Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 38
Filtrer
1.
Biochem Biophys Res Commun ; 718: 149981, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38735134

RÉSUMÉ

In animal cells, vacuoles are absent, but can be induced by diseases and drugs. While phosphoinositides are critical for membrane trafficking, their role in the formation of these vacuoles remains unclear. The immunosuppressive KRP203/Mocravimod, which antagonizes sphingosine-1-phosphate receptors, has been identified as having novel multimodal activity against phosphoinositide kinases. However, the impact of this novel KRP203 activity is unknown. Here, we show that KRP203 disrupts the spatial organization of phosphoinositides and induces extensive vacuolization in tumor cells and immortalized fibroblasts. The KRP203-induced vacuoles are primarily from endosomes, and augmented by inhibition of PIKFYVE and VPS34. Conversely, overexpression of PTEN decreased KRP203-induced vacuole formation. Furthermore, V-ATPase inhibition completely blunted KRP203-induced vacuolization, pointing to a critical requirement of the endosomal maturation process. Importantly, nearly a half of KRP203-induced vacuoles are significantly decorated with PI4P, a phosphoinositide typically enriched at the plasma membrane and Golgi. These results suggest a model that noncanonical spatial reorganization of phosphoinositides by KRP203 alters the endosomal maturation process, leading to vacuolization. Taken together, this study reveals a previously unrecognized bioactivity of KRP203 as a vacuole-inducing agent and its unique mechanism of phosphoinositide modulation, providing a new insight of phosphoinositide regulation into vacuolization-associated diseases and their molecular pathologies.


Sujet(s)
Endosomes , Phosphohydrolase PTEN , Phosphatidyl inositols , Vacuoles , Vacuoles/métabolisme , Vacuoles/effets des médicaments et des substances chimiques , Endosomes/métabolisme , Endosomes/effets des médicaments et des substances chimiques , Humains , Phosphatidyl inositols/métabolisme , Animaux , Phosphohydrolase PTEN/métabolisme , Phosphohydrolase PTEN/génétique , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases de classe III/métabolisme , Phosphatidylinositol 3-kinases de classe III/génétique , Souris , Morpholines/pharmacologie , Vacuolar Proton-Translocating ATPases/métabolisme , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Vacuolar Proton-Translocating ATPases/génétique , Cytoplasme/métabolisme , Cellules HeLa , Aminopyridines , Composés hétérocycliques 3 noyaux
2.
J Infect Dis ; 228(Suppl 5): S337-S354, 2023 10 03.
Article de Anglais | MEDLINE | ID: mdl-37669225

RÉSUMÉ

The National Center for Advancing Translational Sciences (NCATS) Assay Guidance Manual (AGM) Workshop on 3D Tissue Models for Antiviral Drug Development, held virtually on 7-8 June 2022, provided comprehensive coverage of critical concepts intended to help scientists establish robust, reproducible, and scalable 3D tissue models to study viruses with pandemic potential. This workshop was organized by NCATS, the National Institute of Allergy and Infectious Diseases, and the Bill and Melinda Gates Foundation. During the workshop, scientific experts from academia, industry, and government provided an overview of 3D tissue models' utility and limitations, use of existing 3D tissue models for antiviral drug development, practical advice, best practices, and case studies about the application of available 3D tissue models to infectious disease modeling. This report includes a summary of each workshop session as well as a discussion of perspectives and challenges related to the use of 3D tissues in antiviral drug discovery.


Sujet(s)
Antiviraux , Découverte de médicament , Antiviraux/pharmacologie , Antiviraux/usage thérapeutique , Dosage biologique
3.
Biochem Biophys Res Commun ; 679: 116-121, 2023 Oct 30.
Article de Anglais | MEDLINE | ID: mdl-37683456

RÉSUMÉ

Increased phosphoinositide signaling is commonly associated with cancers. While "one-drug one-target" has been a major drug discovery strategy for cancer therapy, a "one-drug multi-targets" approach for phosphoinositide enzymes has the potential to offer a new therapeutic approach. In this study, we sought a new way to target phosphoinositides metabolism. Using a high-throughput phosphatidylinositol 5-phosphate 4-kinase-alpha (PI5P4Kα) assay, we have identified that the immunosuppressor KRP203/Mocravimod induces a significant perturbation in phosphoinositide metabolism in U87MG glioblastoma cells. Despite high sequence similarity of PI5P4K and PI4K isozymes, in vitro kinase assays showed that KRP203 activates some (e.g., PI5P4Kα, PI4KIIß) while inhibiting other phosphoinositide kinases (e.g., PI5P4Kß, γ, PI4KIIα, class I PI3K-p110α, δ, γ). Furthermore, KRP203 enhances PI3P5K/PIKFYVE's substrate selectivity for phosphatidylinositol (PI) while preserving its selectivity for PI(3)P. At cellular levels, 3 h of KRP203 treatment induces a prominent increase of PI(3)P and moderate increase of PI(5)P, PI(3,5)P2, and PI(3,4,5)P3 levels in U87MG cells. Collectively, the finding of multimodal activity of KRP203 towards multi-phosphoinositide kinases may open a novel basis to modulate cellular processes, potentially leading to more effective treatments for diseases associated with phosphoinositide signaling pathways.

4.
Nat Cancer ; 4(3): 365-381, 2023 03.
Article de Anglais | MEDLINE | ID: mdl-36914816

RÉSUMÉ

Adult liver malignancies, including intrahepatic cholangiocarcinoma and hepatocellular carcinoma, are the second leading cause of cancer-related deaths worldwide. Most individuals are treated with either combination chemotherapy or immunotherapy, respectively, without specific biomarkers for selection. Here using high-throughput screens, proteomics and in vitro resistance models, we identify the small molecule YC-1 as selectively active against a defined subset of cell lines derived from both liver cancer types. We demonstrate that selectivity is determined by expression of the liver-resident cytosolic sulfotransferase enzyme SULT1A1, which sulfonates YC-1. Sulfonation stimulates covalent binding of YC-1 to lysine residues in protein targets, enriching for RNA-binding factors. Computational analysis defined a wider group of structurally related SULT1A1-activated small molecules with distinct target profiles, which together constitute an untapped small-molecule class. These studies provide a foundation for preclinical development of these agents and point to the broader potential of exploiting SULT1A1 activity for selective targeting strategies.


Sujet(s)
Agents alcoylants , Tumeurs du foie , Humains , Sulfotransferases , Tumeurs du foie/traitement médicamenteux , Arylsulfotransferase
5.
J Infect Dis ; 224(Supplement_1): S1-S21, 2021 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-34111271

RÉSUMÉ

The NIH Virtual SARS-CoV-2 Antiviral Summit, held on 6 November 2020, was organized to provide an overview on the status and challenges in developing antiviral therapeutics for coronavirus disease 2019 (COVID-19), including combinations of antivirals. Scientific experts from the public and private sectors convened virtually during a live videocast to discuss severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) targets for drug discovery as well as the preclinical tools needed to develop and evaluate effective small-molecule antivirals. The goals of the Summit were to review the current state of the science, identify unmet research needs, share insights and lessons learned from treating other infectious diseases, identify opportunities for public-private partnerships, and assist the research community in designing and developing antiviral therapeutics. This report includes an overview of therapeutic approaches, individual panel summaries, and a summary of the discussions and perspectives on the challenges ahead for antiviral development.


Sujet(s)
Antiviraux/usage thérapeutique , Traitements médicamenteux de la COVID-19 , SARS-CoV-2/effets des médicaments et des substances chimiques , Antiviraux/pharmacologie , COVID-19/virologie , Développement de médicament , Humains , National Institutes of Health (USA) , Peptide hydrolases/métabolisme , Inhibiteurs de protéases/pharmacologie , Inhibiteurs de protéases/usage thérapeutique , États-Unis , Réplication virale/effets des médicaments et des substances chimiques
6.
J Med Chem ; 64(8): 4913-4946, 2021 04 22.
Article de Anglais | MEDLINE | ID: mdl-33822623

RÉSUMÉ

Neomorphic mutations in isocitrate dehydrogenase 1 (IDH1) are oncogenic for a number of malignancies, primarily low-grade gliomas and acute myeloid leukemia. We report a medicinal chemistry campaign around a 7,7-dimethyl-7,8-dihydro-2H-1λ2-quinoline-2,5(6H)-dione screening hit against the R132H and R132C mutant forms of isocitrate dehydrogenase (IDH1). Systematic SAR efforts produced a series of potent pyrid-2-one mIDH1 inhibitors, including the atropisomer (+)-119 (NCATS-SM5637, NSC 791985). In an engineered mIDH1-U87-xenograft mouse model, after a single oral dose of 30 mg/kg, 16 h post dose, between 16 and 48 h, (+)-119 showed higher tumoral concentrations that corresponded to lower 2-HG concentrations, when compared with the approved drug AG-120 (ivosidenib).


Sujet(s)
Antienzymes/composition chimique , Isocitrate dehydrogenases/antagonistes et inhibiteurs , Pyridones/composition chimique , Animaux , Encéphale/métabolisme , Lignée cellulaire tumorale , Évaluation préclinique de médicament , Antienzymes/métabolisme , Antienzymes/usage thérapeutique , Femelle , Glycine/analogues et dérivés , Glycine/usage thérapeutique , Période , Humains , Isocitrate dehydrogenases/génétique , Isocitrate dehydrogenases/métabolisme , Souris , Souris nude , Microsomes du foie/métabolisme , Mutagenèse dirigée , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Pyridines/usage thérapeutique , Pyridones/métabolisme , Pyridones/usage thérapeutique , Rats , Relation structure-activité , Tests d'activité antitumorale sur modèle de xénogreffe
7.
ACS Med Chem Lett ; 11(3): 346-352, 2020 Mar 12.
Article de Anglais | MEDLINE | ID: mdl-32184968

RÉSUMÉ

Phosphatidylinositol 5-phosphate 4-kinases (PI5P4Ks) are important molecular players in a variety of diseases, such as cancer. Currently available PI5P4K inhibitors are reversible small molecules, which may lack selectivity and sufficient cellular on-target activity. In this study, we present a new class of covalent pan-PI5P4K inhibitors with potent biochemical and cellular activity. Our designs are based on THZ-P1-2, a covalent PI5P4K inhibitor previously developed in our lab. Here, we report further structure-guided optimization and structure-activity relationship (SAR) study of this scaffold, resulting in compound 30, which retained biochemical and cellular potency, while demonstrating a significantly improved selectivity profile. Furthermore, we confirm that the inhibitors show efficient binding affinity in the context of HEK 293T cells using isothermal CETSA methods. Taken together, compound 30 represents a highly selective pan-PI5P4K covalent lead molecule.

8.
Clin Cancer Res ; 25(14): 4552-4566, 2019 07 15.
Article de Anglais | MEDLINE | ID: mdl-30979745

RÉSUMÉ

PURPOSE: Ewing sarcoma is an aggressive solid tumor malignancy of childhood. Although current treatment regimens cure approximately 70% of patients with localized disease, they are ineffective for most patients with metastases or relapse. New treatment combinations are necessary for these patients. EXPERIMENTAL DESIGN: Ewing sarcoma cells are dependent on focal adhesion kinase (FAK) for growth. To identify candidate treatment combinations for Ewing sarcoma, we performed a small-molecule library screen to identify compounds synergistic with FAK inhibitors in impairing Ewing cell growth. The activity of a top-scoring class of compounds was then validated across multiple Ewing cell lines in vitro and in multiple xenograft models of Ewing sarcoma. RESULTS: Numerous Aurora kinase inhibitors scored as synergistic with FAK inhibition in this screen. We found that Aurora kinase B inhibitors were synergistic across a larger range of concentrations than Aurora kinase A inhibitors when combined with FAK inhibitors in multiple Ewing cell lines. The combination of AZD-1152, an Aurora kinase B-selective inhibitor, and PF-562271 or VS-4718, FAK-selective inhibitors, induced apoptosis in Ewing sarcoma cells at concentrations that had minimal effects on survival when cells were treated with either drug alone. We also found that the combination significantly impaired tumor progression in multiple xenograft models of Ewing sarcoma. CONCLUSIONS: FAK and Aurora kinase B inhibitors synergistically impair Ewing sarcoma cell viability and significantly inhibit tumor progression. This study provides preclinical support for the consideration of a clinical trial testing the safety and efficacy of this combination for patients with Ewing sarcoma.


Sujet(s)
Aurora kinase B/antagonistes et inhibiteurs , Tumeurs osseuses/traitement médicamenteux , Synergie des médicaments , Focal adhesion kinase 1/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/pharmacologie , Sarcome d'Ewing/traitement médicamenteux , Bibliothèques de petites molécules/pharmacologie , Aminopyridines/pharmacologie , Animaux , Apoptose , Tumeurs osseuses/enzymologie , Tumeurs osseuses/anatomopathologie , Prolifération cellulaire , Association de médicaments , Femelle , Tests de criblage à haut débit , Humains , Indoles/pharmacologie , Souris , Souris nude , Organophosphates/pharmacologie , Quinazolines/pharmacologie , Sarcome d'Ewing/enzymologie , Sarcome d'Ewing/anatomopathologie , Sulfonamides/pharmacologie , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe , Danio zébré
9.
Clin Cancer Res ; 25(4): 1343-1357, 2019 02 15.
Article de Anglais | MEDLINE | ID: mdl-30397176

RÉSUMÉ

PURPOSE: Novel targeted therapeutics have transformed the care of subsets of patients with cancer. In pediatric malignancies, however, with simple tumor genomes and infrequent targetable mutations, there have been few new FDA-approved targeted drugs. The cyclin-dependent kinase (CDK)4/6 pathway recently emerged as a dependency in Ewing sarcoma. Given the heightened efficacy of this class with targeted drug combinations in other cancers, as well as the propensity of resistance to emerge with single agents, we aimed to identify genes mediating resistance to CDK4/6 inhibitors and biologically relevant combinations for use with CDK4/6 inhibitors in Ewing. EXPERIMENTAL DESIGN: We performed a genome-scale open reading frame (ORF) screen in 2 Ewing cell lines sensitive to CDK4/6 inhibitors to identify genes conferring resistance. Concurrently, we established resistance to a CDK4/6 inhibitor in a Ewing cell line. RESULTS: The ORF screen revealed IGF1R as a gene whose overexpression promoted drug escape. We also found elevated levels of phospho-IGF1R in our resistant Ewing cell line, supporting the relevance of IGF1R signaling to acquired resistance. In a small-molecule screen, an IGF1R inhibitor scored as synergistic with CDK4/6 inhibitor treatment. The combination of CDK4/6 inhibitors and IGF1R inhibitors was synergistic in vitro and active in mouse models. Mechanistically, this combination more profoundly repressed cell cycle and PI3K/mTOR signaling than either single drug perturbation. CONCLUSIONS: Taken together, these results suggest that IGF1R inhibitors activation is an escape mechanism to CDK4/6 inhibitors in Ewing sarcoma and that dual targeting of CDK4/6 inhibitors and IGF1R inhibitors provides a candidate synergistic combination for clinical application in this disease.


Sujet(s)
Kinase-4 cycline-dépendante/génétique , Kinase-6 cycline-dépendante/génétique , Récepteur IGF de type 1/génétique , Sarcome d'Ewing/traitement médicamenteux , Animaux , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Lignée cellulaire tumorale , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Résistance aux médicaments antinéoplasiques/génétique , Synergie des médicaments , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Souris , Inhibiteurs de protéines kinases/pharmacologie , Récepteur IGF de type 1/antagonistes et inhibiteurs , Sarcome d'Ewing/génétique , Sarcome d'Ewing/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Bibliothèques de petites molécules/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
10.
Cancer Cell ; 34(6): 922-938.e7, 2018 12 10.
Article de Anglais | MEDLINE | ID: mdl-30537514

RÉSUMÉ

Drug resistance represents a major challenge to achieving durable responses to cancer therapeutics. Resistance mechanisms to epigenetically targeted drugs remain largely unexplored. We used bromodomain and extra-terminal domain (BET) inhibition in neuroblastoma as a prototype to model resistance to chromatin modulatory therapeutics. Genome-scale, pooled lentiviral open reading frame (ORF) and CRISPR knockout rescue screens nominated the phosphatidylinositol 3-kinase (PI3K) pathway as promoting resistance to BET inhibition. Transcriptomic and chromatin profiling of resistant cells revealed that global enhancer remodeling is associated with upregulation of receptor tyrosine kinases (RTKs), activation of PI3K signaling, and vulnerability to RTK/PI3K inhibition. Large-scale combinatorial screening with BET inhibitors identified PI3K inhibitors among the most synergistic upfront combinations. These studies provide a roadmap to elucidate resistance to epigenetic-targeted therapeutics and inform efficacious combination therapies.


Sujet(s)
Azépines/pharmacologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Indazoles/pharmacologie , Thérapie moléculaire ciblée/méthodes , Neuroblastome/traitement médicamenteux , Sulfonamides/pharmacologie , Triazoles/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe , Animaux , Lignée cellulaire tumorale , Survie sans rechute , Épigenèse génétique/effets des médicaments et des substances chimiques , Femelle , Humains , Souris nude , Neuroblastome/génétique , Neuroblastome/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Inhibiteurs des phosphoinositide-3 kinases , Protéines/antagonistes et inhibiteurs , Protéines/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
11.
Sci Rep ; 7(1): 12758, 2017 10 06.
Article de Anglais | MEDLINE | ID: mdl-28986582

RÉSUMÉ

Isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) are key metabolic enzymes that are mutated in a variety of cancers to confer a gain-of-function activity resulting in the accumulation of an oncometabolite, D-2-hydroxyglutarate (2-HG). Accumulation of 2-HG can result in epigenetic dysregulation and a block in cellular differentiation, suggesting these mutations play a role in neoplasia. Based on its potential as a cancer target, a number of small molecule inhibitors have been developed to specifically inhibit mutant forms of IDH (mIDH1 and mIDH2). We present a comprehensive suite of in vitro preclinical drug development assays that can be used as a tool-box to identify lead compounds for mIDH drug discovery programs, as well as what we believe is the most comprehensive publically available dataset on the top mIDH inhibitors. This involved biochemical, cell-based, and tier-one ADME techniques.


Sujet(s)
Découverte de médicament , Évaluation préclinique de médicament/méthodes , Antienzymes/pharmacologie , Isocitrate dehydrogenases/antagonistes et inhibiteurs , Isocitrate dehydrogenases/génétique , Mutation/génétique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Antienzymes/composition chimique , Antienzymes/pharmacocinétique , Stabilité enzymatique , Fluorescence , Glutarates/métabolisme , Tests de criblage à haut débit , Histone/métabolisme , Humains , Isocitrate dehydrogenases/métabolisme , Méthylation , Modèles biologiques , Monocytes/cytologie , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/métabolisme , Cellules THP-1
12.
Clin Cancer Res ; 23(23): 7301-7311, 2017 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-28899971

RÉSUMÉ

Purpose: Although many cancers are showing remarkable responses to targeted therapies, pediatric sarcomas, including Ewing sarcoma, remain recalcitrant. To broaden the therapeutic landscape, we explored the in vitro response of Ewing sarcoma cell lines against a large collection of investigational and approved drugs to identify candidate combinations.Experimental Design: Drugs displaying activity as single agents were evaluated in combinatorial (matrix) format to identify highly active, synergistic drug combinations, and combinations were subsequently validated in multiple cell lines using various agents from each class. Comprehensive metabolomic and proteomic profiling was performed to better understand the mechanism underlying the synergy. Xenograft experiments were performed to determine efficacy and in vivo mechanism.Results: Several promising candidates emerged, including the combination of small-molecule PARP and nicotinamide phosphoribosyltransferase (NAMPT) inhibitors, a rational combination as NAMPTis block the rate-limiting enzyme in the production of nicotinamide adenine dinucleotide (NAD+), a necessary substrate of PARP. Mechanistic drivers of the synergistic cell killing phenotype of these combined drugs included depletion of NMN and NAD+, diminished PAR activity, increased DNA damage, and apoptosis. Combination PARPis and NAMPTis in vivo resulted in tumor regression, delayed disease progression, and increased survival.Conclusions: These studies highlight the potential of these drugs as a possible therapeutic option in treating patients with Ewing sarcoma. Clin Cancer Res; 23(23); 7301-11. ©2017 AACR.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Cytokines/antagonistes et inhibiteurs , Antienzymes/pharmacologie , Nicotinamide phosphoribosyltransferase/antagonistes et inhibiteurs , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Sarcome d'Ewing/traitement médicamenteux , Tests d'activité antitumorale sur modèle de xénogreffe , Animaux , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Cytokines/métabolisme , Tests de criblage d'agents antitumoraux/méthodes , Synergie des médicaments , Antienzymes/administration et posologie , Femelle , Humains , Estimation de Kaplan-Meier , Souris SCID , Nicotinamide phosphoribosyltransferase/métabolisme , Inhibiteurs de poly(ADP-ribose) polymérases/administration et posologie , Sarcome d'Ewing/métabolisme , Sarcome d'Ewing/anatomopathologie , Charge tumorale/effets des médicaments et des substances chimiques
13.
Sci Rep ; 6: 37741, 2016 11 24.
Article de Anglais | MEDLINE | ID: mdl-27883049

RÉSUMÉ

Quality control (QC) metrics are critical in high throughput screening (HTS) platforms to ensure reliability and confidence in assay data and downstream analyses. Most reported HTS QC metrics are designed for plate level or single well level analysis. With the advent of high throughput combination screening there is a need for QC metrics that quantify the quality of combination response matrices. We introduce a predictive, interpretable, matrix-level QC metric, mQC, based on a mix of data-derived and heuristic features. mQC accurately reproduces the expert assessment of combination response quality and correctly identifies unreliable response matrices that can lead to erroneous or misleading characterization of synergy. When combined with the plate-level QC metric, Z', mQC provides a more appropriate determination of the quality of a drug combination screen. Retrospective analysis on a number of completed combination screens further shows that mQC is able to identify problematic screens whereas plate-level QC was not able to. In conclusion, our data indicates that mQC is a reliable QC filter that can be used to identify problematic drug combinations matrices and prevent further analysis on erroneously active combinations as well as for troubleshooting failed screens. The R source code of mQC is available at http://matrix.ncats.nih.gov/mQC.


Sujet(s)
Préparations pharmaceutiques/administration et posologie , Préparations pharmaceutiques/composition chimique , Association médicamenteuse , Tests de criblage à haut débit/méthodes , Humains , Contrôle de qualité , Reproductibilité des résultats , Études rétrospectives
14.
J Biol Chem ; 291(47): 24628-24640, 2016 Nov 18.
Article de Anglais | MEDLINE | ID: mdl-27681596

RÉSUMÉ

Deubiquitinases are important components of the protein degradation regulatory network. We report the discovery of ML364, a small molecule inhibitor of the deubiquitinase USP2 and its use to interrogate the biology of USP2 and its putative substrate cyclin D1. ML364 has an IC50 of 1.1 µm in a biochemical assay using an internally quenched fluorescent di-ubiquitin substrate. Direct binding of ML364 to USP2 was demonstrated using microscale thermophoresis. ML364 induced an increase in cellular cyclin D1 degradation and caused cell cycle arrest as shown in Western blottings and flow cytometry assays utilizing both Mino and HCT116 cancer cell lines. ML364, and not the inactive analog 2, was antiproliferative in cancer cell lines. Consistent with the role of cyclin D1 in DNA damage response, ML364 also caused a decrease in homologous recombination-mediated DNA repair. These effects by a small molecule inhibitor support a key role for USP2 as a regulator of cell cycle, DNA repair, and tumor cell growth.


Sujet(s)
Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Tumeurs colorectales/métabolisme , Cycline D1/métabolisme , Endopeptidases/métabolisme , Lymphome à cellules du manteau/traitement médicamenteux , Protéines tumorales/antagonistes et inhibiteurs , Protéines tumorales/métabolisme , Inhibiteurs de protéases/pharmacologie , Protéolyse/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/génétique , Lignée cellulaire tumorale , Tumeurs colorectales/génétique , Cycline D1/génétique , Altération de l'ADN , Réparation de l'ADN , Endopeptidases/génétique , Humains , Lymphome à cellules du manteau/génétique , Lymphome à cellules du manteau/métabolisme , Protéines tumorales/génétique , Inhibiteurs de protéases/composition chimique , Ubiquitin thiolesterase
15.
Antimicrob Agents Chemother ; 60(10): 6023-33, 2016 10.
Article de Anglais | MEDLINE | ID: mdl-27458230

RÉSUMÉ

Plasmodium falciparum, the deadliest species of malaria parasites, is dependent on glycolysis for the generation of ATP during the pathogenic red blood cell stage. Hexokinase (HK) catalyzes the first step in glycolysis, transferring the γ-phosphoryl group of ATP to glucose to yield glucose-6-phosphate. Here, we describe the validation of a high-throughput assay for screening small-molecule collections to identify inhibitors of the P. falciparum HK (PfHK). The assay, which employed an ADP-Glo reporter system in a 1,536-well-plate format, was robust with a signal-to-background ratio of 3.4 ± 1.2, a coefficient of variation of 6.8% ± 2.9%, and a Z'-factor of 0.75 ± 0.08. Using this assay, we screened 57,654 molecules from multiple small-molecule collections. Confirmed hits were resolved into four clusters on the basis of structural relatedness. Multiple singleton hits were also identified. The most potent inhibitors had 50% inhibitory concentrations as low as ∼1 µM, and several were found to have low-micromolar 50% effective concentrations against asexual intraerythrocytic-stage P. falciparum parasites. These molecules additionally demonstrated limited toxicity against a panel of mammalian cells. The identification of PfHK inhibitors with antiparasitic activity using this validated screening assay is encouraging, as it justifies additional HTS campaigns with more structurally amenable libraries for the identification of potential leads for future therapeutic development.


Sujet(s)
Antipaludiques/pharmacologie , Antienzymes/pharmacologie , Hexokinase/antagonistes et inhibiteurs , Tests de criblage à haut débit , Plasmodium falciparum/effets des médicaments et des substances chimiques , Protéines de protozoaire/antagonistes et inhibiteurs , Bibliothèques de petites molécules/pharmacologie , ADP/métabolisme , Adénosine triphosphate/antagonistes et inhibiteurs , Adénosine triphosphate/biosynthèse , Antipaludiques/composition chimique , Survie cellulaire/effets des médicaments et des substances chimiques , Antienzymes/composition chimique , Érythrocytes/effets des médicaments et des substances chimiques , Érythrocytes/parasitologie , Expression des gènes , Gènes rapporteurs , Glycolyse/effets des médicaments et des substances chimiques , Cellules HEK293 , Cellules HeLa , Hexokinase/génétique , Hexokinase/métabolisme , Humains , Luciferases/génétique , Luciferases/métabolisme , Plasmodium falciparum/enzymologie , Plasmodium falciparum/croissance et développement , Protéines de protozoaire/génétique , Protéines de protozoaire/métabolisme , Rapport signal-bruit , Bibliothèques de petites molécules/composition chimique , Relation structure-activité
16.
Cell ; 165(7): 1698-1707, 2016 Jun 16.
Article de Anglais | MEDLINE | ID: mdl-27238019

RÉSUMÉ

Recent advances in single-particle cryoelecton microscopy (cryo-EM) are enabling generation of numerous near-atomic resolution structures for well-ordered protein complexes with sizes ≥ ∼200 kDa. Whether cryo-EM methods are equally useful for high-resolution structural analysis of smaller, dynamic protein complexes such as those involved in cellular metabolism remains an important question. Here, we present 3.8 Å resolution cryo-EM structures of the cancer target isocitrate dehydrogenase (93 kDa) and identify the nature of conformational changes induced by binding of the allosteric small-molecule inhibitor ML309. We also report 2.8-Å- and 1.8-Å-resolution structures of lactate dehydrogenase (145 kDa) and glutamate dehydrogenase (334 kDa), respectively. With these results, two perceived barriers in single-particle cryo-EM are overcome: (1) crossing 2 Å resolution and (2) obtaining structures of proteins with sizes < 100 kDa, demonstrating that cryo-EM can be used to investigate a broad spectrum of drug-target interactions and dynamic conformational states.


Sujet(s)
Découverte de médicament , Glutamate dehydrogenase/ultrastructure , Isocitrate dehydrogenases/ultrastructure , L-Lactate dehydrogenase/ultrastructure , Aminoquinoléines/composition chimique , Aminoquinoléines/pharmacologie , Animaux , Bovins , Poulets , Cryomicroscopie électronique , Cristallographie aux rayons X , Glutamate dehydrogenase/antagonistes et inhibiteurs , Glutamate dehydrogenase/composition chimique , Humains , Isocitrate dehydrogenases/antagonistes et inhibiteurs , Isocitrate dehydrogenases/composition chimique , L-Lactate dehydrogenase/antagonistes et inhibiteurs , L-Lactate dehydrogenase/composition chimique , Modèles moléculaires , Conformation des protéines , Sulfonamides/composition chimique , Sulfonamides/pharmacologie
17.
Assay Drug Dev Technol ; 14(3): 175-9, 2016 Apr.
Article de Anglais | MEDLINE | ID: mdl-27078679

RÉSUMÉ

Fluorescence is utilized as the output for a range of assay formats used in high-throughput screening (HTS). Interference with these assays from the compounds in libraries utilized in HTS is a well-recognized phenomenon, particularly for assays relying on UV excitation such as for direct detection of the oxidoreductase cofactors NADH or NADPH. In this study, we discuss these interference challenges and highlight the specific case of the diaphorase/resazurin system that can be coupled to enzymes utilizing NADH or NADPH. We review the utilization of this assay system in the literature and argue that the diaphorase/resazurin system is underutilized in assay development. It is the authors' hope that this Perspective and the accompanying Technical Brief in this issue will stimulate interest in a robust and sensitive coupling system to avoid assay fluorescence interference.


Sujet(s)
Artéfacts , NADPH dehydrogenase/métabolisme , Oxazines/métabolisme , Xanthènes/métabolisme , Fluorescence , NAD/métabolisme , NADP/métabolisme , NADPH dehydrogenase/composition chimique , Oxazines/composition chimique , Spectrométrie de fluorescence , Xanthènes/composition chimique
18.
Assay Drug Dev Technol ; 14(3): 207-12, 2016 Apr.
Article de Anglais | MEDLINE | ID: mdl-27078681

RÉSUMÉ

Dehydrogenases are an important target for the development of cancer therapeutics. Dehydrogenases either produce or consume NAD(P)H, which is fluorescent but at a wavelength where many compounds found in chemical libraries are also fluorescent. By coupling dehydrogenases to diaphorase, which utilizes NAD(P)H to produce the fluorescent molecule resorufin from resazurin, the assay can be red-shifted into a spectral region that reduces interference from compound libraries. Dehydrogenases that produce NAD(P)H, such as isocitrate dehydrogenase 1 (IDH1), can be read in kinetic mode. Dehydrogenases that consume NAD(P)H, such as mutant IDH1 R132H, can be read in endpoint mode. Here, we report protocols for robust and miniaturized 1,536-well assays for WT IDH1 and IDH1 R132H coupled to diaphorase, and the counterassays used to further detect compound interference with the coupling reagents. This coupling technique is applicable to dehydrogenases that either produce or consume NAD(P)H, and the examples provided here can act as guidelines for the development of high-throughput screens against this enzyme class.


Sujet(s)
Dosages enzymatiques/méthodes , NADPH dehydrogenase/métabolisme , Clostridium kluyveri/enzymologie , Couleur , NAD/métabolisme , NADP/métabolisme
19.
Methods Mol Biol ; 1376: 1-9, 2016.
Article de Anglais | MEDLINE | ID: mdl-26552670

RÉSUMÉ

Lipid kinases are important regulators of a variety of cellular processes and their dysregulation causes diseases such as cancer and metabolic diseases. Distinct lipid kinases regulate the seven different phosphorylated forms of phosphatidylinositol (PtdIns). Some lipid kinases utilize long-chain lipid substrates that have limited solubility in aqueous solutions, which can lead to difficulties in developing a robust and miniaturizable biochemical assay. The ability to prepare the lipid substrate and develop assays to identify modulators of lipid kinases is important and is the focus of this methods chapter. Herein, we describe a method to prepare a DMSO-based lipid mixture that enables the 1536-well screening of the lipid kinase phosphatidylinositol-5-phosphate 4-kinase α (PI5P4Kα) utilizing the D-myo-di16-PtIns(5)P substrate in quantitative high-throughput screening (qHTS) format using the ADP-Glo™ technology to couple the production of ADP to a bioluminescent readout.


Sujet(s)
Tests de criblage à haut débit/méthodes , Mesures de luminescence/méthodes , Phosphotransferases (Alcohol Group Acceptor)/métabolisme , Adénosine triphosphate , Humains , Phosphatidyl inositols/métabolisme , Phosphorylation , Spécificité du substrat
20.
Methods Mol Biol ; 1360: 47-58, 2016.
Article de Anglais | MEDLINE | ID: mdl-26501901

RÉSUMÉ

Assays in which the detection of a biological phenomenon is coupled to the production of bioluminescence by luciferase have gained widespread use. As firefly luciferases (FLuc) and kinases share a common substrate (ATP), coupling of a kinase to FLuc allows for the amount of ATP remaining following a kinase reaction to be assessed by quantitating the amount of luminescence produced. Alternatively, the amount of ADP produced by the kinase reaction can be coupled to FLuc through a two-step process. This chapter describes the bioluminescent assays that were developed for three classes of kinases (lipid, protein, and metabolic kinases) and miniaturized to 1536-well format, enabling their use for quantitative high-throughput (qHTS) of small-molecule libraries.


Sujet(s)
Glucokinase/analyse , Tests de criblage à haut débit/méthodes , Mesures de luminescence/méthodes , Phosphotransferases (Alcohol Group Acceptor)/analyse , Protéines proto-oncogènes c-yes/analyse , Protéines adaptatrices de la transduction du signal/métabolisme , ADP/analyse , Adénosine triphosphate/analyse , Glucokinase/métabolisme , Tests de criblage à haut débit/instrumentation , Humains , Indicateurs et réactifs , Luciférases des lucioles/métabolisme , Mesures de luminescence/instrumentation , Liaison aux protéines , Spécificité du substrat
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...