Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Chem Inf Model ; 63(17): 5513-5528, 2023 09 11.
Article in English | MEDLINE | ID: mdl-37625010

ABSTRACT

Traditional small-molecule drug discovery is a time-consuming and costly endeavor. High-throughput chemical screening can only assess a tiny fraction of drug-like chemical space. The strong predictive power of modern machine-learning methods for virtual chemical screening enables training models on known active and inactive compounds and extrapolating to much larger chemical libraries. However, there has been limited experimental validation of these methods in practical applications on large commercially available or synthesize-on-demand chemical libraries. Through a prospective evaluation with the bacterial protein-protein interaction PriA-SSB, we demonstrate that ligand-based virtual screening can identify many active compounds in large commercial libraries. We use cross-validation to compare different types of supervised learning models and select a random forest (RF) classifier as the best model for this target. When predicting the activity of more than 8 million compounds from Aldrich Market Select, the RF substantially outperforms a naïve baseline based on chemical structure similarity. 48% of the RF's 701 selected compounds are active. The RF model easily scales to score one billion compounds from the synthesize-on-demand Enamine REAL database. We tested 68 chemically diverse top predictions from Enamine REAL and observed 31 hits (46%), including one with an IC50 value of 1.3 µM.


Subject(s)
High-Throughput Screening Assays , Small Molecule Libraries , Databases, Factual , Drug Discovery , Supervised Machine Learning
2.
PLoS Comput Biol ; 15(8): e1006813, 2019 08.
Article in English | MEDLINE | ID: mdl-31381559

ABSTRACT

Prediction of compounds that are active against a desired biological target is a common step in drug discovery efforts. Virtual screening methods seek some active-enriched fraction of a library for experimental testing. Where data are too scarce to train supervised learning models for compound prioritization, initial screening must provide the necessary data. Commonly, such an initial library is selected on the basis of chemical diversity by some pseudo-random process (for example, the first few plates of a larger library) or by selecting an entire smaller library. These approaches may not produce a sufficient number or diversity of actives. An alternative approach is to select an informer set of screening compounds on the basis of chemogenomic information from previous testing of compounds against a large number of targets. We compare different ways of using chemogenomic data to choose a small informer set of compounds based on previously measured bioactivity data. We develop this Informer-Based-Ranking (IBR) approach using the Published Kinase Inhibitor Sets (PKIS) as the chemogenomic data to select the informer sets. We test the informer compounds on a target that is not part of the chemogenomic data, then predict the activity of the remaining compounds based on the experimental informer data and the chemogenomic data. Through new chemical screening experiments, we demonstrate the utility of IBR strategies in a prospective test on three kinase targets not included in the PKIS.


Subject(s)
Drug Discovery/methods , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Cheminformatics/methods , Cheminformatics/statistics & numerical data , Computational Biology , Computer Simulation , Databases, Chemical , Databases, Pharmaceutical , Drug Discovery/statistics & numerical data , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/statistics & numerical data , High-Throughput Screening Assays/methods , High-Throughput Screening Assays/statistics & numerical data , Humans , Prospective Studies , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protozoan Proteins , Structure-Activity Relationship , User-Computer Interface , Viral Proteins/antagonists & inhibitors
3.
Nat Commun ; 10(1): 516, 2019 01 31.
Article in English | MEDLINE | ID: mdl-30705269

ABSTRACT

Antimicrobial resistance is a global health crisis and few novel antimicrobials have been discovered in recent decades. Natural products, particularly from Streptomyces, are the source of most antimicrobials, yet discovery campaigns focusing on Streptomyces from the soil largely rediscover known compounds. Investigation of understudied and symbiotic sources has seen some success, yet no studies have systematically explored microbiomes for antimicrobials. Here we assess the distinct evolutionary lineages of Streptomyces from insect microbiomes as a source of new antimicrobials through large-scale isolations, bioactivity assays, genomics, metabolomics, and in vivo infection models. Insect-associated Streptomyces inhibit antimicrobial-resistant pathogens more than soil Streptomyces. Genomics and metabolomics reveal their diverse biosynthetic capabilities. Further, we describe cyphomycin, a new molecule active against multidrug resistant fungal pathogens. The evolutionary trajectories of Streptomyces from the insect microbiome influence their biosynthetic potential and ability to inhibit resistant pathogens, supporting the promise of this source in augmenting future antimicrobial discovery.


Subject(s)
Biological Products/pharmacology , Insecta/microbiology , Microbiota , Streptomyces/physiology , Animals , Anti-Bacterial Agents/metabolism , Anti-Infective Agents/pharmacology , Genomics , Metabolomics , Microbial Sensitivity Tests
4.
J Chem Inf Model ; 59(1): 282-293, 2019 01 28.
Article in English | MEDLINE | ID: mdl-30500183

ABSTRACT

Virtual (computational) high-throughput screening provides a strategy for prioritizing compounds for experimental screens, but the choice of virtual screening algorithm depends on the data set and evaluation strategy. We consider a wide range of ligand-based machine learning and docking-based approaches for virtual screening on two protein-protein interactions, PriA-SSB and RMI-FANCM, and present a strategy for choosing which algorithm is best for prospective compound prioritization. Our workflow identifies a random forest as the best algorithm for these targets over more sophisticated neural network-based models. The top 250 predictions from our selected random forest recover 37 of the 54 active compounds from a library of 22,434 new molecules assayed on PriA-SSB. We show that virtual screening methods that perform well on public data sets and synthetic benchmarks, like multi-task neural networks, may not always translate to prospective screening performance on a specific assay of interest.


Subject(s)
Drug Evaluation, Preclinical/methods , Machine Learning , Molecular Docking Simulation , Algorithms , Protein Conformation , Proteins/chemistry , Proteins/metabolism , User-Computer Interface
5.
Org Lett ; 20(18): 5529-5532, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30160121

ABSTRACT

Screening of a marine natural products library for inhibitors of TGF-ß revealed five pyrimidinedione derivatives, biemamides A-E (1-5). The structures were determined by 2D NMR and HRMS experiments; absolute configurations were established by advanced Marfey's analysis and ECD calculations. Biemamides A-E specifically inhibited in vitro TGF-ß induced epithelial to mesenchymal transition in NMuMG cells. Additionally, using Caenorhabditis elegans, selected biemmamides were found to influence in vivo developmental processes related to body size regulation in a dose-dependent manner.


Subject(s)
Biological Products/pharmacology , Caenorhabditis elegans/drug effects , Epithelial-Mesenchymal Transition/drug effects , Pyrimidinones/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Biological Products/chemistry , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Cell Line , Dose-Response Relationship, Drug , Humans , Pyrimidinones/chemistry , Structure-Activity Relationship , Transforming Growth Factor beta/metabolism
6.
SLAS Discov ; 23(1): 94-101, 2018 01.
Article in English | MEDLINE | ID: mdl-28570838

ABSTRACT

Antibiotic-resistant bacterial infections are increasingly prevalent worldwide, and there is an urgent need for novel classes of antibiotics capable of overcoming existing resistance mechanisms. One potential antibiotic target is the bacterial single-stranded DNA binding protein (SSB), which serves as a hub for DNA repair, recombination, and replication. Eight highly conserved residues at the C-terminus of SSB use direct protein-protein interactions (PPIs) to recruit more than a dozen important genome maintenance proteins to single-stranded DNA. Mutations that disrupt PPIs with the C-terminal tail of SSB are lethal, suggesting that small-molecule inhibitors of these critical SSB PPIs could be effective antibacterial agents. As a first step toward implementing this strategy, we have developed orthogonal high-throughput screening assays to identify small-molecule inhibitors of the Klebsiella pneumonia SSB-PriA interaction. Hits were identified from an initial screen of 72,474 compounds using an AlphaScreen (AS) primary screen, and their activity was subsequently confirmed in an orthogonal fluorescence polarization (FP) assay. As an additional control, an FP assay targeted against an unrelated eukaryotic PPI was used to confirm specificity for the SSB-PriA interaction. Nine potent and selective inhibitors produced concentration-response curves with IC50 values of <40 µM, and two compounds were observed to directly bind to PriA, demonstrating the success of this screen strategy.


Subject(s)
DNA-Binding Proteins/metabolism , Drug Discovery/methods , High-Throughput Screening Assays , Protein Interaction Mapping/methods , DNA-Binding Proteins/chemistry , Molecular Structure , Protein Binding/drug effects , Small Molecule Libraries , Thermodynamics , Workflow
7.
J Nat Prod ; 80(9): 2551-2555, 2017 09 22.
Article in English | MEDLINE | ID: mdl-28840714

ABSTRACT

Here we describe the rapid identification and prioritization of novel active marine natural products using an improved dereplication strategy. During the course of our screening of marine natural product libraries, a new cyclic trihydroxamate compound, thalassosamide, was discovered from the α-proteobacterium Thalassospira profundimaris. Its structure was determined by 2D NMR and MS/MS experiments, and the absolute configuration of the lysine-derived units was established by Marfey's analysis, whereas that of C-9, 9', and 9″ was determined via the circular dichroism data of the [Rh2(OCOCF3)4] complex and DFT NMR calculations. Thalassosamide showed moderate in vivo efficacy against Pseudomonas aeruginosa.


Subject(s)
Biological Products/isolation & purification , Biological Products/pharmacology , Hydroxamic Acids/isolation & purification , Pseudomonas aeruginosa/chemistry , Siderophores/chemistry , Siderophores/isolation & purification , Siderophores/pharmacology , Biological Products/chemistry , Circular Dichroism , Hydroxamic Acids/chemistry , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Tandem Mass Spectrometry
8.
J Chem Inf Model ; 57(7): 1579-1590, 2017 07 24.
Article in English | MEDLINE | ID: mdl-28654262

ABSTRACT

In structure-based virtual screening, compound ranking through a consensus of scores from a variety of docking programs or scoring functions, rather than ranking by scores from a single program, provides better predictive performance and reduces target performance variability. Here we compare traditional consensus scoring methods with a novel, unsupervised gradient boosting approach. We also observed increased score variation among active ligands and developed a statistical mixture model consensus score based on combining score means and variances. To evaluate performance, we used the common performance metrics ROCAUC and EF1 on 21 benchmark targets from DUD-E. Traditional consensus methods, such as taking the mean of quantile normalized docking scores, outperformed individual docking methods and are more robust to target variation. The mixture model and gradient boosting provided further improvements over the traditional consensus methods. These methods are readily applicable to new targets in academic research and overcome the potentially poor performance of using a single docking method on a new target.


Subject(s)
Drug Evaluation, Preclinical/methods , Machine Learning , Molecular Targeted Therapy , Proteins/metabolism , Benchmarking , Molecular Docking Simulation , User-Computer Interface
9.
Transplantation ; 100(6): 1198-210, 2016 06.
Article in English | MEDLINE | ID: mdl-26950727

ABSTRACT

BACKGROUND: We hypothesized that nicotinamide adenosine diphosphate oxidase 2 (Nox2) plays an important role in cyclosporine A (CsA)-induced chronic hypoxia. METHODS: We tested this hypothesis in Fisher 344 rats, C57BL/6 J wild type and Nox2-/- mice, and in liver transplant recipients with chronic CsA nephrotoxicity. We used noninvasive molecular imaging (blood oxygen level-dependent magnetic resonance imaging and dynamic contrast-enhanced magnetic resonance imaging) and molecular diagnostic tools to assess intrarenal oxygenation and perfusion, and the molecular phenotype of CsA nephrotoxicity. RESULTS: We observed that chemical and genetic inhibition of Nox2 in rats and mice resulted in the prevention of CsA-induced hypoxia independent of regional perfusion (blood oxygen level-dependent magnetic resonance imaging and dynamic contrast-enhanced magnetic resonance imaging, pimonidazole, HIF-1α). Nicotinamide adenosine diphosphate oxidase 2 knockout was also associated with decreased oxidative stress (Nox2, HIF-1α, hydrogen peroxide, hydroxynonenal), and fibrogenesis (α-smooth muscle actin, picrosirius red, trichrome, vimentin). The molecular signature of chronic CsA nephrotoxicity using transcriptomic analyses demonstrated significant changes in 40 genes involved in injury repair, metabolism, and oxidative stress in Nox2-/- mice. Immunohistochemical analyses of kidney biopsies from liver transplant recipients with chronic CsA nephrotoxicity showed significantly greater Nox2, α-smooth muscle actin and picrosirius levels compared with controls. CONCLUSIONS: These studies suggest that Nox2 is a modulator of CsA-induced hypoxia upstream of HIF-1α and define the molecular characteristics that could be used for the diagnosis and monitoring of chronic calcineurin inhibitor nephrotoxicity.


Subject(s)
Cyclosporine/adverse effects , Hypoxia/chemically induced , Kidney/drug effects , Kidney/pathology , Liver Transplantation , Membrane Glycoproteins/genetics , NADPH Oxidases/genetics , Actins/metabolism , Animals , Azo Compounds/metabolism , Biopsy , Calcineurin Inhibitors/chemistry , Contrast Media/chemistry , Hydrogen Peroxide/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver/pathology , Magnetic Resonance Imaging , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Perfusion , Phenotype , Rats , Rats, Inbred F344 , Vimentin/metabolism
10.
Chem Biol ; 22(12): 1608-21, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26670079

ABSTRACT

ERß is regarded as a "tumor suppressor" in breast cancer due to its anti-proliferative effects. However, unlike ERα, ERß has not been developed as a therapeutic target in breast cancer due to loss of ERß in aggressive cancers. In a small-molecule library screen for ERß stabilizers, we identified Diptoindonesin G (Dip G), which significantly increases ERß protein stability while decreasing ERα protein levels. Dip G enhances the transcription and anti-proliferative activities of ERß, while attenuating the transcription and proliferative effects of ERα. Further investigation revealed that instead of targeting ER, Dip G targets the CHIP E3 ubiquitin ligase shared by ERα and ERß. Thus, Dip G is a dual-functional moiety that reciprocally controls ERα and ERß protein stability and activities via an indirect mechanism. The ERß stabilization effects of Dip G may enable the development of ERß-targeted therapies for human breast cancers.


Subject(s)
Benzofurans/pharmacology , Estrogen Receptor alpha/drug effects , Estrogen Receptor beta/drug effects , Benzofurans/chemistry , Blotting, Western , Breast Neoplasms/therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Molecular Docking Simulation , Protein Stability/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
11.
PLoS One ; 9(2): e89349, 2014.
Article in English | MEDLINE | ID: mdl-24586708

ABSTRACT

Intimal hyperplasia is the cause of the recurrent occlusive vascular disease (restenosis). Drugs currently used to treat restenosis effectively inhibit smooth muscle cell (SMC) proliferation, but also inhibit the growth of the protective luminal endothelial cell (EC) lining, leading to thrombosis. To identify compounds that selectively inhibit SMC versus EC proliferation, we have developed a high-throughput screening (HTS) format using human cells and have employed this to screen a multiple compound collection (NIH Clinical Collection). We developed an automated, accurate proliferation assay in 96-well plates using human aortic SMCs and ECs. Using this HTS format we screened a 447-drug NIH Clinical Library. We identified 11 compounds that inhibited SMC proliferation greater than 50%, among which idarubicin exhibited a unique feature of preferentially inhibiting SMC versus EC proliferation. Concentration-response analysis revealed this differential effect most evident over an ∼10 nM-5 µM window. In vivo testing of idarubicin in a rat carotid injury model at 14 days revealed an 80% reduction of intimal hyperplasia and a 45% increase of lumen size with no significant effect on re-endothelialization. Taken together, we have established a HTS assay of human vascular cell proliferation, and identified idarubicin as a selective inhibitor of SMC versus EC proliferation both in vitro and in vivo. Screening of larger and more diverse compound libraries may lead to the discovery of next-generation therapeutics that can inhibit intima hyperplasia without impairing re-endothelialization.


Subject(s)
Cell Proliferation/drug effects , Idarubicin/pharmacology , Muscle, Smooth, Vascular/drug effects , Tunica Intima/drug effects , Animals , Cells, Cultured , Endothelial Cells/drug effects , High-Throughput Screening Assays/methods , Humans , Hyperplasia/drug therapy , Male , Myocytes, Smooth Muscle/drug effects , Rats , Rats, Sprague-Dawley
12.
Mol Endocrinol ; 27(11): 1946-57, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24002653

ABSTRACT

Myostatin, a member of the TGF superfamily, is sufficient to induce skeletal muscle atrophy. Myostatin-induced atrophy is associated with increases in E3-ligase atrogin-1 expression and protein degradation and decreases in Akt/mechanistic target of rapamycin (mTOR) signaling and protein synthesis. Myostatin signaling activates the transcription factor Smad3 (Small Mothers Against Decapentaplegic), which has been shown to be necessary for myostatin-induced atrogin-1 expression and atrophy; however, it is not known whether Smad3 is sufficient to induce these events or whether Smad3 simply plays a permissive role. Thus, the aim of this study was to address these questions with an in vivo model. To accomplish this goal, in vivo transfection of plasmid DNA was used to create transient transgenic mouse skeletal muscles, and our results show for the first time that Smad3 expression is sufficient to stimulate atrogin-1 promoter activity, inhibit Akt/mTOR signaling and protein synthesis, and induce muscle fiber atrophy. Moreover, we propose that Akt/mTOR signaling is inhibited by a Smad3-induced decrease in microRNA-29 (miR-29) expression and a subsequent increase in the translation of phosphatase and tensin homolog (PTEN) mRNA. Smad3 is also sufficient to inhibit peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α) promoter activity and to increase FoxO (Forkhead Box Protein, Subclass O)-mediated signaling and the promoter activity of plasminogen activator inhibitor 1 (PAI-1). Combined, this study provides the first evidence that Smad3 is sufficient to regulate many of the events associated with myostatin-induced atrophy and therefore suggests that Smad3 signaling may be a viable target for therapies aimed at preventing myostatin-induced muscle atrophy.


Subject(s)
Muscle Proteins/genetics , Muscular Atrophy/metabolism , SKP Cullin F-Box Protein Ligases/genetics , Smad3 Protein/physiology , TOR Serine-Threonine Kinases/metabolism , Transcriptional Activation , Animals , Base Sequence , Female , Gene Expression , Mice , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
13.
Chembiochem ; 14(7): 827-35, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23585185

ABSTRACT

Epigenetics is an emerging field that demands selective cell-permeable chemical probes to perturb, especially in vivo, the activity of specific enzymes involved in modulating the epigenetic codes. Coactivator-associated arginine methyltransferase 1 (CARM1) is a coactivator of estrogen receptor α (ERα), the main target in human breast cancer. We previously showed that twofold overexpression of CARM1 in MCF7 breast cancer cells increased the expression of ERα-target genes involved in differentiation and reduced cell proliferation, thus leading to the hypothesis that activating CARM1 by chemical activators might be therapeutically effective in breast cancer. Selective, potent, cell-permeable CARM1 activators will be essential to test this hypothesis. Here we report the development of a cell-based, time-resolved (TR) FRET assay that uses poly(A) binding protein 1 (PABP1) methylation to monitor cellular activity of CARM1. The LanthaScreen TR-FRET assay uses MCF7 cells expressing GFP-PABP1 fusion protein through BacMam gene delivery system, methyl-PABP1 specific antibody, and terbium-labeled secondary antibody. This assay has been validated as reflecting the expression and/or activity of CARM1 and optimized for high throughput screening to identify CARM1 allosteric activators. This TR-FRET platform serves as a generic tool for functional screening of cell-permeable, chemical modulators of CARM1 for elucidation of its in vivo functions.


Subject(s)
Breast Neoplasms/enzymology , Fluorescence Resonance Energy Transfer , Protein-Arginine N-Methyltransferases/metabolism , Enzyme Activation/drug effects , Female , Humans , MCF-7 Cells , Time Factors
14.
Sci Rep ; 3: 1106, 2013.
Article in English | MEDLINE | ID: mdl-23346363

ABSTRACT

Although two classes of antivirals, NA inhibitors and M2 ion channel blockers, are licensed for influenza treatment, dual resistant mutants, including highly pathogenic H5N1 viruses, have appeared. Alternative treatment options are, therefore, needed. Influenza A viral RNA (vRNA) transcription/replication is a promising target for antiviral development, since it is essential for virus replication. Accordingly, an efficient and reliable method to identify vRNA transcription/replication inhibitors is desirable. Here, we developed a cell-based screening system by establishing a cell line that stably expresses influenza viral ribonucleoprotein complex (vRNP). Compound library screening using this cell line allowed us to identify a compound that inhibits vRNA transcription/replication by using reporter protein expression from virus-like RNA as a readout and virus replication in vitro. vRNP-expressing cells have potential as a simple and convenient high-throughput screening (HTS) system, and, thus, are promising to identify vRNA transcription/replication inhibitors for various RNA viruses, especially for primary screens.


Subject(s)
Antiviral Agents/pharmacology , High-Throughput Screening Assays/methods , Influenza A virus/drug effects , Influenza A virus/physiology , RNA, Viral/drug effects , Virus Replication/drug effects , Animals , Dogs , Drug Evaluation, Preclinical/methods , Genetic Vectors/genetics , HEK293 Cells , Humans , Influenza A virus/genetics , Influenza, Human/drug therapy , Influenza, Human/virology , Madin Darby Canine Kidney Cells , RNA Viruses/drug effects , RNA Viruses/genetics , RNA, Viral/genetics , Vault Ribonucleoprotein Particles/drug effects , Vault Ribonucleoprotein Particles/genetics , Viral Proteins/genetics , Virus Replication/genetics
15.
Fibrogenesis Tissue Repair ; 5(Suppl 1): S28, 2012.
Article in English | MEDLINE | ID: mdl-23259633

ABSTRACT

BACKGROUND: The dynamic process of epithelial-to-mesenchymal transition (EMT) is a causal event in kidney fibrosis. This cellular phenotypic transition involves activation of transcriptional responses and remodeling of cellular structures to change cellular function. The molecular mechanisms that directly contribute to the re-establishment of the epithelial phenotype are poorly understood. RESULTS: Here, we discuss recent studies from our group and other laboratories identifying signaling pathways leading to the reversal of EMT in fibrotic models. We also present evidence that transcriptional factors such as the ZEB proteins are important regulators for reversal of EMT. CONCLUSION: These studies provide insights into cellular plasticity and possible targets for therapeutic intervention.

16.
Matrix Biol ; 31(6): 360-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22986508

ABSTRACT

Fibronectin (FN) is a plasma glycoprotein that circulates in the near micromolar concentration range and is deposited along with locally produced FN in the extracellular matrices of many tissues. The control of FN deposition is tightly controlled by cells. Agents that modulate FN assembly may be useful therapeutically in conditions characterized by excessive FN deposition, such as fibrosis, inflammatory diseases, and malignancies. To identify such agents by high throughput screening (HTS), we developed a microtiter assay of FN deposition by human fibroblasts. The assay provides a robust read-out of FN assembly. Alexa 488-FN (A488-FN) was added to cell monolayers, and the total fluorescence intensity of deposited A488-FN was quantified. The fluorescence intensity of deposited A488-FN correlated with the presence of FN fibrils visualized by fluorescence microscopy. The assay Z' values were 0.67 or 0.54, respectively, when using background values of fluorescence either with no added A488-FN or with A488-FN added together with a known inhibitor of FN deposition. The assay was used to screen libraries comprising 4160 known bioactive compounds. Nine compounds were identified as non- or low-cytotoxic inhibitors of FN assembly. Four (ML-9, HA-100, tyrphostin and imatinib mesylate) are kinase inhibitors, a category of compounds known to inhibit FN assembly; two (piperlongumine and cantharidin) are promoters of cancer cell apoptosis; and three (maprotiline, CGS12066B, and aposcopolamine) are modulators of biogenic amine signaling. The latter six compounds have not been recognized heretofore as affecting FN assembly. The assay is straight-forward, adapts to 96- and 384-well formats, and should be useful for routine measurement of FN deposition and HTS. Screening of more diverse chemical libraries and identification of specific and efficient modulators of FN fibrillogenesis may result in therapeutics to control excessive connective tissue deposition.


Subject(s)
Fibronectins/metabolism , High-Throughput Screening Assays/methods , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Fluorescent Dyes/metabolism , Humans , Protein Kinase Inhibitors/pharmacology , Protein Multimerization , Signal Transduction , Small Molecule Libraries , Titrimetry
17.
PLoS One ; 6(9): e25021, 2011.
Article in English | MEDLINE | ID: mdl-21949838

ABSTRACT

BACKGROUND: Hub proteins are connected through binding interactions to many other proteins. Smad3, a mediator of signal transduction induced by transforming growth factor beta (TGF-ß), serves as a hub protein for over 50 protein-protein interactions. Different cellular responses mediated by Smad3 are the product of cell-type and context dependent Smad3-nucleated protein complexes acting in concert. Our hypothesis is that perturbation of this spectrum of protein complexes by mutation of single protein-binding hot-spots on Smad3 will have distinct consequences on Smad3-mediated responses. METHODOLOGY/PRINCIPAL FINDINGS: We mutated 28 amino acids on the surface of the Smad3 MH2 domain and identified 22 Smad3 variants with reduced binding to subsets of 17 Smad3-binding proteins including Smad4, SARA, Ski, Smurf2 and SIP1. Mutations defective in binding to Smad4, e.g., D408H, or defective in nucleocytoplasmic shuttling, e.g., W406A, were compromised in modulating the expression levels of a Smad3-dependent reporter gene or six endogenous Smad3-responsive genes: Mmp9, IL11, Tnfaip6, Fermt1, Olfm2 and Wnt11. However, the Smad3 mutants Y226A, Y297A, W326A, K341A, and E267A had distinct differences on TGF-ß signaling. For example, K341A and Y226A both reduced the Smad3-mediated activation of the reporter gene by ∼50% but K341A only reduced the TGF-ß inducibilty of Olfm2 in contrast to Y226A which reduced the TGF-ß inducibility of all six endogenous genes as severely as the W406A mutation. E267A had increased protein binding but reduced TGF-ß inducibility because it caused higher basal levels of expression. Y297A had increased TGF-ß inducibility because it caused lower Smad3-induced basal levels of gene expression. CONCLUSIONS/SIGNIFICANCE: Mutations in protein binding hot-spots on Smad3 reduced the binding to different subsets of interacting proteins and caused a range of quantitative changes in the expression of genes induced by Smad3. This approach should be useful for unraveling which Smad3 protein complexes are critical for specific biological responses.


Subject(s)
Gene Expression Regulation , Mutation/genetics , Protein Interaction Domains and Motifs , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Biomarkers/metabolism , Blotting, Western , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Proliferation , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , GTP-Binding Proteins , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Kidney/cytology , Kidney/metabolism , Luciferases/metabolism , Mice , Models, Molecular , Myoblasts/cytology , Myoblasts/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Protein Conformation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/chemistry , Smad4 Protein/genetics , Smad4 Protein/metabolism , Trans-Activators , Transforming Growth Factor beta/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
18.
PLoS One ; 5(11): e15511, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21152098

ABSTRACT

BACKGROUND: Transforming growth factor beta 1 (TGF-ß1) is an inhibitor of muscle cell differentiation that is associated with fibrosis, poor regeneration and poor function in some diseases of muscle. When neutralizing antibodies to TGF-ß1 or the angiotensin II inhibitor losartan were used to reduce TGF-ß1 signaling, muscle morphology and function were restored in mouse models of Marfan Syndrome and muscular dystrophy. The goal of our studies was to identify additional agents that overcome the anti-myogenic effect of TGF-ß1. METHODOLOGY/PRINCIPAL FINDINGS: A high-content cell-based assay was developed in a 96-well plate format that detects the expression of myosin heavy chain (MHC) in C2C12 cells. The assay was used to quantify the dose-dependent responses of C2C12 cell differentiation to TGF-ß1 and to the TGF-ß1 Type 1 receptor kinase inhibitor, SB431542. Thirteen agents previously described as promoting C2C12 differentiation in the absence of TGF-ß1 were screened in the presence of TGF-ß1. Only all-trans retinoic acid and 9-cis retinoic acid allowed a maximal level of C2C12 cell differentiation in the presence of TGF-ß1; the angiotensin-converting enzyme inhibitor captopril and 10 nM estrogen provided partial rescue. Vitamin D was a potent inhibitor of retinoic acid-induced myogenesis in the presence of TGF-ß1. TGF-ß1 inhibits myoblast differentiation through activation of Smad3; however, retinoic acid did not inhibit TGF-ß1-induced activation of a Smad3-dependent reporter gene in C2C12 cells. CONCLUSIONS/SIGNIFICANCE: Retinoic acid alleviated the anti-myogenic effect of TGF-ß1 by a Smad3-independent mechanism. With regard to the goal of improving muscle regeneration and function in individuals with muscle disease, the identification of retinoic acid is intriguing in that some retinoids are already approved for human therapy. However, retinoids also have well-described adverse effects. The quantitative, high-content assay will be useful to screen for less-toxic retinoids or combinations of agents that promote myoblast differentiation in the presence of TGF-ß1.


Subject(s)
Muscle Development/drug effects , Myoblasts/drug effects , Transforming Growth Factor beta1/pharmacology , Tretinoin/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Antioxidants/pharmacology , Benzamides/pharmacology , Captopril/pharmacology , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Dioxoles/pharmacology , Dose-Response Relationship, Drug , Mice , Myoblasts/cytology , Myoblasts/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Resveratrol , Signal Transduction/drug effects , Smad3 Protein/metabolism , Stilbenes/pharmacology , Vitamin D/pharmacology , Vitamins/pharmacology
19.
Biochem Pharmacol ; 80(8): 1221-9, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20599778

ABSTRACT

Xenoestrogenic compounds are abundant in the modern environment including phytoestrogens from plants, chemical by-products from industry, and secondary metabolites from microbes; all can profoundly affect human health. Consequently mechanism-based screens are urgently needed to improve the rate at which the xenoestrogens are discovered. Estrogen Receptor (ER) dimerization is required for target gene transcription. The three ER dimer pairs (ERalpha/alpha homodimers, ERbeta/beta homodimers, and ERalpha/beta heterodimers) exhibit diverse physiological responses in response to ligand-dependent activation with ERalpha/alpha homodimers being pro-proliferative and ERbeta/beta homodimers being anti-proliferative. The biological role of the ERalpha/beta heterodimer remains unclear. We previously developed a cell-based, bioluminescence resonance energy transfer (BRET) assay that can distinguish natural estrogenic compounds based on their abilities to activate the three diverse ER dimer pairs. Using BRET assays, we sought to identify novel xenoestrogens from soil bacteria that preferentially activate ERalpha/beta heterodimer with hopes of shedding light on the biological function of this elusive dimer pair. Here we describe the application of BRET assays in high throughput screens of crude bacterial extracts not previously screened for ER modulatory function and originating from unique ecological niches. Here we report the discovery and biological evaluation of a new natural product, actinopolymorphol A (1), that preferentially induces ERalpha/beta dimerization. Actinopolymorphol A represents the first representative of a new ER modulatory scaffold.


Subject(s)
Acetates/chemistry , Acetates/pharmacology , Bacteria/metabolism , Estrogens/chemistry , Estrogens/pharmacology , Phenols/chemistry , Phenols/pharmacology , Receptors, Estrogen/agonists , Animals , Biological Assay , Breast Neoplasms , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Humans , Models, Molecular , Molecular Structure , Protein Binding , Protein Conformation , Rats , Receptors, Estrogen/chemistry , Soil Microbiology
20.
Cancer Res ; 70(11): 4560-8, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20460526

ABSTRACT

Relatively high oxidative stress levels in the prostate are postulated to be a major factor for prostate carcinogenesis and prostate cancer (CaP) progression. We focused on elucidating metabolic pathways of oxidative stress generation in CaP cells. Previously, we showed that the transcription factor JunD is essential for androgen-induced reactive oxygen species (ROS) production in androgen-dependent human CaP cells. We also recently showed that androgen induces the first and regulatory enzyme spermidine/spermine N1-acetyltransferase (SSAT) in a polyamine catabolic pathway that produces copious amounts of metabolic ROS. Here, we present coimmunoprecipitation and Gaussia luciferase reconstitution assay data that show that JunD forms a complex with androgen-activated androgen receptor (AR) in situ. Our chromatin immunoprecipitation assay data show that JunD binds directly to a specific SSAT promoter sequence only in androgen-treated LNCaP cells. Using a vector containing a luciferase reporter gene connected to the SSAT promoter and a JunD-silenced LNCaP cell line, we show that JunD is essential for androgen-induced SSAT gene expression. The elucidation of JunD-AR complex inducing SSAT expression leading to polyamine oxidation establishes the mechanistic basis of androgen-induced ROS production in CaP cells and opens up a new prostate-specific target for CaP chemopreventive/chemotherapeutic drug development.


Subject(s)
Oxidative Stress/physiology , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Androgen/metabolism , Acetyltransferases/biosynthesis , Acetyltransferases/genetics , Acetyltransferases/metabolism , Androgens/metabolism , Androgens/pharmacology , Base Sequence , Cell Line, Tumor , Cell Nucleus/metabolism , Gene Expression , Humans , Immunoprecipitation , Male , Molecular Sequence Data , Promoter Regions, Genetic , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...