Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
mBio ; 13(3): e0078422, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35471084

ABSTRACT

The main protease, Mpro, of SARS-CoV-2 is required to cleave the viral polyprotein into precise functional units for virus replication and pathogenesis. Here, we report quantitative reporters for Mpro function in living cells in which protease inhibition by genetic or chemical methods results in robust signal readouts by fluorescence (enhanced green fluorescent protein [eGFP]) or bioluminescence (firefly luciferase). These gain-of-signal systems are scalable to high-throughput platforms for quantitative discrimination between Mpro mutants and/or inhibitor potencies as evidenced by validation of several reported inhibitors. Additional utility is shown by single Mpro amino acid variants and structural information combining to demonstrate that both inhibitor conformational dynamics and amino acid differences are able to influence inhibitor potency. We further show that a recent variant of concern (Omicron) has an unchanged response to a clinically approved drug, nirmatrelvir, whereas proteases from divergent coronavirus species show differential susceptibility. Together, we demonstrate that these gain-of-signal systems serve as robust, facile, and scalable assays for live cell quantification of Mpro inhibition, which will help expedite the development of next-generation antivirals and enable the rapid testing of emerging variants. IMPORTANCE The main protease, Mpro, of SARS-CoV-2 is an essential viral protein required for the earliest steps of infection. It is therefore an attractive target for antiviral drug development. Here, we report the development and implementation of two complementary cell-based systems for quantification of Mpro inhibition by genetic or chemical approaches. The first is fluorescence based (eGFP), and the second is luminescence based (firefly luciferase). Importantly, both systems rely upon gain-of-signal readouts such that stronger inhibitors yield higher fluorescent or luminescent signal. The high versatility and utility of these systems are demonstrated by characterizing Mpro mutants and natural variants, including Omicron, as well as a panel of existing inhibitors. These systems rapidly, safely, and sensitively identify Mpro variants with altered susceptibilities to inhibition, triage-nonspecific, or off-target molecules and validate bona fide inhibitors, with the most potent thus far being the first-in-class drug nirmatrelvir.


Subject(s)
Antiviral Agents , Coronavirus 3C Proteases , Protease Inhibitors , SARS-CoV-2 , Amino Acids , Antiviral Agents/pharmacology , Coronavirus 3C Proteases/antagonists & inhibitors , Luciferases, Firefly , Protease Inhibitors/pharmacology , SARS-CoV-2/drug effects , SARS-CoV-2/genetics
2.
SLAS Discov ; 27(1): 8-19, 2022 01.
Article in English | MEDLINE | ID: mdl-35058179

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 responsible for COVID-19 remains a persistent threat to mankind, especially for the immunocompromised and elderly for which the vaccine may have limited effectiveness. Entry of SARS-CoV-2 requires a high affinity interaction of the viral spike protein with the cellular receptor angiotensin-converting enzyme 2. Novel mutations on the spike protein correlate with the high transmissibility of new variants of SARS-CoV-2, highlighting the need for small molecule inhibitors of virus entry into target cells. We report the identification of such inhibitors through a robust high-throughput screen testing 15,000 small molecules from unique libraries. Several leads were validated in a suite of mechanistic assays, including whole cell SARS-CoV-2 infectivity assays. The main lead compound, calpeptin, was further characterized using SARS-CoV-1 and the novel SARS-CoV-2 variant entry assays, SARS-CoV-2 protease assays and molecular docking. This study reveals calpeptin as a potent and specific inhibitor of SARS-CoV-2 and some variants.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Virus Attachment/drug effects , Virus Internalization/drug effects , Angiotensin-Converting Enzyme 2/metabolism , Animals , Cathepsin L/antagonists & inhibitors , Cell Line , Chlorocebus aethiops , Drug Evaluation, Preclinical , Drug Repositioning , HEK293 Cells , Humans , Molecular Docking Simulation , SARS-CoV-2/drug effects , SARS-CoV-2/growth & development , Serine Endopeptidases/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Vero Cells
3.
PLoS Negl Trop Dis ; 15(11): e0009898, 2021 11.
Article in English | MEDLINE | ID: mdl-34731172

ABSTRACT

Given the worldwide burden of neglected tropical diseases, there is ongoing need to develop novel anthelmintic agents to strengthen the pipeline of drugs to combat these burdensome infections. Many diseases caused by parasitic flatworms are treated using the anthelmintic drug praziquantel (PZQ), employed for decades as the key clinical agent to treat schistosomiasis. PZQ activates a flatworm transient receptor potential (TRP) channel within the melastatin family (TRPMPZQ) to mediate sustained Ca2+ influx and worm paralysis. As a druggable target present in many parasitic flatworms, TRPMPZQ is a promising target for a target-based screening campaign with the goal of discovering novel regulators of this channel complex. Here, we have optimized methods to miniaturize a Ca2+-based reporter assay for Schistosoma mansoni TRPMPZQ (Sm.TRPMPZQ) activity enabling a high throughput screening (HTS) approach. This methodology will enable further HTS efforts against Sm.TRPMPZQ as well as other flatworm ion channels. A pilot screen of ~16,000 compounds yielded a novel activator of Sm.TRPMPZQ, and numerous potential blockers. The new activator of Sm.TRPMPZQ represented a distinct chemotype to PZQ, but is a known chemical entity previously identified by phenotypic screening. The fact that a compound prioritized from a phenotypic screening campaign is revealed to act, like PZQ, as an Sm.TRPMPZQ agonist underscores the validity of TRPMPZQ as a druggable target for antischistosomal ligands.


Subject(s)
Anthelmintics/pharmacology , Helminth Proteins/antagonists & inhibitors , Praziquantel/pharmacology , Schistosoma mansoni/metabolism , Schistosomiasis mansoni/parasitology , Transient Receptor Potential Channels/antagonists & inhibitors , Animals , Anthelmintics/chemistry , Calcium/metabolism , Drug Evaluation, Preclinical , Female , Helminth Proteins/genetics , Helminth Proteins/metabolism , Humans , Male , Mice , Praziquantel/chemistry , Schistosoma mansoni/drug effects , Schistosoma mansoni/genetics , Schistosomiasis mansoni/drug therapy , Schistosomiasis mansoni/metabolism , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism
4.
SLAS Discov ; 26(9): 1177-1188, 2021 10.
Article in English | MEDLINE | ID: mdl-34112017

ABSTRACT

Regulators of G protein signaling (RGS) proteins serve as critical regulatory nodes to limit the lifetime and extent of signaling via G protein-coupled receptors (GPCRs). Previously, approaches to pharmacologically inhibit RGS activity have mostly focused on the inhibition of GTPase activity by interrupting the interaction of RGS proteins with the G proteins they regulate. However, several RGS proteins are also regulated by association with binding partners. A notable example is the mammalian RGS7 protein, which has prominent roles in metabolic control, vision, reward, and actions of opioid analgesics. In vivo, RGS7 exists in complex with the binding partners type 5 G protein ß subunit (Gß5) and R7 binding protein (R7BP), which control its stability and activity, respectively. Targeting the whole RGS7/Gß5/R7BP protein complex affords the opportunity to allosterically tune opioid receptor signaling following opioid engagement while potentially bypassing undesirable side effects. Hence, we implemented a novel strategy to pharmacologically target the interaction between RGS7/Gß5 and R7BP. To do so, we searched for protein complex inhibitors using a time-resolved fluorescence resonance energy transfer (FRET)-based high-throughput screening (HTS) assay that measures compound-mediated alterations in the FRET signal between RGS7/Gß5 and R7BP. We performed two HTS campaigns, each screening ~100,000 compounds from the Scripps Drug Discovery Library (SDDL). Each screen yielded more than 100 inhibitors, which will be described herein.


Subject(s)
Drug Discovery , GTP-Binding Protein beta Subunits/metabolism , Multiprotein Complexes/metabolism , RGS Proteins/metabolism , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Humans , Multiprotein Complexes/agonists , Multiprotein Complexes/antagonists & inhibitors , Protein Binding/drug effects , Small Molecule Libraries
5.
SLAS Discov ; 26(2): 205-215, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33016182

ABSTRACT

Cystic fibrosis (CF) is caused by a mutation of the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) gene, which disrupts an ion channel involved in hydration maintenance via anion homeostasis. Nearly 5% of CF patients possess one or more copies of the G542X allele, which results in a stop codon at residue 542, preventing full-length CFTR protein synthesis. Identifying small-molecule modulators of mutant CFTR biosynthesis that affect the readthrough of this and other premature termination codons to synthesize a fully functional CFTR protein represents a novel target area of drug discovery. We describe the implementation and integration for large-scale screening of a homogeneous, 1536-well functional G542X-CFTR readthrough assay. The assay uses HEK 293 cells engineered to overexpress the G542X-CFTR mutant, whose functional activity is monitored with a membrane potential dye. Cells are co-incubated with a CFTR amplifier and CFTR corrector to maximize mRNA levels and trafficking of CFTR to the cell surface. Compounds that allow translational readthrough and synthesis of functional CFTR chloride channels are reflected by changes in membrane potential in response to cAMP stimulation with forskolin and CFTR channel potentiation with genistein. Assay statistics yielded Z' values of 0.69 ± 0.06. As further evidence of its suitability for high-throughput screening, we completed automated screening of approximately 666,000 compounds, identifying 7761 initial hits. Following secondary and tertiary assays, we identified 188 confirmed hit compounds with low and submicromolar potencies. Thus, this approach takes advantage of a phenotypic screen with high-throughput scalability to identify new small-molecule G542X-CFTR readthrough modulators.


Subject(s)
Codon, Nonsense , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Drug Discovery/methods , Gene Expression Regulation/drug effects , Protein Biosynthesis/drug effects , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Drug Evaluation, Preclinical/methods , Gene Expression , Genes, Reporter , HEK293 Cells , High-Throughput Screening Assays/methods , Humans , Plasmids/genetics , Small Molecule Libraries , Transfection/methods
6.
Sci Rep ; 10(1): 10579, 2020 06 29.
Article in English | MEDLINE | ID: mdl-32601341

ABSTRACT

Molecules that correct the folding of protein mutants, restoring their functional trafficking, are called pharmacoperones. Most are clinically irrelevant and possess intrinsic antagonist or agonist activity. Here, we identify compounds capable of rescuing the activity of mutant gonadotropin-releasing hormone receptor or GnRHR which, is sequestered within the cell and if dysfunctional leads to Hypogonadotropic Hypogonadism. To do this we screened the E90K GnRHR mutant vs. a library of 645,000 compounds using a cell-based calcium detection system. Ultimately, we identified 399 compounds with EC50 ≤ 5 µM with no effect in counterscreen assays. Medicinal chemistry efforts confirmed activity of 70 pure samples and mode of action studies, including radioligand binding, inositol phosphate, and toxicity assays, proved that we have a series of tractable compounds that can be categorized into structural clusters. These early lead molecules rescue mutant GnRHR function and are neither agonist nor antagonists of the GnRHR cognate receptor, a feature required for potential clinical utility.


Subject(s)
Receptors, LHRH/agonists , Receptors, LHRH/metabolism , Small Molecule Libraries/pharmacology , Calcium/metabolism , Drug Evaluation, Preclinical , Gonadotropin-Releasing Hormone/agonists , Gonadotropin-Releasing Hormone/metabolism , HeLa Cells , High-Throughput Screening Assays , Humans , Inositol Phosphates/metabolism , Mutation , Protein Folding , Protein Transport , Receptors, LHRH/genetics
7.
Sci Rep ; 9(1): 9000, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31227747

ABSTRACT

Neurons created from human induced pluripotent stem cells (hiPSCs) provide the capability of identifying biological mechanisms that underlie brain disorders. IPSC-derived human neurons, or iNs, hold promise for advancing precision medicine through drug screening, though it remains unclear to what extent iNs can support early-stage drug discovery efforts in industrial-scale screening centers. Despite several reported approaches to generate iNs from iPSCs, each suffer from technological limitations that challenge their scalability and reproducibility, both requirements for successful screening assays. We addressed these challenges by initially removing the roadblocks related to scaling of iNs for high throughput screening (HTS)-ready assays. We accomplished this by simplifying the production and plating of iNs and adapting them to a freezer-ready format. We then tested the performance of freezer-ready iNs in an HTS-amenable phenotypic assay that measured neurite outgrowth. This assay successfully identified small molecule inhibitors of neurite outgrowth. Importantly, we provide evidence that this scalable iN-based assay was both robust and highly reproducible across different laboratories. These streamlined approaches are compatible with any iPSC line that can produce iNs. Thus, our findings indicate that current methods for producing iPSCs are appropriate for large-scale drug-discovery campaigns (i.e. >10e5 compounds) that read out simple neuronal phenotypes. However, due to the inherent limitations of currently available iN differentiation protocols, technological advances are required to achieve similar scalability for screens that require more complex phenotypes related to neuronal function.


Subject(s)
Cell Differentiation/physiology , High-Throughput Screening Assays/methods , Induced Pluripotent Stem Cells/physiology , Neurons/physiology , Biological Assay/methods , Cells, Cultured , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Humans , Induced Pluripotent Stem Cells/cytology , Neuronal Outgrowth/drug effects , Neuronal Outgrowth/physiology , Neurons/cytology , Phenotype , Reproducibility of Results
8.
SLAS Technol ; 24(4): 420-428, 2019 08.
Article in English | MEDLINE | ID: mdl-31225974

ABSTRACT

Affordable and physiologically relevant three-dimensional (3D) cell-based assays used in high-throughput screening (HTS) are on the rise in early drug discovery. These technologies have been aided by the recent adaptation of novel microplate treatments and spheroid culturing techniques. One such technology involves the use of nanoparticle (NanoShuttle-PL) labeled cells and custom magnetic drives to assist in cell aggregation to ensure rapid 3D structure formation after the cells have been dispensed into microtiter plates. Transitioning this technology from a low-throughput manual benchtop application, as previously published by our lab, into a robotically enabled format achieves orders of magnitude greater throughput but required the development of specialized support hardware. This effort included in-house development, fabrication, and testing of ancillary devices that assist robotic handing and high-precision placement of microtiter plates into an incubator embedded with magnetic drives. Utilizing a "rapid prototyping" approach facilitated by cloud-based computer-aided design software, we built the necessary components using hobby-grade 3D printers with turnaround times that rival those of traditional manufacturing/development practices at a substantially reduced cost. This approach culminated in a first-in-class HTS-compatible 3D system in which we have coupled 3D bioprinting to a fully automated HTS robotic platform utilizing our novel magnetic incubator shelf assemblies.


Subject(s)
Automation, Laboratory/methods , Cell Culture Techniques/methods , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays , Magnetics , Robotics/methods , Spheroids, Cellular/drug effects , Automation, Laboratory/instrumentation , Cell Culture Techniques/instrumentation , Drug Evaluation, Preclinical/instrumentation , Robotics/instrumentation
9.
SLAS Discov ; 23(6): 574-584, 2018 07.
Article in English | MEDLINE | ID: mdl-29673279

ABSTRACT

Traditional high-throughput drug screening in oncology routinely relies on two-dimensional (2D) cell models, which inadequately recapitulate the physiologic context of cancer. Three-dimensional (3D) cell models are thought to better mimic the complexity of in vivo tumors. Numerous methods to culture 3D organoids have been described, but most are nonhomogeneous and expensive, and hence impractical for high-throughput screening (HTS) purposes. Here we describe an HTS-compatible method that enables the consistent production of organoids in standard flat-bottom 384- and 1536-well plates by combining the use of a cell-repellent surface with a bioprinting technology incorporating magnetic force. We validated this homogeneous process by evaluating the effects of well-characterized anticancer agents against four patient-derived pancreatic cancer KRAS mutant-associated primary cells, including cancer-associated fibroblasts. This technology was tested for its compatibility with HTS automation by completing a cytotoxicity pilot screen of ~3300 approved drugs. To highlight the benefits of the 3D format, we performed this pilot screen in parallel in both the 2D and 3D assays. These data indicate that this technique can be readily applied to support large-scale drug screening relying on clinically relevant, ex vivo 3D tumor models directly harvested from patients, an important milestone toward personalized medicine.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Evaluation, Preclinical/methods , Drug Screening Assays, Antitumor/methods , Organoids/drug effects , Pancreatic Neoplasms/drug therapy , Cell Line, Tumor , HT29 Cells , High-Throughput Screening Assays , Humans , Precision Medicine/methods
10.
Assay Drug Dev Technol ; 15(8): 395-406, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29172645

ABSTRACT

Cystic fibrosis (CF), an inherited genetic disease, is caused by mutation of the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) gene, which encodes an ion channel involved in hydration maintenance by anion homeostasis. Ninety percent of CF patients possess one or more copies of the F508del CFTR mutation. This mutation disrupts trafficking of the protein to the plasma membrane and diminishes function of mature CFTR. Identifying small molecule modulators of mutant CFTR activity or biosynthesis may yield new tools for discovering novel CF treatments. One strategy utilizes a 384-well, cell-based fluorescence-quenching assay, which requires extensive wash steps, but reports sensitive changes in fluorescence-quenching kinetic rates. In this study, we describe the methods of adapting the protocol to a homogeneous, miniaturized 1,536-well format and further optimization of this functional F508del CFTR assay. The assay utilizes a cystic fibrosis bronchial epithelial (CFBE41o-) cell line, which was engineered to report CFTR-mediated intracellular flux of iodide by a halide-sensitive yellow fluorescence protein (YFP) reporter. We also describe the limitations of quench rate analysis and the subsequent incorporation of a novel, kinetic data analysis modality to quickly and efficiently find active CFTR modulators. This format yields a Z' value interval of 0.61 ± 0.05. As further evidence of high-throughput screen suitability, we subsequently completed a screening campaign of >645,000 compounds, identifying 2,811 initial hits. After completing secondary and tertiary follow-up assays, we identified 187 potential CFTR modulators, which EC50's < 5 µM. Thus, the assay has integrated the advantages of a phenotypic screen with high-throughput scalability to discover new small-molecule CFTR modulators.


Subject(s)
Bacterial Proteins/analysis , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Drug Evaluation, Preclinical/methods , Luminescent Proteins/analysis , Small Molecule Libraries/analysis , Small Molecule Libraries/pharmacology , Bacterial Proteins/metabolism , Cells, Cultured , Cystic Fibrosis/drug therapy , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/agonists , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , High-Throughput Screening Assays/methods , Humans , Luminescent Proteins/metabolism , Small Molecule Libraries/chemistry
11.
SLAS Discov ; 22(7): 887-896, 2017 08.
Article in English | MEDLINE | ID: mdl-28346094

ABSTRACT

Primary hyperoxaluria is the underlying cause of oxalosis and is a life-threatening autosomal recessive disease, for which treatment may require dialysis or dual liver-kidney transplantation. The most common primary hyperoxaluria type 1 (PH1) is caused by genetic mutations of a liver-specific enzyme alanine:glyoxylate aminotransferase (AGT), which results in the misrouting of AGT from the peroxisomes to the mitochondria. Pharmacoperones are small molecules with the ability to modify misfolded proteins and route them correctly within the cells, which may present an effective strategy to treat AGT misrouting in PH1 disorders. We miniaturized a cell-based high-content assay into 1536-well plate format and screened ~4200 pharmacologically relevant compounds including Food and Drug Administration, European Union, and Japanese-approved drugs. This assay employs CHO cells stably expressing AGT-170, a mutant that predominantly resides in the mitochondria, where we monitor for its relocation to the peroxisomes through automated image acquisition and analysis. The miniaturized 1536-well assay yielded a Z' averaging 0.70 ± 0.07. Three drugs were identified as potential pharmacoperones from this pilot screen, demonstrating the applicability of this assay for large-scale high-throughput screening.


Subject(s)
Hyperoxaluria/drug therapy , Ionophores/pharmacology , Kidney Diseases/drug therapy , Animals , CHO Cells , Cricetulus , Drug Evaluation, Preclinical/methods , Hyperoxaluria/genetics , Hyperoxaluria/metabolism , Hyperoxaluria, Primary/drug therapy , Hyperoxaluria, Primary/genetics , Hyperoxaluria, Primary/metabolism , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Transplantation/methods , Liver/drug effects , Liver/metabolism , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Mutation/genetics , Peroxisomes/drug effects , Peroxisomes/genetics , Peroxisomes/metabolism , Renal Dialysis/methods , Transaminases/genetics , Transaminases/metabolism
12.
Elife ; 52016 07 20.
Article in English | MEDLINE | ID: mdl-27435961

ABSTRACT

Imbalances in endoplasmic reticulum (ER) proteostasis are associated with etiologically-diverse degenerative diseases linked to excessive extracellular protein misfolding and aggregation. Reprogramming of the ER proteostasis environment through genetic activation of the Unfolded Protein Response (UPR)-associated transcription factor ATF6 attenuates secretion and extracellular aggregation of amyloidogenic proteins. Here, we employed a screening approach that included complementary arm-specific UPR reporters and medium-throughput transcriptional profiling to identify non-toxic small molecules that phenocopy the ATF6-mediated reprogramming of the ER proteostasis environment. The ER reprogramming afforded by our molecules requires activation of endogenous ATF6 and occurs independent of global ER stress. Furthermore, our molecules phenocopy the ability of genetic ATF6 activation to selectively reduce secretion and extracellular aggregation of amyloidogenic proteins. These results show that small molecule-dependent ER reprogramming, achieved through preferential activation of the ATF6 transcriptional program, is a promising strategy to ameliorate imbalances in ER function associated with degenerative protein aggregation diseases.


Subject(s)
Activating Transcription Factor 6/biosynthesis , Protein Aggregation, Pathological/prevention & control , Proteostasis/drug effects , Unfolded Protein Response/drug effects , Cell Line , Drug Evaluation, Preclinical/methods , Humans
13.
Assay Drug Dev Technol ; 13(1): 16-24, 2015.
Article in English | MEDLINE | ID: mdl-25710543

ABSTRACT

Primary hyperoxaluria is a severe disease for which the best current therapy is dialysis or organ transplantation. These are risky, inconvenient, and costly procedures. In some patients, pyridoxine treatment can delay the need for these surgical procedures. The underlying cause of particular forms of this disease is the misrouting of a specific enzyme, alanine:glyoxylate aminotransferase (AGT), to the mitochondria instead of the peroxisomes. Pharmacoperones are small molecules that can rescue misfolded proteins and redirect them to their correct location, thereby restoring their function and potentially curing disease. In the present study, we miniaturized a cell-based assay to identify pharmacoperone drugs present in large chemical libraries to selectively correct AGT misrouting. This assay employs AGT-170, a mutant form of AGT that predominantly resides in the mitochondria, which we monitor for its relocation to the peroxisomes through automated image acquisition and analysis. Over the course of a pilot screen of 1,280 test compounds, we achieved an average Z'-factor of 0.72±0.02, demonstrating the suitability of this assay for HTS.


Subject(s)
Biological Assay/methods , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Hyperoxaluria, Primary/drug therapy , Hyperoxaluria, Primary/pathology , Molecular Chaperones/pharmacology , Animals , CHO Cells , Cell Survival/drug effects , Cricetulus , Drug Design , Humans , Molecular Chaperones/chemical synthesis , Molecular Chaperones/classification , Phenotype , Technology, Pharmaceutical/methods
14.
ACS Chem Biol ; 10(4): 925-32, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25602368

ABSTRACT

Platelet-activating factor acetylhydrolases (PAFAHs) 1b2 and 1b3 are poorly characterized serine hydrolases that form a complex with a noncatalytic protein (1b1) to regulate brain development, spermatogenesis, and cancer pathogenesis. Determining physiological substrates and biochemical functions for the PAFAH1b complex would benefit from selective chemical probes that can perturb its activity in living systems. Here, we report a class of tetrahydropyridine reversible inhibitors of PAFAH1b2/3 discovered using a fluorescence polarization-activity-based protein profiling (fluopol-ABPP) screen of the NIH 300,000+ compound library. The most potent of these agents, P11, exhibited IC50 values of ∼40 and 900 nM for PAFAH1b2 and 1b3, respectively. We confirm selective inhibition of PAFAH1b2/3 in cancer cells by P11 using an ABPP protocol adapted for in situ analysis of reversible inhibitors and show that this compound impairs tumor cell survival, supporting a role for PAFAH1b2/3 in cancer.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , 1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , 1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Animals , Brain/drug effects , Brain/metabolism , Cell Line , Cell Line, Tumor/drug effects , Cell Survival/drug effects , Fluorescence Polarization/methods , Humans , Inhibitory Concentration 50 , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Proteomics/methods , Pyridines/chemistry , Pyridines/pharmacology , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
15.
Mol Biochem Parasitol ; 180(2): 127-31, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21939693

ABSTRACT

Recent success in the global reduction campaign against malaria has resulted in the possibility that it may be feasible to drastically reduce or even eradicate malaria even without the introduction of a vaccine. However, while there has been significant effort to design the next generation of antimalarial drugs, one area that is underrepresented in the current antimalarial pharmacopeia is that of transmission blocking drugs directed at late-stage gametocytes. Here we describe the development of a robust and simple assay that is amenable to a high throughput format for the discovery of new antigametocyte drugs.


Subject(s)
Antimalarials/pharmacology , Drug Evaluation, Preclinical/methods , Malaria, Falciparum/physiopathology , Plasmodium falciparum/drug effects , Humans , Malaria, Falciparum/drug therapy , Parasitic Sensitivity Tests , Plasmodium falciparum/growth & development
16.
Proc Natl Acad Sci U S A ; 108(17): 6811-6, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21398589

ABSTRACT

National Institutes of Health (NIH)-sponsored screening centers provide academic researchers with a special opportunity to pursue small-molecule probes for protein targets that are outside the current interest of, or beyond the standard technologies employed by, the pharmaceutical industry. Here, we describe the outcome of an inhibitor screen for one such target, the enzyme protein phosphatase methylesterase-1 (PME-1), which regulates the methylesterification state of protein phosphatase 2A (PP2A) and is implicated in cancer and neurodegeneration. Inhibitors of PME-1 have not yet been described, which we attribute, at least in part, to a dearth of substrate assays compatible with high-throughput screening. We show that PME-1 is assayable by fluorescence polarization-activity-based protein profiling (fluopol-ABPP) and use this platform to screen the 300,000+ member NIH small-molecule library. This screen identified an unusual class of compounds, the aza-ß-lactams (ABLs), as potent (IC(50) values of approximately 10 nM), covalent PME-1 inhibitors. Interestingly, ABLs did not derive from a commercial vendor but rather an academic contribution to the public library. We show using competitive-ABPP that ABLs are exquisitely selective for PME-1 in living cells and mice, where enzyme inactivation leads to substantial reductions in demethylated PP2A. In summary, we have combined advanced synthetic and chemoproteomic methods to discover a class of ABL inhibitors that can be used to selectively perturb PME-1 activity in diverse biological systems. More generally, these results illustrate how public screening centers can serve as hubs to create spontaneous collaborative opportunities between synthetic chemistry and chemical biology labs interested in creating first-in-class pharmacological probes for challenging protein targets.


Subject(s)
Carboxylic Ester Hydrolases/antagonists & inhibitors , Enzyme Inhibitors , Animals , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Mice , Mice, Knockout , National Institutes of Health (U.S.) , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , United States
17.
Mol Pharmacol ; 77(1): 46-57, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19837904

ABSTRACT

The role of neuropeptide Y Y2 receptor (Y2R) in human diseases such as obesity, mood disorders, and alcoholism could be better resolved by the use of small-molecule chemical probes that are substantially different from the currently available Y2R antagonist, N-[(1S)-4-[(aminoiminomethyl)amino]-1-[[[2-(3,5-dioxo-1,2-diphenyl-1,2,4-triazolidin-4-yl)ethyl]amino]carbonyl]butyl]-1-[2-[4-(6,11-dihydro-6-oxo-5H-dibenz[b,e]azepin-11-yl)-1-piperazinyl]-2-oxoethyl]-cyclopentaneacetamide) (BIIE0246). Presented here are five potent, selective, and publicly available Y2R antagonists identified by a high-throughput screening approach. These compounds belong to four chemical scaffolds that are structurally distinct from the peptidomimetic BIIE0246. In functional assays, IC(50) values between 199 and 4400 nM against the Y2R were measured, with no appreciable activity against the related NPY-Y1 receptor (Y1R). Compounds also displaced radiolabeled peptide YY from the Y2R with high affinity (K(i) values between 1.55 and 60 nM) while not displacing the same ligand from the Y1R. In contrast to BIIE0246, Schild analysis with NPY suggests that two of the five compounds behave as competitive antagonists. Profiling against a panel of 40 receptors, ion channels, and transporters found in the central nervous system showed that the five Y2R antagonists demonstrate greater selectivity than BIIE0246. Furthermore, the ability of these antagonists to penetrate the blood-brain barrier makes them better suited for pharmacological studies of Y2R function in both the brain and periphery.


Subject(s)
Drug Evaluation, Preclinical/methods , Heterocyclic Compounds/pharmacokinetics , Receptors, Neuropeptide Y/antagonists & inhibitors , Animals , Arginine/analogs & derivatives , Arginine/pharmacokinetics , Arginine/pharmacology , Benzazepines/pharmacokinetics , Benzazepines/pharmacology , Binding, Competitive , Blood-Brain Barrier/metabolism , Cell Line , Drug Stability , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Heterocyclic Compounds, 3-Ring/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Inhibitory Concentration 50 , Ligands , Mice , Mice, Inbred C57BL , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL