Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Article in English | MEDLINE | ID: mdl-31427291

ABSTRACT

The suboptimal effectiveness of ß-lactam antibiotics against Mycobacterium tuberculosis has hindered the utility of this compound class for tuberculosis treatment. However, the results of treatment with a second-line regimen containing meropenem plus a ß-lactamase inhibitor were found to be encouraging in a case study of extensively drug-resistant tuberculosis (M. C. Payen, S. De Wit, C. Martin, R. Sergysels, et al., Int J Tuberc Lung Dis 16:558-560, 2012, https://doi.org/10.5588/ijtld.11.0414). We hypothesized that the innate resistance of M. tuberculosis to ß-lactams is mediated in part by noncanonical accessory proteins that are not considered the classic targets of ß-lactams and that small-molecule inhibitors of those accessory targets might sensitize M. tuberculosis to ß-lactams. In this study, we screened an NIH small-molecule library for the ability to sensitize M. tuberculosis to meropenem. We identified six hit compounds, belonging to either the N-arylindole or benzothiophene chemotype. Verification studies confirmed the synthetic lethality phenotype for three of the N-arylindoles and one benzothiophene derivative. The latter was demonstrated to be partially bioavailable via oral administration in mice. Structure-activity relationship studies of both structural classes identified analogs with potent antitubercular activity, alone or in combination with meropenem. Transcriptional profiling revealed that oxidoreductases, MmpL family proteins, and a 27-kDa benzoquinone methyltransferase could be the targets of the N-arylindole potentiator. In conclusion, our compound-compound synthetic lethality screening revealed novel small molecules that were capable of potentiating the action of meropenem, presumably via inhibition of the innate resistance conferred by ß-lactam accessory proteins. ß-Lactam compound-compound synthetic lethality may be an alternative approach for drug-resistant tuberculosis.


Subject(s)
Antitubercular Agents/pharmacology , Mycobacterium tuberculosis/drug effects , Synthetic Lethal Mutations/drug effects , Tuberculosis, Multidrug-Resistant/drug therapy , beta-Lactams/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Extensively Drug-Resistant Tuberculosis/drug therapy , Extensively Drug-Resistant Tuberculosis/metabolism , Female , Meropenem/pharmacology , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests/methods , Tuberculosis, Multidrug-Resistant/metabolism , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/metabolism
2.
ChemMedChem ; 14(1): 78-82, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30376607

ABSTRACT

Antibiotics with novel mechanisms of action are desperately needed to combat the increasing rates of multidrug-resistant infections. Bacterial pantothenate kinase (PanK) has emerged as a target of interest to cut off the biosynthesis of coenzyme A. Herein we report the results of an in vitro high-throughput screen of over 10 000 small molecules against Bacillus anthracis PanK, as well as a follow-up screen of hits against PanK isolated from Pseudomonas aeruginosa and Burkholderia cenocepacia. Nine hits are structurally categorized and analyzed to set the stage for future drug development.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacillus anthracis/drug effects , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Bacillus anthracis/enzymology , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Molecular Structure , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
3.
Aging (Albany NY) ; 9(1): 68-97, 2017 01 05.
Article in English | MEDLINE | ID: mdl-28077781

ABSTRACT

A progressive loss of genome maintenance has been implicated as both a cause and consequence of aging. Here we present evidence supporting the hypothesis that an age-associated decay in genome maintenance promotes aging in Saccharomyces cerevisiae (yeast) due to an inability to sense or repair DNA damage by topoisomerase 2 (yTop2). We describe the characterization of LS1, identified in a high throughput screen for small molecules that shorten the replicative lifespan of yeast. LS1 accelerates aging without affecting proliferative growth or viability. Genetic and biochemical criteria reveal LS1 to be a weak Top2 poison. Top2 poisons induce the accumulation of covalent Top2-linked DNA double strand breaks that, if left unrepaired, lead to genome instability and death. LS1 is toxic to cells deficient in homologous recombination, suggesting that the damage it induces is normally mitigated by genome maintenance systems. The essential roles of yTop2 in proliferating cells may come with a fitness trade-off in older cells that are less able to sense or repair yTop2-mediated DNA damage. Consistent with this idea, cells live longer when yTop2 expression levels are reduced. These results identify intrinsic yTop2-mediated DNA damage as potentially manageable cause of aging.


Subject(s)
Cellular Senescence/genetics , DNA Topoisomerases, Type II/genetics , Poisons/pharmacology , Saccharomyces cerevisiae/metabolism , Topoisomerase II Inhibitors/pharmacology , Cellular Senescence/drug effects , DNA Breaks, Double-Stranded/drug effects , DNA Damage/drug effects , Genomic Instability/drug effects , Saccharomyces cerevisiae/genetics
4.
PLoS One ; 11(10): e0163615, 2016.
Article in English | MEDLINE | ID: mdl-27732613

ABSTRACT

Small molecules that correct the folding defects and enhance surface localization of the F508del mutation in the Cystic Fibrosis Transmembrane conductance Regulator (CFTR) comprise an important therapeutic strategy for cystic fibrosis lung disease. However, compounds that rescue the F508del mutant protein to wild type (WT) levels have not been identified. In this report, we consider obstacles to obtaining robust and therapeutically relevant levels of F508del CFTR. For example, markedly diminished steady state amounts of F508del CFTR compared to WT CFTR are present in recombinant bronchial epithelial cell lines, even when much higher levels of mutant transcript are present. In human primary airway cells, the paucity of Band B F508del is even more pronounced, although F508del and WT mRNA concentrations are comparable. Therefore, to augment levels of "repairable" F508del CFTR and identify small molecules that then correct this pool, we developed compound library screening protocols based on automated protein detection. First, cell-based imaging measurements were used to semi-quantitatively estimate distribution of F508del CFTR by high content analysis of two-dimensional images. We evaluated ~2,000 known bioactive compounds from the NIH Roadmap Molecular Libraries Small Molecule Repository in a pilot screen and identified agents that increase the F508del protein pool. Second, we analyzed ~10,000 compounds representing diverse chemical scaffolds for effects on total CFTR expression using a multi-plate fluorescence protocol and describe compounds that promote F508del maturation. Together, our findings demonstrate proof of principle that agents identified in this fashion can augment the level of endoplasmic reticulum (ER) resident "Band B" F508del CFTR suitable for pharmacologic correction. As further evidence in support of this strategy, PYR-41-a compound that inhibits the E1 ubiquitin activating enzyme-was shown to synergistically enhance F508del rescue by C18, a small molecule corrector. Our combined results indicate that increasing the levels of ER-localized CFTR available for repair provides a novel route to correct F508del CFTR.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/pathology , Endoplasmic Reticulum/metabolism , Small Molecule Libraries/chemistry , Alleles , Benzoates/chemistry , Benzoates/pharmacology , Cells, Cultured , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Furans/chemistry , Furans/pharmacology , Gene Deletion , HEK293 Cells , HeLa Cells , High-Throughput Screening Assays , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Microscopy, Fluorescence , Protein Folding , Protein Structure, Tertiary , Pyrazoles/chemistry , Pyrazoles/pharmacology , RNA, Messenger/metabolism , Small Molecule Libraries/pharmacology , Ubiquitination/drug effects , Vorinostat
5.
J Biol Chem ; 291(46): 24188-24199, 2016 Nov 11.
Article in English | MEDLINE | ID: mdl-27679486

ABSTRACT

The enzyme cytochrome c oxidase (CcO) or complex IV (EC 1.9.3.1) is a large transmembrane protein complex that serves as the last enzyme in the respiratory electron transport chain of eukaryotic mitochondria. CcO promotes the switch from glycolytic to oxidative phosphorylation (OXPHOS) metabolism and has been associated with increased self-renewal characteristics in gliomas. Increased CcO activity in tumors has been associated with tumor progression after chemotherapy failure, and patients with primary glioblastoma multiforme and high tumor CcO activity have worse clinical outcomes than those with low tumor CcO activity. Therefore, CcO is an attractive target for cancer therapy. We report here the characterization of a CcO inhibitor (ADDA 5) that was identified using a high throughput screening paradigm. ADDA 5 demonstrated specificity for CcO, with no inhibition of other mitochondrial complexes or other relevant enzymes, and biochemical characterization showed that this compound is a non-competitive inhibitor of cytochrome c When tested in cellular assays, ADDA 5 dose-dependently inhibited the proliferation of chemosensitive and chemoresistant glioma cells but did not display toxicity against non-cancer cells. Furthermore, treatment with ADDA 5 led to significant inhibition of tumor growth in flank xenograft mouse models. Importantly, ADDA 5 inhibited CcO activity and blocked cell proliferation and neurosphere formation in cultures of glioma stem cells, the cells implicated in tumor recurrence and resistance to therapy in patients with glioblastoma. In summary, we have identified ADDA 5 as a lead CcO inhibitor for further optimization as a novel approach for the treatment of glioblastoma and related cancers.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Electron Transport Complex IV/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Glioma , Neoplasm Proteins/antagonists & inhibitors , Animals , Cell Line, Tumor , Cytochromes c/metabolism , Electron Transport Complex IV/metabolism , Glioma/drug therapy , Glioma/enzymology , Humans , Mice , Neoplasm Proteins/metabolism , Xenograft Model Antitumor Assays
6.
Methods Mol Biol ; 1439: 181-95, 2016.
Article in English | MEDLINE | ID: mdl-27316996

ABSTRACT

High-throughput screening is a valuable way to identify hit compounds that combined with a robust medicinal chemistry program could lead to the identification of new antibiotics. Here, we discuss our method for screening large compound libraries with virulent Mycobacterium tuberculosis, possibly one of the more difficult bacteria to use because of its slow growth and assignment to Biosafety Level-3 by the CDC and NIH. The principles illuminated here, however, are relevant to the execution of most bacteria high-throughput screens.


Subject(s)
Antitubercular Agents/pharmacology , High-Throughput Screening Assays/methods , Microbial Sensitivity Tests/methods , Mycobacterium tuberculosis/drug effects , Tuberculosis/drug therapy , Animals , Cell Culture Techniques/methods , Chlorocebus aethiops , Humans , Mycobacterium tuberculosis/growth & development , Small Molecule Libraries/pharmacology , Tuberculosis/microbiology , Vero Cells
7.
Am J Respir Crit Care Med ; 194(9): 1092-1103, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27104944

ABSTRACT

RATIONALE: Premature termination codons (PTCs) in the cystic fibrosis transmembrane conductance regulator (CFTR) gene cause cystic fibrosis (CF). Several agents are known to suppress PTCs but are poorly efficacious or toxic. OBJECTIVES: To determine whether there are clinically available agents that elicit translational readthrough and improve CFTR function sufficient to confer therapeutic benefit to patients with CF with PTCs. METHODS: Two independent screens, firefly luciferase and CFTR-mediated transepithelial chloride conductance assay, were performed on a library of 1,600 clinically approved compounds using fisher rat thyroid cells stably transfected with stop codons. Select agents were further evaluated using secondary screening assays including short circuit current analysis on primary cells from patients with CF. In addition, the effect of CFTR modulators (ivacaftor) was tested in combination with the most efficacious agents. MEASUREMENTS AND MAIN RESULTS: From the primary screen, 48 agents were selected as potentially active. Following confirmatory tests in the transepithelial chloride conductance assay and prioritizing agents based on favorable pharmacologic properties, eight agents were advanced for secondary screening. Ivacaftor significantly increased short circuit current following forskolin stimulation in cells treated with pyranoradine tetraphosphate, potassium p-aminobenzoate, and escin as compared with vehicle control. Escin, an herbal agent, consistently induced readthrough activity as demonstrated by enhanced CFTR expression and function in vitro. CONCLUSIONS: Clinically approved drugs identified as potential readthrough agents, in combination with ivacaftor, may induce nonsense suppression to restore therapeutic levels of CFTR function. One or more agents may be suitable to advance to human testing.


Subject(s)
Codon, Nonsense/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/drug therapy , Drug Discovery/methods , Animals , Cell Line , Codon, Nonsense/genetics , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Drug Evaluation, Preclinical/methods , Humans , Luciferases/metabolism , Rats, Inbred F344 , Real-Time Polymerase Chain Reaction
8.
Biochem J ; 473(8): 1027-35, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26846349

ABSTRACT

Historically, drugs used in the treatment of cancers also tend to cause damage to healthy cells while affecting cancer cells. Therefore, the identification of novel agents that act specifically against cancer cells remains a high priority in the search for new therapies. In contrast with normal cells, most cancer cells contain multiple centrosomes which are associated with genome instability and tumorigenesis. Cancer cells can avoid multipolar mitosis, which can cause cell death, by clustering the extra centrosomes into two spindle poles, thereby enabling bipolar division. Kinesin-like protein KIFC1 plays a critical role in centrosome clustering in cancer cells, but is not essential for normal cells. Therefore, targeting KIFC1 may provide novel insight into selective killing of cancer cells. In the present study, we identified a small-molecule KIFC1 inhibitor, SR31527, which inhibited microtubule (MT)-stimulated KIFC1 ATPase activity with an IC50 value of 6.6 µM. By using bio layer interferometry technology, we further demonstrated that SR31527 bound directly to KIFC1 with high affinity (Kd=25.4 nM). Our results from computational modelling and saturation-transfer difference (STD)-NMR experiments suggest that SR31527 bound to a novel allosteric site of KIFC1 that appears suitable for developing selective inhibitors of KIFC1. Importantly, SR31527 prevented bipolar clustering of extra centrosomes in triple negative breast cancer (TNBC) cells and significantly reduced TNBC cell colony formation and viability, but was less toxic to normal fibroblasts. Therefore, SR31527 provides a valuable tool for studying the biological function of KIFC1 and serves as a potential lead for the development of novel therapeutic agents for breast cancer treatment.


Subject(s)
Drug Discovery , Kinesins/antagonists & inhibitors , Kinesins/metabolism , Thiadiazoles/chemistry , Thiadiazoles/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Drug Discovery/methods , Humans , Kinesins/chemistry , Protein Binding/physiology , Protein Structure, Secondary , Thiadiazoles/pharmacology
9.
J Lab Autom ; 21(1): 198-203, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26663785

ABSTRACT

The development of acoustic droplet ejection (ADE) technology has resulted in many positive changes associated with the operations in a high-throughput screening (HTS) laboratory. Originally, this liquid transfer technology was used to simply transfer DMSO solutions of primarily compounds. With the introduction of Labcyte's Echo 555, which has aqueous dispense capability, the application of this technology has been expanded beyond its original use. This includes the transfer of many biological reagents solubilized in aqueous buffers, including siRNAs. The Echo 555 is ideal for siRNA dispensing because it is accurate at low volumes and a step-down dilution is not necessary. The potential for liquid carryover and cross-contamination is eliminated, as no tips are needed. Herein, we describe the siRNA screening platform at Southern Research's HTS Center using the ADE technology. With this technology, an siRNA library can be dispensed weeks or even months in advance of the assay itself. The protocol has been optimized to achieve assay parameters comparable to small-molecule screening parameters, and exceeding the norm reported for genomewide siRNA screens.


Subject(s)
Biomedical Technology/methods , Genetic Association Studies/methods , High-Throughput Screening Assays/methods , RNA Interference , Acoustics , Solutions
10.
J Lab Autom ; 21(1): 188-97, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26663786

ABSTRACT

When acoustic droplet ejection technology was first introduced for high-throughput applications, it was used primarily for dispensing compounds dissolved in DMSO. The high precision and accuracy achieved for low-volume transfers in this application were noted by those working outside of the compound management area, and interest was generated in expanding the scope of the technology to include other liquid types. Later-generation instruments included calibrations for several aqueous buffers that were applicable to the life sciences. The High Throughput Screening Center at Southern Research has made use of this range of liquid calibrations for the Infectious Disease Program. The original calibration for DMSO has allowed the preparation of assay-ready plates that can be sent to remote locations. This process was used as part of the collaboration between Southern Research and Galveston National Laboratory, University of Texas Medical Branch, to develop high-throughput screening for biological safety level 4 containment and to provide compounds for two pilot screens that were run there with BSL-4-level pathogens. The aqueous calibrations have been instrumental in miniaturizing assays used for infectious disease, such as qPCR, tissue culture infectious dose 50, and bacterial motility, to make them compatible with HTS operations.


Subject(s)
High-Throughput Screening Assays/methods , Microbiological Techniques/methods , Specimen Handling/methods , Acoustics , Containment of Biohazards/methods , Solutions , Texas
11.
Cell ; 161(6): 1252-65, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26046436

ABSTRACT

Small-molecule probes can illuminate biological processes and aid in the assessment of emerging therapeutic targets by perturbing biological systems in a manner distinct from other experimental approaches. Despite the tremendous promise of chemical tools for investigating biology and disease, small-molecule probes were unavailable for most targets and pathways as recently as a decade ago. In 2005, the NIH launched the decade-long Molecular Libraries Program with the intent of innovating in and broadening access to small-molecule science. This Perspective describes how novel small-molecule probes identified through the program are enabling the exploration of biological pathways and therapeutic hypotheses not otherwise testable. These experiences illustrate how small-molecule probes can help bridge the chasm between biological research and the development of medicines but also highlight the need to innovate the science of therapeutic discovery.


Subject(s)
Drug Discovery , Small Molecule Libraries , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Humans , National Institutes of Health (U.S.) , United States
12.
J Biol Chem ; 290(16): 10504-17, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25724652

ABSTRACT

APOBEC3G (A3G) is a cellular cytidine deaminase that restricts HIV-1 replication by inducing G-to-A hypermutation in viral DNA and by deamination-independent mechanisms. HIV-1 Vif binds to A3G, resulting in its degradation via the 26 S proteasome. Therefore, this interaction represents a potential therapeutic target. To identify compounds that inhibit interaction between A3G and HIV-1 Vif in a high throughput format, we developed a homogeneous time-resolved fluorescence resonance energy transfer assay. A 307,520 compound library from the NIH Molecular Libraries Small Molecule Repository was screened. Secondary screens to evaluate dose-response performance and off-target effects, cell-based assays to identify compounds that attenuate Vif-dependent degradation of A3G, and assays testing antiviral activity in peripheral blood mononuclear cells and T cells were employed. One compound, N.41, showed potent antiviral activity in A3G(+) but not in A3G(-) T cells and had an IC50 as low as 8.4 µM and a TC50 of >100 µM when tested against HIV-1Ba-L replication in peripheral blood mononuclear cells. N.41 inhibited the Vif-A3G interaction and increased cellular A3G levels and incorporation of A3G into virions, thereby attenuating virus infectivity in a Vif-dependent manner. N.41 activity was also species- and Vif-dependent. Preliminary structure-activity relationship studies suggest that a hydroxyl moiety located at a phenylamino group is critical for N.41 anti-HIV activity and identified N.41 analogs with better potency (IC50 as low as 4.2 µM). These findings identify a new lead compound that attenuates HIV replication by liberating A3G from Vif regulation and increasing its innate antiviral activity.


Subject(s)
Anti-HIV Agents/pharmacology , Cytidine Deaminase/genetics , HIV-1/drug effects , Leukocytes, Mononuclear/drug effects , Small Molecule Libraries/pharmacology , T-Lymphocytes/drug effects , vif Gene Products, Human Immunodeficiency Virus/genetics , APOBEC-3G Deaminase , Anti-HIV Agents/chemistry , Biological Assay , Cell Line , Cytidine Deaminase/metabolism , Fluorescence Resonance Energy Transfer , Gene Expression Regulation , HEK293 Cells , HIV-1/genetics , HIV-1/metabolism , Host-Pathogen Interactions , Humans , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Primary Cell Culture , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Signal Transduction , Small Molecule Libraries/chemistry , Structure-Activity Relationship , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Virus Replication/drug effects , vif Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , vif Gene Products, Human Immunodeficiency Virus/metabolism
13.
Assay Drug Dev Technol ; 13(1): 44-54, 2015.
Article in English | MEDLINE | ID: mdl-25710545

ABSTRACT

High-throughput screening (HTS) has been integrated into the drug discovery process, and multiple assay formats have been widely used in many different disease areas but with limited focus on infectious agents. In recent years, there has been an increase in the number of HTS campaigns using infectious wild-type pathogens rather than surrogates or biochemical pathogen-derived targets. Concurrently, enhanced emerging pathogen surveillance and increased human mobility have resulted in an increase in the emergence and dissemination of infectious human pathogens with serious public health, economic, and social implications at global levels. Adapting the HTS drug discovery process to biocontainment laboratories to develop new drugs for these previously uncharacterized and highly pathogenic agents is now feasible, but HTS at higher biosafety levels (BSL) presents a number of unique challenges. HTS has been conducted with multiple bacterial and viral pathogens at both BSL-2 and BSL-3, and pilot screens have recently been extended to BSL-4 environments for both Nipah and Ebola viruses. These recent successful efforts demonstrate that HTS can be safely conducted at the highest levels of biological containment. This review outlines the specific issues that must be considered in the execution of an HTS drug discovery program for high-containment pathogens. We present an overview of the requirements for HTS in high-level biocontainment laboratories.


Subject(s)
Biological Assay/instrumentation , Containment of Biohazards/instrumentation , Drug Evaluation, Preclinical/instrumentation , High-Throughput Screening Assays/instrumentation , Laboratories , Technology, Pharmaceutical/instrumentation , Drug Design , Equipment Design , Equipment Failure Analysis , Robotics/instrumentation , Specimen Handling/instrumentation
14.
J Med Chem ; 57(24): 10314-28, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25399509

ABSTRACT

A quinazolinedione-derived screening hit 2 was discovered with cellular antiviral activity against respiratory syncytial virus (CPE EC50 = 2.1 µM), moderate efficacy in reducing viral progeny (4.2 log at 10 µM), and marginal cytotoxic liability (selectivity index, SI ∼ 24). Scaffold optimization delivered analogs with improved potency and selectivity profiles. Most notable were compounds 15 and 19 (EC50 = 300-500 nM, CC50 > 50 µM, SI > 100), which significantly reduced viral titer (>400,000-fold), and several analogs were shown to block the activity of the RNA-dependent RNA-polymerase complex of RSV.


Subject(s)
Antiviral Agents/pharmacology , Benzamides/pharmacology , Cell Proliferation/drug effects , Quinazolinones/pharmacology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/drug effects , Virus Replication/drug effects , Antiviral Agents/chemical synthesis , Benzamides/chemical synthesis , Drug Design , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Quinazolinones/chemical synthesis , Quinazolinones/chemistry , RNA-Dependent RNA Polymerase/metabolism , Respiratory Syncytial Virus Infections/virology , Structure-Activity Relationship
15.
J Med Chem ; 57(20): 8608-21, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25244572

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is an emerging pathogenic alphavirus that can cause significant disease in humans. Given the absence of therapeutic options available and the significance of VEEV as a weaponized agent, an optimization effort was initiated around a quinazolinone screening hit 1 with promising cellular antiviral activity (EC50 = 0.8 µM), limited cytotoxic liability (CC50 > 50 µM), and modest in vitro efficacy in reducing viral progeny (63-fold at 5 µM). Scaffold optimization revealed a novel rearrangement affording amidines, specifically compound 45, which was found to potently inhibit several VEEV strains in the low nanomolar range without cytotoxicity (EC50 = 0.02-0.04 µM, CC50 > 50 µM) while limiting in vitro viral replication (EC90 = 0.17 µM). Brain exposure was observed in mice with 45. Significant protection was observed in VEEV-infected mice at 5 mg kg(-1) day(-1) and viral replication appeared to be inhibited through interference of viral nonstructural proteins.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Benzamides/pharmacology , Encephalitis Virus, Venezuelan Equine/drug effects , Piperazines/pharmacology , Animals , Benzamides/chemistry , Drug Evaluation, Preclinical/methods , Encephalomyelitis, Venezuelan Equine/drug therapy , Heterocyclic Compounds, 2-Ring/chemistry , Mice, Inbred C3H , Mice, Inbred C57BL , Piperazines/chemistry , Quinazolinones/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Virus Replication/drug effects
16.
J Biol Chem ; 289(47): 32937-51, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25228699

ABSTRACT

Pathogenic mutations in the LRRK2 gene can cause late-onset Parkinson disease. The most common mutation, G2019S, resides in the kinase domain and enhances activity. LRRK2 possesses the unique property of cis-autophosphorylation of its own GTPase domain. Because high-resolution structures of the human LRRK2 kinase domain are not available, we used novel high-throughput assays that measured both cis-autophosphorylation and trans-peptide phosphorylation to probe the ATP-binding pocket. We disclose hundreds of commercially available activity-selective LRRK2 kinase inhibitors. Some compounds inhibit cis-autophosphorylation more strongly than trans-peptide phosphorylation, and other compounds inhibit G2019S-LRRK2 more strongly than WT-LRRK2. Through exploitation of structure-activity relationships revealed through high-throughput analyses, we identified a useful probe inhibitor, SRI-29132 (11). SRI-29132 is exquisitely selective for LRRK2 kinase activity and is effective in attenuating proinflammatory responses in macrophages and rescuing neurite retraction phenotypes in neurons. Furthermore, the compound demonstrates excellent potency, is highly blood-brain barrier-permeant, but suffers from rapid first-pass metabolism. Despite the observed selectivity of SRI-29132, docking models highlighted critical interactions with residues conserved in many protein kinases, implying a unique structural configuration for the LRRK2 ATP-binding pocket. Although the human LRRK2 kinase domain is unstable and insoluble, we demonstrate that the LRRK2 homolog from ameba can be mutated to approximate some aspects of the human LRRK2 ATP-binding pocket. Our results provide a rich resource for LRRK2 small molecule inhibitor development. More broadly, our results provide a precedent for the functional interrogation of ATP-binding pockets when traditional approaches to ascertain structure prove difficult.


Subject(s)
Adenosine Triphosphate/chemistry , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , Biocatalysis/drug effects , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Hep G2 Cells , Humans , Kinetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mice , Models, Molecular , Molecular Sequence Data , Molecular Structure , Mutation , Phosphorylation/drug effects , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , Pyridazines/chemistry , Pyridazines/metabolism , Pyridazines/pharmacology , Sequence Homology, Amino Acid , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Triazoles/chemistry , Triazoles/metabolism , Triazoles/pharmacology
17.
J Biomol Screen ; 19(10): 1338-49, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25156556

ABSTRACT

Alzheimer disease (AD) is the most common neurodegenerative disease, and with Americans' increasing longevity, it is becoming an epidemic. There are currently no effective treatments for this disorder. Abnormalities of Tau track more closely with cognitive decline than the most studied therapeutic target in AD, amyloid-ß, but the optimal strategy for targeting Tau has not yet been identified. On the basis of considerable preclinical data from AD models, we hypothesize that interactions between Tau and the Src-family tyrosine kinase, Fyn, are pathogenic in AD. Genetically reducing either Tau or Fyn is protective in AD mouse models, and a dominant negative fragment of Tau that alters Fyn localization is also protective. Here, we describe a new AlphaScreen assay and a live-cell bioluminescence resonance energy transfer (BRET) assay using a novel BRET pair for quantifying the Tau-Fyn interaction. We used these assays to map the binding site on Tau for Fyn to the fifth and sixth PXXP motifs to show that AD-associated phosphorylation at microtubule affinity regulating kinase sites increases the affinity of the Tau-Fyn interaction and to identify Tau-Fyn interaction inhibitors by high-throughput screening. This screen has identified a variety of chemically tractable hits, suggesting that the Tau-Fyn interaction may represent a good drug target for AD.


Subject(s)
Bioluminescence Resonance Energy Transfer Techniques/methods , High-Throughput Screening Assays/methods , Proto-Oncogene Proteins c-fyn/metabolism , Small Molecule Libraries/pharmacology , tau Proteins/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Humans , Molecular Sequence Data , Phosphorylation , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-fyn/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , src Homology Domains , tau Proteins/genetics
18.
PLoS One ; 9(7): e96054, 2014.
Article in English | MEDLINE | ID: mdl-24983234

ABSTRACT

Uropathogenic Escherichia coli (UPEC) is the leading cause of community-acquired urinary tract infections (UTIs), with over 100 million UTIs occurring annually throughout the world. Increasing antimicrobial resistance among UPEC limits ambulatory care options, delays effective treatment, and may increase overall morbidity and mortality from complications such as urosepsis. The polysaccharide capsules of UPEC are an attractive target a therapeutic, based on their importance in defense against the host immune responses; however, the large number of antigenic types has limited their incorporation into vaccine development. The objective of this study was to identify small-molecule inhibitors of UPEC capsule biogenesis. A large-scale screening effort entailing 338,740 compounds was conducted in a cell-based, phenotypic screen for inhibition of capsule biogenesis in UPEC. The primary and concentration-response assays yielded 29 putative inhibitors of capsule biogenesis, of which 6 were selected for further studies. Secondary confirmatory assays identified two highly active agents, named DU003 and DU011, with 50% inhibitory concentrations of 1.0 µM and 0.69 µM, respectively. Confirmatory assays for capsular antigen and biochemical measurement of capsular sugars verified the inhibitory action of both compounds and demonstrated minimal toxicity and off-target effects. Serum sensitivity assays demonstrated that both compounds produced significant bacterial death upon exposure to active human serum. DU011 administration in mice provided near complete protection against a lethal systemic infection with the prototypic UPEC K1 isolate UTI89. This work has provided a conceptually new class of molecules to combat UPEC infection, and future studies will establish the molecular basis for their action along with efficacy in UTI and other UPEC infections.


Subject(s)
Anti-Bacterial Agents , Bacterial Capsules/metabolism , Escherichia coli Infections/drug therapy , Urinary Tract Infections/drug therapy , Uropathogenic Escherichia coli/metabolism , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Escherichia coli Infections/pathology , Female , Humans , Mice , Urinary Tract Infections/pathology
19.
PLoS Pathog ; 10(6): e1004213, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24967809

ABSTRACT

Alphaviruses present serious health threats as emerging and re-emerging viruses. Venezuelan equine encephalitis virus (VEEV), a New World alphavirus, can cause encephalitis in humans and horses, but there are no therapeutics for treatment. To date, compounds reported as anti-VEEV or anti-alphavirus inhibitors have shown moderate activity. To discover new classes of anti-VEEV inhibitors with novel viral targets, we used a high-throughput screen based on the measurement of cell protection from live VEEV TC-83-induced cytopathic effect to screen a 340,000 compound library. Of those, we identified five novel anti-VEEV compounds and chose a quinazolinone compound, CID15997213 (IC50 = 0.84 µM), for further characterization. The antiviral effect of CID15997213 was alphavirus-specific, inhibiting VEEV and Western equine encephalitis virus, but not Eastern equine encephalitis virus. In vitro assays confirmed inhibition of viral RNA, protein, and progeny synthesis. No antiviral activity was detected against a select group of RNA viruses. We found mutations conferring the resistance to the compound in the N-terminal domain of nsP2 and confirmed the target residues using a reverse genetic approach. Time of addition studies showed that the compound inhibits the middle stage of replication when viral genome replication is most active. In mice, the compound showed complete protection from lethal VEEV disease at 50 mg/kg/day. Collectively, these results reveal a potent anti-VEEV compound that uniquely targets the viral nsP2 N-terminal domain. While the function of nsP2 has yet to be characterized, our studies suggest that the protein might play a critical role in viral replication, and further, may represent an innovative opportunity to develop therapeutic interventions for alphavirus infection.


Subject(s)
Antiviral Agents/pharmacology , Encephalitis Virus, Venezuelan Equine/drug effects , Encephalomyelitis, Venezuelan Equine/drug therapy , Quinazolinones/pharmacology , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Disease Models, Animal , Drug Resistance, Viral/genetics , Encephalitis Virus, Venezuelan Equine/genetics , Encephalomyelitis, Venezuelan Equine/virology , High-Throughput Screening Assays , Mice , Mice, Inbred C3H , Species Specificity , Structure-Activity Relationship , Vero Cells , Viral Plaque Assay , Virus Replication/drug effects
20.
Assay Drug Dev Technol ; 12(3): 155-61, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24735442

ABSTRACT

Nipah virus is a biosafety level 4 (BSL-4) pathogen that causes severe respiratory illness and encephalitis in humans. To identify novel small molecules that target Nipah virus replication as potential therapeutics, Southern Research Institute and Galveston National Laboratory jointly developed an automated high-throughput screening platform that is capable of testing 10,000 compounds per day within BSL-4 biocontainment. Using this platform, we screened a 10,080-compound library using a cell-based, high-throughput screen for compounds that inhibited the virus-induced cytopathic effect. From this pilot effort, 23 compounds were identified with EC50 values ranging from 3.9 to 20.0 µM and selectivities >10. Three sulfonamide compounds with EC50 values <12 µM were further characterized for their point of intervention in the viral replication cycle and for broad antiviral efficacy. Development of HTS capability under BSL-4 containment changes the paradigm for drug discovery for highly pathogenic agents because this platform can be readily modified to identify prophylactic and postexposure therapeutic candidates against other BSL-4 pathogens, particularly Ebola, Marburg, and Lassa viruses.


Subject(s)
Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Drug Evaluation, Preclinical/instrumentation , High-Throughput Screening Assays/instrumentation , Nipah Virus/drug effects , Nipah Virus/physiology , Sulfonamides/antagonists & inhibitors , Virus Replication/drug effects , Animals , Chlorocebus aethiops , Containment of Biohazards/instrumentation , Dose-Response Relationship, Drug , Equipment Design , Equipment Failure Analysis , Robotics/instrumentation , Vero Cells , Virus Replication/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...