Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 372(3): 267-276, 2020 03.
Article in English | MEDLINE | ID: mdl-31857349

ABSTRACT

Heterologous sensitization of adenylyl cyclase (AC) is revealed as enhanced or exaggerated AC/cAMP signaling that occurs following persistent activation of Gα i/o-coupled receptors. This paradoxical phenomenon was discovered more than 40 years ago and was proposed as a cellular mechanism to explain the adaptive changes that occur following chronic exposure to drugs of abuse. However, the underlying molecular mechanisms of heterologous sensitization of AC remain largely unknown. In the present study, we performed a genome-wide cell-based RNA interference screen as an unbiased approach to identify genes associated with heterologous sensitization of AC. Following a series of validation and confirmation assays, three genes that form an E3 ligase complex, cullin3 (CUL3), neural precursor-cell-expressed and developmentally downregulated 8 (NEDD8), and really interesting new gene (RING)-box protein 1 (RBX1), were identified as specific modulators of heterologous sensitization of AC. Furthermore, based on the downstream actions of these genes, we evaluated the activity of proteasome inhibitors as well as the specific NEDD8-activating enzyme inhibitor, MLN4924 (Pevonedistat), in AC sensitization. We demonstrate that MG-132 and bortezomib treatments could mimic the inhibitory effects observed with gene knockdown, and MLN4924 was potent and efficacious in blocking the development of heterologous sensitization of endogenous and recombinant AC isoforms, including AC1, AC2, AC5, and AC6. Together, by using genetic and pharmacological approaches, we identified, for the first time, cullin3-RING ligases and the protein degradation pathway as essential modulators for heterologous sensitization of AC. SIGNIFICANCE STATEMENT: Through a genome-wide cell-based RNA interference screening, we identified three genes that form an E3 ligase complex, cullin3, neural precursor-cell-expressed and developmentally downregulated 8 (NEDD8), and really interesting new gene-box protein 1, as specific modulators of heterologous sensitization of AC. The effect of cullin3, NEDD8, or really interesting new gene-box protein 1 small interfering RNAs on heterologous sensitization was recapitulated by proteasome inhibitors, MG132 and bortezomib, and the specific NEDD8-activating enzyme inhibitor, MLN4924. These results suggest a novel hypothesis in which protein degradation is involved in the sensitization of AC signaling that occurs following chronic activation of Gαi/o-coupled receptors.


Subject(s)
Adenylyl Cyclases/metabolism , Carrier Proteins/genetics , Cullin Proteins/genetics , NEDD8 Protein/genetics , Ubiquitin-Protein Ligases/genetics , Adenylyl Cyclase Inhibitors/pharmacology , Adenylyl Cyclases/genetics , Cell Survival/drug effects , Cyclic AMP/metabolism , Cyclopentanes/pharmacology , Enzyme Activation , Gene Knockdown Techniques , Genome-Wide Association Study , HEK293 Cells , Humans , Pyrimidines/pharmacology , RNA, Small Interfering , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Signal Transduction
2.
Clin Cancer Res ; 24(24): 6509-6522, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30185422

ABSTRACT

PURPOSE: Testosterone suppression in prostate cancer is limited by serious side effects and resistance via restoration of androgen receptor (AR) functionality. ELK1 is required for AR-dependent growth in various hormone-dependent and castration-resistant prostate cancer models. The amino-terminal domain of AR docks at two sites on ELK1 to coactivate essential growth genes. This study explores the ability of small molecules to disrupt the ELK1-AR interaction in the spectrum of prostate cancer, inhibiting AR activity in a manner that would predict functional tumor selectivity. EXPERIMENTAL DESIGN: Small-molecule drug discovery and extensive biological characterization of a lead compound. RESULTS: We have discovered a lead molecule (KCI807) that selectively disrupts ELK1-dependent promoter activation by wild-type and variant ARs without interfering with ELK1 activation by ERK. KCI807 has an obligatory flavone scaffold and functional hydroxyl groups on C5 and C3'. KCI807 binds to AR, blocking ELK1 binding, and selectively blocks recruitment of AR to chromatin by ELK1. KCI807 primarily affects a subset of AR target growth genes selectively suppressing AR-dependent growth of prostate cancer cell lines with a better inhibitory profile than enzalutamide. KCI807 also inhibits in vivo growth of castration/enzalutamide-resistant cell line-derived and patient-derived tumor xenografts. In the rodent model, KCI807 has a plasma half-life of 6 hours, and maintenance of its antitumor effect is limited by self-induced metabolism at its 3'-hydroxyl. CONCLUSIONS: The results offer a mechanism-based therapeutic paradigm for disrupting the AR growth-promoting axis in the spectrum of prostate tumors while reducing global suppression of testosterone actions. KCI807 offers a good lead molecule for drug development.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Androgen Receptor Antagonists/chemistry , Androgen Receptor Antagonists/therapeutic use , Animals , Antineoplastic Agents, Hormonal/chemistry , Antineoplastic Agents, Hormonal/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Drug Discovery/methods , Drug Screening Assays, Antitumor , Gene Expression Profiling , High-Throughput Screening Assays , Humans , Male , Mice , Promoter Regions, Genetic , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Binding , Structure-Activity Relationship , Xenograft Model Antitumor Assays , ets-Domain Protein Elk-1/metabolism
3.
JCI Insight ; 3(14)2018 07 26.
Article in English | MEDLINE | ID: mdl-30046004

ABSTRACT

Soluble cytokine receptors function as decoy receptors to attenuate cytokine-mediated signaling and modulate downstream cellular responses. Dysregulated overproduction of soluble receptors can be pathological, such as soluble ST2 (sST2), a prognostic biomarker in cardiovascular diseases, ulcerative colitis, and graft-versus-host disease (GVHD). Although intervention using an ST2 antibody improves survival in murine GVHD models, sST2 is a challenging target for drug development because it binds to IL-33 via an extensive interaction interface. Here, we report the discovery of small-molecule ST2 inhibitors through a combination of high-throughput screening and computational analysis. After in vitro and in vivo toxicity assessment, 3 compounds were selected for evaluation in 2 experimental GVHD models. We show that the most effective compound, iST2-1, reduces plasma sST2 levels, alleviates disease symptoms, improves survival, and maintains graft-versus-leukemia activity. Our data suggest that iST2-1 warrants further optimization to develop treatment for inflammatory diseases mediated by sST2.


Subject(s)
Drug Discovery , Interleukin-1 Receptor-Like 1 Protein/drug effects , Interleukin-1 Receptor-Like 1 Protein/metabolism , Proteomics , Receptors, Cytokine/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biomarkers , Cell Line, Tumor , Computational Biology , Drug Evaluation, Preclinical , Graft vs Host Disease , High-Throughput Screening Assays , Interleukin-33/metabolism , Leukemia/drug therapy , Mice , Models, Animal , Stem Cell Transplantation
4.
SLAS Discov ; 23(1): 47-54, 2018 01.
Article in English | MEDLINE | ID: mdl-28686847

ABSTRACT

microRNAs (miRNAs) are small gene regulatory RNAs, and their expression has been found to be dysregulated in a number of human diseases. To facilitate the discovery of small molecules capable of selectively modulating the activity of a specific miRNA, we have utilized new high-throughput screening technology targeting Dicer-mediated pre-miRNA maturation. Pilot screening of ~50,000 small molecules and ~33,000 natural product extract libraries against pre-miR-21 processing indicated the potential of our assay for this goal, yielding a campaign Z' factor of 0.52 and an average plate signal-to-background (S/B) ratio of 13. Using two-dimensional screening against a second pre-miRNA, pre-let-7d, we evaluated the selectivity of confirmed hits. The results presented demonstrate how high-throughput screening can be used to identify selective small molecules for a target RNA.


Subject(s)
Drug Discovery/methods , Gene Expression Regulation/drug effects , High-Throughput Screening Assays , Ligands , MicroRNAs/genetics , RNA Precursors/genetics , Small Molecule Libraries , MicroRNAs/chemistry , Molecular Structure , RNA Precursors/chemistry , Reproducibility of Results , Workflow
5.
Brain ; 139(11): 2891-2908, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27645800

ABSTRACT

No disease-modifying treatment exists for the fatal neurodegenerative polyglutamine disease known both as Machado-Joseph disease and spinocerebellar ataxia type 3. As a potential route to therapy, we identified small molecules that reduce levels of the mutant disease protein, ATXN3. Screens of a small molecule collection, including 1250 Food and Drug Administration-approved drugs, in a novel cell-based assay, followed by secondary screens in brain slice cultures from transgenic mice expressing the human disease gene, identified the atypical antipsychotic aripiprazole as one of the hits. Aripiprazole increased longevity in a Drosophila model of Machado-Joseph disease and effectively reduced aggregated ATXN3 species in flies and in brains of transgenic mice treated for 10 days. The aripiprazole-mediated decrease in ATXN3 abundance may reflect a complex response culminating in the modulation of specific components of cellular protein homeostasis. Aripiprazole represents a potentially promising therapeutic drug for Machado-Joseph disease and possibly other neurological proteinopathies.


Subject(s)
Antipsychotic Agents/therapeutic use , Aripiprazole/therapeutic use , Ataxin-3/metabolism , Machado-Joseph Disease/drug therapy , Machado-Joseph Disease/metabolism , Mutant Proteins/drug effects , Animals , Animals, Genetically Modified , Ataxin-3/genetics , Brain/drug effects , Brain/metabolism , Brain/ultrastructure , Disease Models, Animal , Drosophila , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , HEK293 Cells/drug effects , HEK293 Cells/metabolism , HEK293 Cells/ultrastructure , Humans , Machado-Joseph Disease/genetics , Mice , Mutant Proteins/metabolism , Nerve Tissue Proteins/metabolism , Organ Culture Techniques , Peptides/genetics , Piperidines/pharmacology , Pyrans/pharmacology , Pyrazoles/pharmacology
6.
ACS Chem Biol ; 11(11): 3084-3092, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27622287

ABSTRACT

Inorganic pyrophosphatase (PPiase) is an essential enzyme that hydrolyzes inorganic pyrophosphate (PPi), driving numerous metabolic processes. We report a discovery of an allosteric inhibitor (2,4-bis(aziridin-1-yl)-6-(1-phenylpyrrol-2-yl)-s-triazine) of bacterial PPiases. Analogues of this lead compound were synthesized to target specifically Mycobacterium tuberculosis (Mtb) PPiase (MtPPiase). The best analogue (compound 16) with a Ki of 11 µM for MtPPiase is a species-specific inhibitor. Crystal structures of MtPPiase in complex with the lead compound and one of its analogues (compound 6) demonstrate that the inhibitors bind in a nonconserved interface between monomers of the hexameric MtPPiase in a yet unprecedented pairwise manner, while the remote conserved active site of the enzyme is occupied by a bound PPi substrate. Consistent with the structural studies, the kinetic analysis of the most potent inhibitor has indicated that it functions uncompetitively, by binding to the enzyme-substrate complex. The inhibitors appear to allosterically lock the active site in a closed state causing its dysfunctionalization and blocking the hydrolysis. These inhibitors are the first examples of allosteric, species-selective inhibitors of PPiases, serving as a proof-of-principle that PPiases can be selectively targeted.


Subject(s)
Enzyme Inhibitors/pharmacology , Inorganic Pyrophosphatase/antagonists & inhibitors , Mycobacterium tuberculosis/enzymology , Allosteric Regulation , Crystallography, X-Ray , Drug Discovery , Enzyme Inhibitors/chemistry , Inorganic Pyrophosphatase/metabolism , Molecular Structure
7.
ACS Med Chem Lett ; 6(8): 877-81, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26288688

ABSTRACT

As part of the International Cooperative Biodiversity Groups (ICBG) Program, we were interested in identifying biologically active unfolded protein response (UPR) inducing compounds from marine microorganisms isolated from Costa Rican biota. With this aim in mind we have now generated more than 33,000 unique prefractionated natural product extracts from marine and terrestrial organisms that have been submitted to the Center of Chemical Genomics (CCG) at the University of Michigan for high throughput screening (HTS). An effective complementary cell-based assay to identify novel modulators of UPR signaling was used for screening extracts. Active fractions were iteratively subjected to reverse-phase HPLC chromatographic analysis, and together with lobophorin A, B, E, and F (1-4), three new lobophorin congeners, designated as CR1 (5), CR2 (6), and CR3 (7) were isolated. Herein, we report that secondary assays revealed that the new lobophorins induced UPR-associated gene expression, inhibited oral squamous cell carcinoma cell growth, and led to UPR-dependent cell death in murine embryonic fibroblast (MEF) cells.

8.
Chem Biol Drug Des ; 86(6): 1331-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26147927

ABSTRACT

Novel antimicrobials that effectively inhibit bacterial growth are essential to fight the growing threat of antibiotic resistance. A promising target is the bacterial ribosome, a 2.5 MDa organelle susceptible to several biorthogonal modes of action used by different classes of antibiotics. To promote the discovery of unique inhibitors, we have miniaturized a coupled transcription/translation assay using E. coli and applied it to screen a natural product library of ~30 000 extracts. We significantly reduced the scale of the assay to 2 µL in a 1536-well plate format and decreased the effective concentration of costly reagents. The improved assay returned 1327 hits (4.6% hit rate) with %CV and Z' values of 8.5% and 0.74, respectively. This assay represents a significant advance in molecular screening, both in miniaturization and its application to a natural product extract library, and we intend to apply it to a broad array of pathogenic microbes in the search for novel anti-infective agents.


Subject(s)
Biological Products/pharmacology , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Anti-Infective Agents/pharmacology , Escherichia coli/drug effects , Escherichia coli/genetics , Luciferases/genetics , Miniaturization/methods , Protein Biosynthesis/drug effects , Ribosomes/drug effects , Ribosomes/genetics , Small Molecule Libraries , Transcription, Genetic/drug effects
9.
Antimicrob Agents Chemother ; 59(7): 3880-6, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25870073

ABSTRACT

Campylobacter jejuni is a major cause of food-borne illness due to its ability to reside within the gastrointestinal tracts of chickens. Multiple studies have identified the flagella of C. jejuni as a major determinant of chicken colonization. An inhibitor screen of approximately 147,000 small molecules was performed to identify compounds that are able to inhibit flagellar expression in a reporter strain of C. jejuni. Several compounds that modestly inhibited motility of wild-type C. jejuni in standard assays were identified, as were a number of small molecules that robustly inhibited C. jejuni growth, in vitro. Examination of similar bacterial screens found that many of these small molecules inhibited only the growth of C. jejuni. Follow-up assays demonstrated inhibition of other strains of C. jejuni and Campylobacter coli but no inhibition of the closely related Helicobacter pylori. The compounds were determined to be bacteriostatic and nontoxic to eukaryotic cells. Preliminary results from a day-of-hatch chick model of colonization suggest that at least one of the compounds demonstrates promise for reducing Campylobacter colonization loads in vivo, although further medicinal chemistry may be required to enhance bioavailability.


Subject(s)
Anti-Bacterial Agents/pharmacology , Campylobacter jejuni/drug effects , Campylobacter jejuni/metabolism , Flagella/drug effects , Animals , Anti-Bacterial Agents/toxicity , Campylobacter coli/drug effects , Campylobacter jejuni/growth & development , Cell Survival/drug effects , Chick Embryo , Chickens/microbiology , Dose-Response Relationship, Drug , Eukaryotic Cells/drug effects , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Helicobacter pylori/drug effects , High-Throughput Screening Assays , Microbial Sensitivity Tests , Poultry Diseases/microbiology , Small Molecule Libraries , Species Specificity
10.
J Biomol Screen ; 20(5): 673-80, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25550354

ABSTRACT

Alphaviruses are a prominent class of reemergent pathogens due to their globally expanding ranges, potential for lethality, and possible use as bioweapons. The absence of effective treatments for alphaviruses highlights the need for innovative strategies to identify antiviral agents. Primary screens that use noninfectious self-replicating RNAs, termed replicons, have been used to identify potential antiviral compounds for alphaviruses. Only inhibitors of viral genome replication, however, will be identified using replicons, which excludes many other druggable steps in the viral life cycle. To address this limitation, we developed a western equine encephalitis virus pseudoinfectious particle system that reproduces several crucial viral life cycle steps in addition to genome replication. We used this system to screen a library containing ~26,000 extracts derived from marine microbes, and we identified multiple bacterial strains that produce compounds with potential antiviral activity. We subsequently used pseudoinfectious particle and replicon assays in parallel to counterscreen candidate extracts, and followed antiviral activity during biochemical fractionation and purification to differentiate between inhibitors of viral entry and genome replication. This novel process led to the isolation of a known alphavirus entry inhibitor, bafilomycin, thereby validating the approach for the screening and identification of potential antiviral compounds.


Subject(s)
Alphavirus/drug effects , Alphavirus/physiology , Antiviral Agents/pharmacology , Biological Products/pharmacology , Drug Discovery/methods , Animals , Antiviral Agents/chemistry , Biological Products/chemistry , Cell Line , Dose-Response Relationship, Drug , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Microbial Sensitivity Tests/methods , Reproducibility of Results , Small Molecule Libraries , Virus Replication/drug effects
11.
mBio ; 5(2): e01089-13, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24570372

ABSTRACT

The urinary tract is one of the most common sites of infection in humans, and uropathogenic Escherichia coli (UPEC) is the main causative agent of urinary tract infections. Bacteria colonizing the urinary tract face extremely low iron availability. To counteract this, UPEC expresses a wide variety of iron acquisition systems. To exploit iron acquisition in UPEC as a global target for small-molecule inhibition, we developed and carried out a whole-cell growth-based high throughput screen of 149,243 compounds. Our primary assay was carried out under iron-limiting conditions. Hits in the primary screen were assayed using two counterscreens that ruled out iron chelators and compounds that inhibit growth by means other than inhibition of iron acquisition. We determined dose-response curves under two different iron conditions and purchased fresh compounds for selected hits. After retesting dose-response relationships, we identified 16 compounds that arrest growth of UPEC only under iron-limiting conditions. All compounds are bacteriostatic and do not inhibit proton motive force. A loss-of-target strategy was employed to identify the cellular target of these inhibitors. Two compounds lost inhibitory activity against a strain lacking TonB and were shown to inhibit irreversible adsorption of a TonB-dependent bacteriophage. Our results validate iron acquisition as a target for antibacterial strategies against UPEC and identify TonB as one of the cellular targets. IMPORTANCE Half of women will suffer at least one episode of urinary tract infection (UTI) during their lifetime. The current treatment for UTI involves antibiotic therapy. Resistance to currently used antibiotics has steadily increased over the last decade, generating a pressing need for the development of new therapeutic agents. Since iron is essential for colonization and scarce in the urinary tract, targeting iron acquisition would seem to be an attractive strategy. However, the multiplicity and redundancy of iron acquisition systems in uropathogenic Escherichia coli (UPEC) make it difficult to pinpoint a specific cellular target. Here, we identified 16 iron acquisition inhibitors through a whole-cell high-throughput screen, validating iron acquisition as a target for antibacterial strategies against UPEC. We also identified the cellular target of two of the inhibitors as the TonB system.


Subject(s)
Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/pharmacology , Biological Transport/drug effects , Escherichia coli Proteins/antagonists & inhibitors , Iron/metabolism , Membrane Proteins/antagonists & inhibitors , Uropathogenic Escherichia coli/drug effects , Coliphages/physiology , Drug Evaluation, Preclinical , Uropathogenic Escherichia coli/growth & development , Uropathogenic Escherichia coli/metabolism , Virus Attachment
12.
Comb Chem High Throughput Screen ; 17(3): 210-30, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24409957

ABSTRACT

High throughput screening (HTS) is an integral part of a highly collaborative approach to drug discovery at the University of Michigan. The HTS lab is one of four core centers that provide services to identify, produce, screen and follow-up on biomedical targets for faculty. Key features of this system are: protein cloning and purification, protein crystallography, small molecule and siRNA HTS, medicinal chemistry and pharmacokinetics. Therapeutic areas that have been targeted include anti-bacterial, metabolic, neurodegenerative, cardiovascular, anti-cancer and anti-viral. The centers work in a coordinated, interactive environment to affordably provide academic investigators with the technology, informatics and expertise necessary for successful drug discovery. This review provides an overview of these centers at the University of Michigan, along with case examples of successful collaborations with faculty.


Subject(s)
Drug Discovery/methods , High-Throughput Screening Assays/methods , Universities/organization & administration , Biological Products/chemistry , Biological Products/pharmacology , Chemistry, Pharmaceutical , Drug Discovery/organization & administration , Genomics/methods , Humans , Michigan , Molecular Biology/methods , Molecular Biology/organization & administration , Pharmacokinetics , Protein Folding , Proteins/chemistry , Proteins/isolation & purification , Small Molecule Libraries/pharmacology
13.
Nat Med ; 19(3): 313-21, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23396211

ABSTRACT

Emerging evidence suggests that inflammation provides a link between obesity and insulin resistance. The noncanonical IκB kinases IKK-ɛ and TANK-binding kinase 1 (TBK1) are induced in liver and fat by NF-κB activation upon high-fat diet feeding and in turn initiate a program of counterinflammation that preserves energy storage. Here we report that amlexanox, an approved small-molecule therapeutic presently used in the clinic to treat aphthous ulcers and asthma, is an inhibitor of these kinases. Treatment of obese mice with amlexanox elevates energy expenditure through increased thermogenesis, producing weight loss, improved insulin sensitivity and decreased steatosis. Because of its record of safety in patients, amlexanox may be an interesting candidate for clinical evaluation in the treatment of obesity and related disorders.


Subject(s)
Aminopyridines/pharmacology , Anti-Obesity Agents/pharmacology , Energy Metabolism/drug effects , I-kappa B Kinase/antagonists & inhibitors , Insulin Resistance , Obesity/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Anti-Allergic Agents/pharmacology , Cell Line , Diet, High-Fat , Enzyme Activation , Fatty Liver/drug therapy , Glucose Metabolism Disorders/drug therapy , I-kappa B Kinase/metabolism , Insulin Resistance/immunology , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , NF-kappa B/metabolism , Obesity/drug therapy , Obesity/immunology , Oxygen Consumption/drug effects , Protein Serine-Threonine Kinases/metabolism , Weight Loss/drug effects
14.
Article in English | MEDLINE | ID: mdl-24533288

ABSTRACT

Profound neuropeptide diversity characterizes the nematode nervous system, but it has proven challenging to match neuropeptide G protein-coupled receptors (GPCR) with their cognate ligands in heterologous systems. We have expressed the Caenorhabditis elegans GPCR encoded in the locus T19F4.1, previously matched with FMRFamide-like peptides encoded on the flp-2 precursor gene, in mammalian cells and in the yeast Saccharomyces cerevisiae. Pharmacological characterization revealed that the receptor is potently activated by flp-2 peptides in CHO cells (∼10 nM EC50) and in yeast (∼100 nM EC50), signaling through a Gqα pathway in each system. The yeast GPCR expression system provides a robust assay for screening for agonists of the flp-2 receptor and is the target of an ongoing high-throughput screening exercise.

15.
ACS Chem Neurosci ; 3(7): 546-56, 2012 Jul 18.
Article in English | MEDLINE | ID: mdl-22860224

ABSTRACT

ΔFosB protein accumulates in the striatum in response to chronic administration of drugs of abuse, L-DOPA, or stress, triggering long lasting neural and behavioral changes that underlie aspects of drug addiction, abnormal involuntary movements (dyskinesia), and depression. ΔFosB binds AP-1 DNA consensus sequences found in promoters of many genes and can both repress or activate gene transcription. In the striatum, ΔFosB is thought to dimerize with JunD to form a functional transcription factor, though strikingly JunD does not accumulate in parallel. One explanation is that ΔFosB can recruit different partners, including itself, depending on the neuron type in which it is induced and the chronic stimulus, generating protein complexes with different effects on gene transcription. To develop chemical probes to study ΔFosB, a high-throughput screen was carried out to identify small molecules that modulate ΔFosB function. Two compounds with low micromolar activity, termed C2 and C6, disrupt the binding of ΔFosB to DNA via different mechanisms, and in in vitro assays stimulate ΔFosB-mediated transcription. In cocaine-treated mice, C2 significantly elevates mRNA levels of the AMPA glutamate receptor GluR2 subunit with specificity, a known target gene of ΔFosB that plays a role in drug addiction and endogenous resilience mechanisms. C2 and C6 show different activities against ΔFosB homodimers compared to ΔFosB/JunD heterodimers, suggesting that these compounds can be used as probes to study the contribution of different ΔFosB-containing complexes on the regulation of gene transcription in biological systems and to assess the utility of ΔFosB as a therapeutic target.


Subject(s)
Pharmaceutical Preparations/chemistry , Proto-Oncogene Proteins c-fos/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Cell Line, Tumor , Cell Survival/physiology , Drug Evaluation, Preclinical/methods , Insecta , Mice , Pharmaceutical Preparations/metabolism , Protein Binding/physiology , Proto-Oncogene Proteins c-fos/antagonists & inhibitors , Proto-Oncogene Proteins c-fos/physiology , Transcription Factors/antagonists & inhibitors , Transcription Factors/physiology
16.
J Biomol Screen ; 17(8): 1080-7, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22706349

ABSTRACT

High-throughput screening (HTS) has historically been used by the pharmaceutical industry to rapidly test hundreds of thousands of compounds to identify potential drug candidates. More recently, academic groups have used HTS to identify new chemical probes or small interfering RNA (siRNA) that can serve as experimental tools to examine the biology or physiology of novel proteins, processes, or interactions. HTS presents a significant challenge with the vast and complex nature of data generated. This report describes MScreen, a Web-based, open-source cheminformatics application for chemical library and siRNA plate management, primary HTS and dose-response data handling, structure search, and administrative functions. Each project in MScreen can be secured with passwords or shared in an open-information environment that enables collaborators to easily compare data from many screens, providing a useful means to identify compounds with desired selectivity. Unique features include compound, substance, mixture, and siRNA plate creation and formatting; automated dose-response fitting and quality control (QC); and user, target, and assay method administration. MScreen provides an effective means to facilitate HTS information handling and analysis in the academic setting so that users can efficiently view their screening data and evaluate results for follow-up.


Subject(s)
Databases, Chemical , High-Throughput Screening Assays , Information Storage and Retrieval , RNA, Small Interfering , Small Molecule Libraries/pharmacology , Internet
17.
Proc Natl Acad Sci U S A ; 109(9): 3469-74, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22331877

ABSTRACT

The widespread occurrence of antibiotic resistance among human pathogens is a major public health problem. Conventional antibiotics typically target bacterial killing or growth inhibition, resulting in strong selection for the development of antibiotic resistance. Alternative therapeutic approaches targeting microbial pathogenicity without inhibiting growth might minimize selection for resistant organisms. Compounds inhibiting gene expression of streptokinase (SK), a critical group A streptococcal (GAS) virulence factor, were identified through a high-throughput, growth-based screen on a library of 55,000 small molecules. The lead compound [Center for Chemical Genomics 2979 (CCG-2979)] and an analog (CCG-102487) were confirmed to also inhibit the production of active SK protein. Microarray analysis of GAS grown in the presence of CCG-102487 showed down-regulation of a number of important virulence factors in addition to SK, suggesting disruption of a general virulence gene regulatory network. CCG-2979 and CCG-102487 both enhanced granulocyte phagocytosis and killing of GAS in an in vitro assay, and CCG-2979 also protected mice from GAS-induced mortality in vivo. These data suggest that the class of compounds represented by CCG-2979 may be of therapeutic value for the treatment of GAS and potentially other gram-positive infections in humans.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Gene Expression Regulation, Bacterial/drug effects , Quinazolines/therapeutic use , Streptococcal Infections/drug therapy , Streptococcus pyogenes/drug effects , Streptokinase/antagonists & inhibitors , Animals , Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/pharmacology , Depression, Chemical , Drug Evaluation, Preclinical , Enzyme Induction/drug effects , High-Throughput Screening Assays , Host Specificity/genetics , Humans , Kanamycin Resistance/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Structure , Phagocytosis/drug effects , Plasminogen/genetics , Promoter Regions, Genetic/genetics , Quinazolines/isolation & purification , Quinazolines/pharmacology , Small Molecule Libraries , Streptococcus pyogenes/enzymology , Streptococcus pyogenes/genetics , Streptococcus pyogenes/pathogenicity , Streptokinase/biosynthesis , Streptokinase/genetics , Virulence/drug effects , Virulence/genetics
18.
J Biomol Screen ; 16(8): 825-35, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21844328

ABSTRACT

Despite advances toward understanding the prevention and treatment of many cancers, patients who suffer from oral squamous cell carcinoma (OSCC) confront a survival rate that has remained unimproved for more than 2 decades, indicating our ability to treat them pharmacologically has reached a plateau. In an ongoing effort to improve the clinical outlook for this disease, we previously reported that an essential component of the mechanism by which the proteasome inhibitor bortezomib (PS-341, Velcade) induced apoptosis in OSCC required the activation of a terminal unfolded protein response (UPR). Predicated on these studies, the authors hypothesized that high-throughput screening (HTS) of large diverse chemical libraries might identify more potent or selective small-molecule activators of the apoptotic arm of the UPR to control or kill OSCC. They have developed complementary cell-based assays using stably transfected CHO-K1 cell lines that individually assess the PERK/eIF2α/CHOP (apoptotic) or the IRE1/XBP1 (adaptive) UPR subpathways. An 66 K compound collection was screened at the University of Michigan Center for Chemical Genomics that included a unique library of prefractionated natural product extracts. The mycotoxin methoxycitrinin was isolated from a natural extract and found to selectively activate the CHOP-luciferase reporter at 80 µM. A series of citrinin derivatives was isolated from these extracts, including a unique congener that has not been previously described. In an effort to identify more potent compounds, the authors examined the ability of citrinin and the structurally related mycotoxins ochratoxin A and patulin to activate the UPR. Strikingly, it was found that patulin at 2.5 to 10 µM induced a terminal UPR in a panel of OSCC cells that was characterized by an increase in CHOP, GADD34, and ATF3 gene expression and XBP1 splicing. A luminescent caspase assay and the induction of several BH3-only genes indicated that patulin could induce apoptosis in OSCC cells. These data support the use of this complementary HTS strategy to identify novel modulators of UPR signaling and tumor cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays , Mouth Neoplasms/drug therapy , Mycotoxins/pharmacology , Animals , Apoptosis , Boronic Acids/pharmacology , Bortezomib , CHO Cells , Carcinoma, Squamous Cell/pathology , Caspases/genetics , Caspases/metabolism , Cell Proliferation/drug effects , Cricetinae , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Genes, Reporter , Humans , Luciferases/analysis , Mouth Neoplasms/pathology , Pyrazines/pharmacology , Signal Transduction , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Transduction, Genetic , Unfolded Protein Response/drug effects , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
19.
Anal Biochem ; 410(1): 133-40, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21073853

ABSTRACT

Homocitrate synthase (HCS) catalyzes the first step of l-lysine biosynthesis in fungi by condensing acetyl-coenzyme A and 2-oxoglutarate to form 3R-homocitrate and coenzyme A. Due to its conservation in pathogenic fungi, HCS has been proposed as a candidate for antifungal drug design. Here we report the development and validation of a robust fluorescent assay for HCS that is amenable to high-throughput screening for inhibitors in vitro. Using this assay, Schizosaccharomyces pombe HCS was screened against a diverse library of approximately 41,000 small molecules. Following confirmation, counter screens, and dose-response analysis, we prioritized more than 100 compounds for further in vitro and in vivo analysis. This assay can be readily adapted to screen for small molecule modulators of other acyl-CoA-dependent acyltransferases or enzymes that generate a product with a free sulfhydryl group, including histone acetyltransferases, aminoglycoside N-acetyltransferases, thioesterases, and enzymes involved in lipid metabolism.


Subject(s)
Drug Evaluation, Preclinical/methods , Enzyme Assays/methods , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Histone Acetyltransferases/metabolism , Oxo-Acid-Lyases/antagonists & inhibitors , Spectrometry, Fluorescence/methods , Acyl Coenzyme A/metabolism , Chelating Agents/chemistry , Chelating Agents/pharmacology , Enzyme Inhibitors/chemistry , Metals/chemistry , Naphthalenes/chemistry , Oxo-Acid-Lyases/chemistry , Oxo-Acid-Lyases/metabolism , Pyrroles/chemistry , Reproducibility of Results , Schizosaccharomyces/enzymology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Sulfhydryl Compounds/chemistry
20.
J Biomol Screen ; 15(10): 1211-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20926844

ABSTRACT

Members of the heat shock protein 70 (Hsp70) family of molecular chaperones are emerging as potential therapeutic targets. Their ATPase activity has classically been measured using colorimetric phosphate detection reagents, such as quinaldine red (QR). Although such assays are suitable for 96-well plate formats, they typically lose sensitivity when attempted in lower volume due to path length and meniscus effects. These limitations and Hsp70's weak enzymatic activity have combined to create significant challenges in high-throughput screening. To overcome these difficulties, the authors have adopted an energy transfer strategy that was originally reported by Zuck et al. (Anal Biochem 2005;342:254-259). Briefly, white 384-well plates emit fluorescence when irradiated at 430 nm. In turn, this intrinsic fluorescence can be quenched by energy transfer with the QR-based chromophore. Using this more sensitive approach, the authors tested 55,400 compounds against DnaK, a prokaryotic member of the Hsp70 family. The assay performance was good (Z' ~0.6, coefficient of variation ~8%), and at least one promising new inhibitor was identified. In secondary assays, this compound specifically blocked stimulation of DnaK by its co-chaperone, DnaJ. Thus, this simple and inexpensive adaptation of a colorimetric method might be suitable for screening against Hsp70 family members.


Subject(s)
Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/metabolism , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/metabolism , High-Throughput Screening Assays/methods , Adenosine Triphosphatases/metabolism , Drug Discovery , Fluorescence Resonance Energy Transfer , HSP40 Heat-Shock Proteins/antagonists & inhibitors , HSP40 Heat-Shock Proteins/metabolism , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/metabolism , Small Molecule Libraries
SELECTION OF CITATIONS
SEARCH DETAIL
...