimidin-2-ylamine. We further studied this compound by systematically modifying the core pyrimidine moiety, the methylpiperazine at position 4, the NH2 at position 2, and positions 5 and 6 of the pyrimidine ring. The pyrimidine 6 position benefited the most from this optimization, especially in analogs in which the 6- tert-butyl was replaced with aromatic and secondary amine moieties. The highlight of the optimization campaign was compound 4, 4-[2-amino-6-(4-methylpiperazin-1-yl)pyrimidin-4-yl]benzonitrile, which was potent in vitro and was active as an anti-inflammatory agent in an animal model and had antinociceptive activity in a pain model, which supports the potential of H 4R antagonists in pain.


Subject(s)
Histamine Antagonists/chemical synthesis , Histamine Antagonists/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Receptors, Histamine/metabolism , Animals , Biomarkers , Histamine Antagonists/chemistry , Humans , Hyperplasia/chemically induced , Hyperplasia/prevention & control , Ligands , Locomotion/drug effects , Mice , Molecular Structure , Pyrimidines/chemistry , Rats , Structure-Activity Relationship , Substrate Specificity
5.
J Biomol Screen ; 13(6): 527-37, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18566484

ABSTRACT

Prioritization of compounds based on human hepatotoxicity potential is currently a key unmet need in drug discovery, as it can become a major problem for several lead compounds in later stages of the drug discovery pipeline. The authors report the validation and implementation of a high-content multiparametric cytotoxicity assay based on simultaneous measurement of 8 key cell health indicators associated with nuclear morphology, plasma membrane integrity, mitochondrial function, and cell proliferation. Compounds are prioritized by (a) computing an in vitro safety margin using the minimum cytotoxic concentration (IC(20)) across all 8 indicators and cell-based efficacy data and (b) using the minimal cytotoxic concentration alone to take into account concentration of drug in tissues. Feasibility data using selected compounds, including quinolone antibiotics, thiazolidinediones, and statins, suggest the viability of this approach. To increase overall throughput of compound prioritization, the authors have identified the higher throughput, plate reader-based CyQUANT assay that is similar to the high-content screening (HCS) assay in sensitivity of measuring inhibition of cell proliferation. It is expected that the phenotypic output from the multiparametric HCS assay in combination with other highly sensitive approaches, such as microarray-based expression analysis of toxic signatures, will contribute to a better understanding and predictivity of human hepatotoxicity potential.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Hepatocytes/drug effects , Toxicity Tests/methods , Xenobiotics/toxicity , Biological Assay , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug-Related Side Effects and Adverse Reactions/classification , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Inhibitory Concentration 50 , Liver Neoplasms/pathology , Membrane Potential, Mitochondrial/drug effects , Predictive Value of Tests , Reproducibility of Results , Xenobiotics/classification
6.
Bioorg Med Chem Lett ; 18(11): 3173-7, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18479921

ABSTRACT

Halosalicylamide derivatives were identified from high-throughput screening as potent inhibitors of HCV NS5B polymerase. The subsequent structure and activity relationship revealed the absolute requirement of the salicylamide moiety for optimum activity. Methylation of either the hydroxyl group or the amide group of the salicylamide moiety abolished the activity while the substitutions on both phenyl rings are acceptable. The halosalicylamide derivatives were shown to be non-competitive with respect to elongation nucleotide and demonstrated broad genotype activity against genotype 1-3 HCV NS5B polymerases. Inhibitor competition studies indicated an additive binding mode to the initiation pocket that is occupied by the thiadiazine class of compounds and an additive binding mode to the elongation pocket that is occupied by diketoacids, but a mutually exclusive binding mode with respect to the allosteric thumb pocket that is occupied by the benzimidazole class of inhibitors. Therefore, halosalicylamides represent a novel class of allosteric inhibitors of HCV NS5B polymerase.


Subject(s)
Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Hepacivirus/drug effects , Salicylamides/chemical synthesis , Salicylamides/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Antiviral Agents/chemistry , Combinatorial Chemistry Techniques , Drug Design , Humans , Molecular Structure , Salicylamides/chemistry , Structure-Activity Relationship
7.
J Neurosci Res ; 86(10): 2214-26, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18438921

ABSTRACT

Activation of the Rho kinase (ROCK) pathway has been associated with inhibition of neurite regeneration and outgrowth in spinal cord injury. Growth-inhibitory substances present in the glial scar such as chondroitin sulfate proteoglycans (CSPGs) have been shown to create a nonpermissive environment for axon regeneration that results in growth cone collapse. In this study, an in vitro model was developed in nerve growth factor-differentiated PC12 cells where the Rho/ROCK pathway was modulated by CSPG. CSPG elicited concentration-dependent inhibition of neurite outgrowth in PC12 cells, which was reversed by ROCK inhibitors such as fasudil, dimethylfasudil, and Y27632. Further studies on the interactions of CSPG with ROCK inhibitors revealed that the modulation of ROCK by CSPG is noncompetitive in nature. It was also observed that ROCK inhibitors increased neurite outgrowth in undifferentiated PC12 cells, indicating constitutive ROCK activity in the cells. Analysis of signaling pathways demonstrated that the effect of CSPG increases the phosphorylation of myosin phosphatase, a substrate immediately downstream of ROCK activation. Fasudil, dimethylfasudil, and Y27632 inhibited the phosphorylation of myosin phosphatase induced by CSPG with rank order potencies comparable to those observed in the neurite outgrowth assay. In addition, ROCK inhibitors reversed cofilin phosphorylation induced by CSPG with similar rank order potencies. Taken together, our data demonstrate that the interaction of CSPG with the ROCK pathway involves downstream effectors of ROCK such as myosin phosphatase and cofilin.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Neurites/metabolism , Signal Transduction/physiology , rho-Associated Kinases/metabolism , Animals , Cofilin 1/metabolism , Enzyme Inhibitors/pharmacology , Myosin-Light-Chain Phosphatase/metabolism , Neurites/drug effects , PC12 Cells , Phosphorylation , Rats , Signal Transduction/drug effects
8.
Proc Natl Acad Sci U S A ; 105(1): 174-9, 2008 Jan 08.
Article in English | MEDLINE | ID: mdl-18172210

ABSTRACT

Hypoxia-inducible factor-1 (HIF-1) plays an essential role in tumor development and progression by regulating genes that are vital for proliferation, glycolysis, angiogenesis, and metastasis. To identify strategies of targeting the HIF-1 pathway, we screened a siRNA library against the entire druggable genome and a small-molecule library consisting of 691,200 compounds using a HIF-1 reporter cell line. Although the siRNA library screen failed to reveal any druggable targets, the small-molecule library screen identified a class of alkyliminophenylacetate compounds that inhibit hypoxia-induced HIF-1 reporter activity at single-digit nanomolar concentrations. These compounds were found to inhibit hypoxia but not deferoxamine-induced HIF-1alpha protein stabilization. Further analysis indicated that the alkyliminophenylacetate compounds likely inhibit the HIF-1 pathway through blocking the hypoxia-induced mitochondrial reactive oxygen species (ROS) production. Strikingly, all of the nonalkyliminophenylacetate HIF-1 inhibitors identified from the small-molecule library screen were also found to target mitochondria like the alkyliminophenylacetate compounds. The exclusive enrichment of mitochondria inhibitors from a library of >600,000 diverse compounds by using the HIF-1 reporter assay highlights the essential role of mitochondria in HIF-1 regulation. These results also suggest that targeting mitochondrial ROS production might be a highly effective way of blocking HIF-1 activity in tumors.


Subject(s)
Gene Library , Genomics/methods , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Mitochondria/metabolism , Small Molecule Libraries , Acetates/chemistry , Chemistry, Pharmaceutical/methods , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Models, Biological , Nuclear Proteins/chemistry , Pharmacogenetics/methods , RNA, Small Interfering/metabolism , Reactive Oxygen Species , Technology, Pharmaceutical/methods
9.
J Biomol Screen ; 12(7): 938-45, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17942786

ABSTRACT

A 384-well soft agar assay was developed to identify potential novel anticancer compounds. Normally used to detect cell transformation, the assay is used here to quantitate cell proliferation in a 3-dimensional (3-D) anchorage-independent format. HCC827 cells, which are highly sensitive to epithelial growth factor receptor (EGFR) tyrosine kinase inhibitors, were used to develop the method and a set of 9600 compounds used to validate the assay. Results were compared to a monolayer assay using the same compound set. The assay provides a robust method to discover compounds that could be missed using traditional monolayer formats.


Subject(s)
Antineoplastic Agents/chemistry , Enzyme Inhibitors/chemistry , Agar , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , Humans
10.
Anal Biochem ; 368(2): 156-67, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17644059

ABSTRACT

Maturation of the hepatitis C virus (HCV) polyprotein occurs by a series of proteolytic processes catalyzed by host cell proteases and the virally encoded proteases NS2 and NS3. Although several peptidomimetic inhibitors of NS3 protease have been published, only a few small molecule inhibitors have been reported. In an effort to improve screening efficiency by minimizing the spectral interference of various test compounds, a substrate that contains the longer wavelength fluorescence resonance energy transfer (FRET) pair, TAMRA/QSY-7, was devised. For the optimized substrate T-Abu-Q, with sequence Ac-Asp-Glu-Lys(TAMRA)-Glu-Glu-Abu-Psi(COO)Ala-Ser-Lys(QSY-7)-amide, the kinetic parameters with HCV NS3 protease are K(m)=30 microM, k(cat)=0.6s(-1), and k(cat)/K(m)=20,100s(-1)M(-1). We show that this substrate is suitable for inhibitor analysis and mechanistic studies so long as the substrate concentration is low enough (0.5 microM) to avoid complications from high inner filter effects. The substrate is especially useful with ultra-high-density screening formats, such as microarrayed compound screening technology, because there is less spectral interference from the compounds being tested than with more traditional (EDANS/DABCYL) FRET protease substrates. The merits of the new substrate, as well as potential applications of this FRET pair to other protease substrates, are discussed.


Subject(s)
Fluorescence Resonance Energy Transfer , Oligopeptides/chemistry , Viral Nonstructural Proteins/chemistry , Dose-Response Relationship, Drug , Fluorescent Dyes/chemistry , Hydrolysis , Kinetics , Oligopeptides/chemical synthesis , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Rhodamines/chemistry , Sensitivity and Specificity , Substrate Specificity , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism
11.
J Biomol Screen ; 12(1): 61-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17099245

ABSTRACT

Despite increasing use of cell-based assays in high-throughput screening (HTS) and lead optimization, one challenge is the adequate supply of high-quality cells expressing the target of interest. To this end, cell lines stably expressing targets are often established, maintained, and scaled up by cell culture. These steps require large investments of time and resources. Moreover, significant variability invariably occurs in cell yield, viability, expression levels, and target activities. In particular, stable expression of targets such as transient receptor potential A1 (TRPA1) causes toxicity, cell line degeneration, and loss of functional activity. Therefore, in an effort to identify TRPA1 antagonists, the authors used large-scale transiently transfected (LSTT) cells, enabling rapid establishment of assays suitable for HTS. LSTT cells, which could- be stored frozen for a long period of time (e.g., at least 42 weeks), retained TRPA1 protein expression and could be easily revived to produce robust and consistent signals in calcium influx and electrophysiological assays. Using cells from a single transfection, a chemical library of 700,000 compounds was screened, and TRPA1 antagonists were identified. The use of LSTT circumvented issues associated with stable TRPA1 expression, increased flexibility and consistency, and greatly reduced labor and cost. This approach will also be applicable to other pharmaceutical targets.


Subject(s)
Membrane Transport Modulators/analysis , Membrane Transport Modulators/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Transfection , Transient Receptor Potential Channels/agonists , Calcium/metabolism , Calcium Channels/metabolism , Clone Cells , Electrophysiology , Fluorescence , Freezing , Humans , Nerve Tissue Proteins/metabolism , TRPA1 Cation Channel , Transient Receptor Potential Channels/metabolism
12.
J Neurosci Methods ; 161(1): 47-54, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17083980

ABSTRACT

Fatty acid amide hydrolase (FAAH) is the enzyme responsible for the rapid degradation of fatty acid amides such as the endocannabinoid anandamide. Inhibition of FAAH activity has been suggested as a therapeutic approach for the treatment of chronic pain, depression and anxiety, through local activation of the cannabinoid receptor CB1. We have developed a high throughput screening assay for identification of FAAH inhibitors using a novel substrate, decanoyl 7-amino-4-methyl coumarin (D-AMC) that is cleaved by FAAH to release decanoic acid and the highly fluorescent molecule 7-amino-4-methyl coumarin (AMC). This assay gives an excellent signal window for measuring FAAH activity and, as a continuous assay, inherently offers improved sensitivity and accuracy over previously reported endpoint assays. The assay was validated using a panel of known FAAH inhibitors and purified recombinant human FAAH, then converted to a 384 well format and used to screen a large library of compounds (>600,000 compounds) to identify FAAH inhibitors. This screen identified numerous novel FAAH inhibitors of diverse chemotypes. These hits confirmed using a native FAAH substrate, anandamide, and had very similar rank order potency to that obtained using the D-AMC substrate. Collectively these data demonstrate that D-AMC can be successfully used to rapidly and effectively identify novel FAAH inhibitors for potential therapeutic use.


Subject(s)
Amidohydrolases/metabolism , Biological Assay/methods , Fluorescent Dyes/analysis , Automation/methods , Coumarins/pharmacokinetics , Fluorescent Dyes/chemistry , Humans , Indicators and Reagents/pharmacokinetics , Reproducibility of Results
13.
J Biomol Screen ; 10(1): 46-55, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15695343

ABSTRACT

Although fluorescence imaging plate reader (FLIPR)-based assays have been widely used in high-throughput screening, improved efficiencies in throughput and fidelity continue to be investigated. This study presents an offline compound addition protocol coupled with a testing strategy using mixtures of compounds in a 384-well format to identify antagonists of the neurokinin-1 receptor expressed in the human astrocytoma cell line (U373 MG). Substance P evoked a concentration-dependent increase in intracellular cellular Ca(2+) with an EC(50) value of 0.30 +/- 0.17 nM, which was inhibited by neurokinin-1 (NK1) antagonists L-733,060 and L-703,606. Test compounds, as mixtures of 10 compounds/well, were added to the cells offline using an automated dispensing unit and incubated prior to performing the assay in the FLIPR. Using the offline protocol, a higher through put of ~200,000 compounds was achieved in an 8-h working day, and several novel structural classes of compounds were identified as antagonists for the NK1 receptor. These studies demonstrate that the offline compound addition format using a mixture of compounds in a 384-well FLIPR assay provides an efficient platform for screening and identifying modulators for G-protein-coupled receptors.


Subject(s)
Drug Evaluation, Preclinical/instrumentation , Drug Evaluation, Preclinical/methods , Receptors, G-Protein-Coupled/metabolism , Cell Line, Tumor , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Humans , Inhibitory Concentration 50 , Kinetics , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Neurokinin-1/metabolism
14.
Anal Biochem ; 328(2): 131-8, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15113688

ABSTRACT

Bcl-xL is a member of the Bcl-2 family of proteins that are implicated to play a vital role in several diseases including cancer. Bcl-xL suppresses apoptosis; thus the inhibition of Bcl-xL function could restore the apoptotic process. To identify antagonists of Bcl-xL function, two ultra-high-throughput screens were implemented. An activity assay utilized fluorescence polarization, based on the binding of fluorescein-labeled peptide [the BH3 domain of BAD protein (F-Bad 6)] to Bcl-xL. A 384-well plate assay with mixtures of 10 drug compounds per well, combined with a fast plate reader, resulted in a throughput of 46,080 data points/day. Utilizing this screening format, 370,400 compounds were screened in duplicate and 425 inhibitors with an IC(50) below 100 microM were identified. The second assay format, affinity selection/mass spectrometry (ASMS), used ultrafiltration to separate Bcl-xL binders from nonbinders in mixtures of 2400 compounds. The bound species were subsequently separated from the protein and analyzed by flow injection electrospray mass spectrometry. Utilizing the ASMS format, 263,382 compounds were screened in duplicate and 29 binders with affinities below 100 microM were identified. Two novel classes of Bcl-xL inhibitors were identified by both methods and confirmed to bind (13)C-labeled Bcl-xL using heteronuclear magnetic resonance spectroscopy.


Subject(s)
Combinatorial Chemistry Techniques/methods , Pharmaceutical Preparations/analysis , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Apoptosis/genetics , Apoptosis/physiology , Binding, Competitive , Carrier Proteins/analysis , Carrier Proteins/metabolism , Dimethyl Sulfoxide/chemistry , Fluorescence Polarization , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Mass Spectrometry , Titrimetry , bcl-X Protein
15.
J Biomol Screen ; 9(2): 112-21, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15006134

ABSTRACT

A novel and innovative high-throughput screening assay was developed to identify both activators and inhibitors of AMP-activated protein kinase (AMPK) using microarrayed compound screening (microARCS) technology. Test compounds were arrayed at a density of 8640 on a polystyrene sheet, and the enzyme and peptide substrate were introduced into the assay by incorporating them into an agarose gel followed by placement of the gels onto the compound sheet. Adenosine triphosphate (ATP) was delivered via a membrane, and the phosphorylated biotinylated substrate was captured onto a streptavidin affinity membrane (SAM trade mark ). For detection, the SAM trade mark was removed, washed, and imaged on a phosphor screen overnight. A library of more than 700,000 compounds was screened using this format to identify novel activators and inhibitors of AMPK.


Subject(s)
Multienzyme Complexes/antagonists & inhibitors , Oligonucleotide Array Sequence Analysis/methods , Protein Serine-Threonine Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases , Adenosine Triphosphate/analysis , Animals , Miniaturization , Multienzyme Complexes/analysis , Protein Serine-Threonine Kinases/analysis , Rats , Sensitivity and Specificity , Time Factors
16.
J Biomol Screen ; 9(1): 3-11, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15006143

ABSTRACT

The authors report the development of a high-throughput screen for inhibitors of Streptococcus pneumoniae transcription and translation (TT) using a luciferase reporter, and the secondary assays used to determine the biochemical spectrum of activity and bacterial specificity. More than 220,000 compounds were screened in mixtures of 10 compounds per well, with 10,000 picks selected for further study. False-positive hits from inhibition of luciferase activity were an extremely common artifact. After filtering luciferase inhibitors and several known classes of antibiotics, approximately 50 hits remained. These compounds were examined for their ability to inhibit Escherichia coli TT, uncoupled S. pneumoniae translation or transcription, rabbit reticulocyte translation, and in vitro toxicity in human and bacterial cells. One of these compounds had the desired profile of broad-spectrum biochemical activity in bacteria and selectivity versus mammalian biochemical and whole-cell assays.


Subject(s)
Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests/methods , Protein Biosynthesis , Streptococcus pneumoniae/drug effects , Transcription, Genetic , Anti-Bacterial Agents/adverse effects , Base Sequence , Cell Line, Tumor , DNA, Bacterial , Genes, Reporter , Humans , Luciferases/genetics , Molecular Sequence Data , Streptococcus pneumoniae/genetics
17.
J Biomol Screen ; 9(1): 12-21, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15006144

ABSTRACT

Protein tyrosine kinases play critical roles in cell signaling and are considered attractive targets for drug discovery. The authors have applied microARCS (microarrayed compound screening) technology to develop a high-throughput screen for finding inhibitors of the p56lck tyrosine kinase. Initial assay development was performed in a homogeneous time-resolved (LANCE) format in 96-well microplates and then converted into the gel-based microARCS format. The microARCS methodology is a well-less screening format in which 8640 compounds are arrayed on a microplate-sized piece of polystyene and subsequently assayed by placing reagents cast in agarose gels in contact with these compound sheets. A blotting paper soaked with adenosine triphosphate is applied on the gel to initiate the kinase reaction in the gel. Using this screening methodology, 300,000 compounds were screened in less than 40 h. Substantial reagent reduction was achieved by converting this tyrosine kinase assay from a 96-well plate assay to microARCS, resulting in significant cost savings.


Subject(s)
Enzyme Inhibitors/pharmacology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/antagonists & inhibitors , Oligonucleotide Array Sequence Analysis , Electrophoresis, Agar Gel , Substrate Specificity
18.
Anal Biochem ; 321(2): 192-201, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14511684

ABSTRACT

The identification of agonist and antagonist leads for G-protein-coupled receptors (GPCRs) is of critical importance to the pharmaceutical and biotechnology industries. We report on the utilization of a novel, high-density, well-less screening platform known as microarrayed compound screening microARCS) that tests 8640 compounds in the footprint of a standard microtiter plate for the identification of novel agonists for a specific G-protein-coupled receptor. Although receptors coupled to the G alpha(q) protein can readily be assessed by fluorescence-based Ca(2+) release measurements, many GPCRs that are coupled to G alpha(s) or G alpha(i/o) proteins are not amenable to functional evaluation in such a high-throughput manner. In this study, the human dopamine D(4.4) receptor, which normally couples through the G alpha(i/o) protein to inhibit adenylate cyclase and to reduce levels of intracellular cAMP, was coupled to intracellular Ca(2+) release by stably coexpressing this receptor with a chimeric G(alpha qo5) protein in HEK-293 cells. In microARCS format, the cells expressing D(4.4) receptor and G alpha(qo5) protein were preloaded with fluo-4, cast into a 1% agarose gel, placed above the compound sheets, and imaged successively using a ViewLux charge-coupled device imaging system. Dopamine and other agonists evoked an increase in fluorescence response that appeared as bright spots in a time- and concentration-dependent manner. Utilizing this technology, a library of 260,000 compounds was rapidly screened and led to the identification of several novel agonists. These agonists were further characterized using a fluorometric imaging plate reader assay. Excellent confirmation rates coupled with enhanced efficiency and throughput enable microARCS to serve as an alternative platform for the screening and identification of novel GPCR agonists.


Subject(s)
Protein Array Analysis , Receptors, Dopamine D2/agonists , Apomorphine/pharmacology , Calcium Signaling , Cell Line , Dopamine/analysis , Dopamine/pharmacology , Drug Evaluation, Preclinical , Fluorometry , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Quinpirole/pharmacology , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/analysis , Receptors, Calcium-Sensing/metabolism , Receptors, Dopamine D2/analysis , Receptors, Dopamine D2/genetics , Receptors, Dopamine D4
19.
J Biomol Screen ; 8(3): 273-82, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12857381

ABSTRACT

Microarrayed compound screening format (muARCS) is a novel high-throughput screening technology that uses agarose matrices to integrate various biochemical or biological reagents in the assay. To evaluate the feasibility of using the muARCS technology for nucleic acid polymerization assays, the authors developed HIV reverse transcription (RT) and E1-dependent human papillomavirus (HPV) replication assays in this format. HIV RT is an RNA-dependent DNA polymerase, whereas HPV E1 is a DNA helicase. To ensure the efficient capture of the nucleic acid polymerization reaction and to minimize the nonspecific binding, the authors used a SAM(2) biotin capture membrane in the assay. In both studies, the nucleic acid substrate was biotinylated on one end and was bound to the SAM(2) membrane. A low melting-point agarose gel containing the rest of the reaction components was first placed on a polystyrene sheet spotted with compounds to allow passive diffusion of the compounds into the gel. The gel was removed from the compound sheet and applied to the SAM(2) membrane with the immobilized nucleic acid template to initiate the polymerization. After the incubation, the membrane was washed with a high-salt buffer and exposed for imaging. Potential inhibitors can be seen as white spots on a dark background. The sensitivity for the known inhibitors appears to be comparable in muARCS as in a traditional 96-well plate assay. The methodology described in this paper further expands the applications of muARCS technology.


Subject(s)
Biotin/pharmacology , Oligonucleotide Array Sequence Analysis/methods , Alkynes , Anti-HIV Agents/pharmacology , Benzoxazines , Biotin/chemistry , Biotinylation , Cyclopropanes , DNA/chemistry , Dose-Response Relationship, Drug , HIV Reverse Transcriptase/genetics , Inhibitory Concentration 50 , Nevirapine/pharmacology , Nucleic Acids/chemistry , Oxazines/pharmacology , Plasmids/metabolism , RNA/chemistry , RNA-Directed DNA Polymerase/chemistry , Sensitivity and Specificity , Templates, Genetic
20.
J Biomol Screen ; 8(3): 324-31, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12857386

ABSTRACT

Eotaxin, an inducer of eosinophil migration and activation, exerts its activity by binding to CCR3, the C-C chemokine receptor 3. An inhibitor of the eotaxin-CCR3 binding interaction may have potential as an anti-inflammatory drug for treatment of asthma, parasitic infections, and allergic disorders. A radioligand binding assay was developed using HEK cells transfected with CCR3, with (125)I eotaxin as the ligand. Whole cells grown on polylysine-coated plates were used as the receptor source for the screen. Screening of more than 200,000 compounds with this assay yielded a number of screening hits, and of these, 2 active novel antagonists were identified. These compounds showed inhibitory effects on eosinophil chemotaxis in both in vitro and in vivo assays.


Subject(s)
Biochemistry/methods , Receptors, Chemokine/chemistry , Receptors, Chemokine/metabolism , Animals , Calcium/metabolism , Cell Line , Cell Membrane/metabolism , Cell Movement , Chemokine CCL11 , Chemokines, CC/chemistry , Chemokines, CC/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Eosinophils/metabolism , Humans , Ligands , Mice , Mice, Inbred BALB C , Models, Chemical , Polylysine/chemistry , Protein Binding , Radioligand Assay , Receptors, CCR3 , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL