Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 51
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.
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.

3.
Arterioscler Thromb Vasc Biol ; 43(10): 1808-1817, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37345522

RÉSUMÉ

BACKGROUND: Heparin-induced thrombocytopenia (HIT) is a major concern for all individuals that undergo cardiac bypass surgeries or require prolonged heparin exposure. HIT is a life- and limb-threatening adverse drug reaction with an immune response following the formation of ultra-large immune complexes that drive platelet activation through the receptor FcγRIIA. Thrombotic events remain high following the standard of care treatment with anticoagulants, while increasing risk of bleeding complications. This study sought to investigate a novel approach to treatment of HIT. Recent reports demonstrate increased procoagulant activity in HIT; however, these reports required analysis ex vivo, and relevance in vivo remains unclear. METHODS: Using human and mouse model systems, we investigated the cooperativity of PARs (protease-activated receptors) and FcγRIIA in HIT. We challenged humanized FcγRIIA transgenic mice with or without endogenous mouse Par4 (denoted as IIA-Par4+/+ or IIA-Par4-/-, respectively) with a well-established model IgG immune complex (anti [α]-CD9). Furthermore, we assessed the procoagulant phenotype and efficacy to treat HIT utilizing inhibitor of 12-LOX (12[S]-lipoxygenase), VLX-1005, previously reported to decrease platelet activation downstream of FcγRIIA and PAR4, using the triple allele HIT mouse model. RESULTS: IIA-Par4+/+ mice given αCD9 were severely thrombocytopenic, with extensive platelet-fibrin deposition in the lung. In contrast, IIA-Par4-/- mice had negligible thrombocytopenia or pulmonary platelet-fibrin thrombi. We observed that pharmacological inhibition of 12-LOX resulted in a significant reduction in both platelet procoagulant phenotype ex vivo, and thrombocytopenia and thrombosis in our humanized mouse model of HIT in vivo. CONCLUSIONS: These data demonstrate for the first time the need for dual platelet receptor (PAR and FcγRIIA) stimulation for fibrin formation in HIT in vivo. These results extend our understanding of HIT pathophysiology and provide a scientific rationale for targeting the procoagulant phenotype as a possible therapeutic strategy in HIT.


Sujet(s)
Thrombopénie , Humains , Souris , Animaux , Thrombopénie/induit chimiquement , Héparine/effets indésirables , Plaquettes , Anticoagulants/effets indésirables , Souris transgéniques , Phénotype , Fibrine/génétique , Facteur-4 plaquettaire/génétique
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.
Sci Transl Med ; 13(607)2021 08 18.
Article de Anglais | MEDLINE | ID: mdl-34408078

RÉSUMÉ

Dystonias are a group of chronic movement-disabling disorders for which highly effective oral medications or disease-modifying therapies are lacking. The most effective treatments require invasive procedures such as deep brain stimulation. In this study, we used a high-throughput assay based on a monogenic form of dystonia, DYT1 (DYT-TOR1A), to screen a library of compounds approved for use in humans, the NCATS Pharmaceutical Collection (NPC; 2816 compounds), and identify drugs able to correct mislocalization of the disease-causing protein variant, ∆E302/3 hTorsinA. The HIV protease inhibitor, ritonavir, was among 18 compounds found to normalize hTorsinA mislocalization. Using a DYT1 knock-in mouse model to test efficacy on brain pathologies, we found that ritonavir restored multiple brain abnormalities across development. Ritonavir acutely corrected striatal cholinergic interneuron physiology in the mature brain and yielded sustained correction of diffusion tensor magnetic resonance imaging signals when delivered during a discrete early developmental window. Mechanistically, we found that, across the family of HIV protease inhibitors, efficacy correlated with integrated stress response activation. These preclinical results identify ritonavir as a drug candidate for dystonia with disease-modifying potential.


Sujet(s)
Dystonie , Inhibiteurs de protéase du VIH , Animaux , Encéphale/imagerie diagnostique , Dystonie/traitement médicamenteux , Souris , Chaperons moléculaires , Phénotype , Ritonavir
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.
Mol Cancer Ther ; 19(7): 1520-1529, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32371575

RÉSUMÉ

Despite a growing body of knowledge about the genomic landscape of Ewing sarcoma, translation of basic discoveries into targeted therapies and significant clinical gains has remained elusive. Recent insights have revealed that the oncogenic transcription factor EWS-FLI1 can impact Ewing sarcoma cellular metabolism, regulating expression of 3-phosphoglycerate dehydrogenase (PHGDH), the first enzyme in de novo serine synthesis. Here, we have examined the importance of serine metabolism in Ewing sarcoma tumorigenesis and evaluated the therapeutic potential of targeting serine metabolism in preclinical models of Ewing sarcoma. We show that PHGDH knockdown resulted in decreased Ewing sarcoma cell proliferation, especially under serine limitation, and significantly inhibited xenograft tumorigenesis in preclinical orthotopic models of Ewing sarcoma. In addition, the PHGDH inhibitor NCT-503 caused a dose-dependent decrease in cellular proliferation. Moreover, we report a novel drug combination in which nicotinamide phosphoribosyltransferase (NAMPT) inhibition, which blocks production of the PHGDH substrate NAD+, synergized with NCT-503 to abolish Ewing sarcoma cell proliferation and tumor growth. Furthermore, we show that serine deprivation inhibited Ewing sarcoma cell proliferation and tumorigenesis, indicating that Ewing sarcoma cells depend on exogenous serine in addition to de novo serine synthesis. Our findings suggest that serine metabolism is critical for Ewing sarcoma tumorigenesis, and that targeting metabolic dependencies should be further investigated as a potential therapeutic strategy for Ewing sarcoma. In addition, the combination strategy presented herein may have broader clinical applications in other PHGDH-overexpressing cancers as well.


Sujet(s)
Tumeurs osseuses/anatomopathologie , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Protéines de fusion oncogènes/métabolisme , Phosphoglycerate dehydrogenase/métabolisme , Protéine proto-oncogène c-fli-1/métabolisme , Protéine EWS de liaison à l'ARN/métabolisme , Sarcome d'Ewing/anatomopathologie , Sérine/métabolisme , Animaux , Apoptose , Tumeurs osseuses/génétique , Tumeurs osseuses/métabolisme , Femelle , Humains , Souris , Souris SCID , Protéines de fusion oncogènes/génétique , Protéine proto-oncogène c-fli-1/génétique , Protéine EWS de liaison à l'ARN/génétique , Sarcome d'Ewing/génétique , Sarcome d'Ewing/métabolisme , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
8.
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.

9.
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
10.
J Comput Aided Mol Des ; 33(4): 405-417, 2019 04.
Article de Anglais | MEDLINE | ID: mdl-30806949

RÉSUMÉ

Classic Galactosemia is a potentially lethal autosomal recessive metabolic disorder caused by deficient galactose-1-phosphate uridyltransferase (GALT) that results in the buildup of galactose-1-phosphate (gal-1-p) in cells. Galactokinase (GALK1) is the enzyme responsible for converting galactose into gal-1-p. A pharmacological inhibitor of GALK1 is hypothesized to be therapeutic strategy for treating galactosemia by reducing production of gal-1-p. In this study, we report the discovery of novel series of GALK1 inhibitors by structure-based virtual screening (VS). Followed by an extensive structural modeling and binding mode analysis of the active compounds identified from quantitative high-throughput screen (qHTS), we developed an efficient pharmacophore-based VS approach and applied for a large-scale in silico database screening. Out of 230,000 compounds virtually screened, 350 compounds were cherry-picked based on multi-factor prioritization procedure, and 75 representing a diversity of chemotypes exhibited inhibitory activity in GALK1 biochemical assay. Furthermore, a phenylsulfonamide series with excellent in vitro ADME properties was selected for downstream characterization and demonstrated its ability to lower gal-1-p in primary patient fibroblasts. The compounds described herein should provide a starting point for further development of drug candidates for the GALK1 modulation in the Classic Galactosemia.


Sujet(s)
Galactokinase/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , Domaine catalytique/effets des médicaments et des substances chimiques , Conception de médicament , Découverte de médicament , Galactokinase/composition chimique , Galactokinase/métabolisme , Humains , Simulation de docking moléculaire , Bibliothèques de petites molécules/composition chimique , Bibliothèques de petites molécules/pharmacologie
11.
Bioorg Med Chem ; 26(8): 1727-1739, 2018 05 01.
Article de Anglais | MEDLINE | ID: mdl-29555419

RÉSUMÉ

Proliferating cells, including cancer cells, obtain serine both exogenously and via the metabolism of glucose. By catalyzing the first, rate-limiting step in the synthesis of serine from glucose, phosphoglycerate dehydrogenase (PHGDH) controls flux through the biosynthetic pathway for this important amino acid and represents a putative target in oncology. To discover inhibitors of PHGDH, a coupled biochemical assay was developed and optimized to enable high-throughput screening for inhibitors of human PHGDH. Feedback inhibition was minimized by coupling PHGDH activity to two downstream enzymes (PSAT1 and PSPH), providing a marked improvement in enzymatic turnover. Further coupling of NADH to a diaphorase/resazurin system enabled a red-shifted detection readout, minimizing interference due to compound autofluorescence. With this protocol, over 400,000 small molecules were screened for PHGDH inhibition, and following hit validation and triage work, a piperazine-1-thiourea was identified. Following rounds of medicinal chemistry and SAR exploration, two probes (NCT-502 and NCT-503) were identified. These molecules demonstrated improved target activity and encouraging ADME properties, enabling in vitro assessment of the biological importance of PHGDH, and its role in the fate of serine in PHGDH-dependent cancer cells. This manuscript reports the assay development and medicinal chemistry leading to the development of NCT-502 and -503 reported in Pacold et al. (2016).


Sujet(s)
Antienzymes/pharmacologie , Phosphoglycerate dehydrogenase/antagonistes et inhibiteurs , Pipérazines/pharmacologie , Thiourée/analogues et dérivés , Thiourée/pharmacologie , Relation dose-effet des médicaments , Découverte de médicament , Antienzymes/synthèse chimique , Antienzymes/composition chimique , Tests de criblage à haut débit , Humains , Structure moléculaire , Phosphoglycerate dehydrogenase/génétique , Phosphoglycerate dehydrogenase/métabolisme , Pipérazines/synthèse chimique , Pipérazines/composition chimique , Relation structure-activité , Thiourée/synthèse chimique , Thiourée/composition chimique
12.
Oncotarget ; 8(53): 90959-90968, 2017 Oct 31.
Article de Anglais | MEDLINE | ID: mdl-29207616

RÉSUMÉ

Cancer metabolism has emerged as an increasingly attractive target for interfering with tumor growth. Small molecule activators of pyruvate kinase isozyme M2 (PKM2) suppress tumor formation but have an unknown effect on established tumors. We demonstrate that TEPP-46, a PKM2 activator, results in increased glucose consumption, providing the rationale for combining PKM2 activators with the toxic glucose analog, 2-deoxy-D-glucose (2-DG). Combination treatment resulted in reduced viability of a range of cell lines in standard cell culture conditions at concentrations of drugs that had no effect when used alone. This effect was replicated in vivo on established subcutaneous tumors. We further demonstrated the ability to detect acute metabolic differences in combination treatment using hyperpolarized magnetic resonance spectroscopy (MRS). Combination treated tumors displayed a higher pyruvate to lactate 13C-label exchange 2 hr post-treatment. This ability to assess the effect of drugs non-invasively may accelerate the implementation and clinical translation of drugs that target cancer metabolism.

13.
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
15.
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
16.
Proc Natl Acad Sci U S A ; 113(34): 9587-92, 2016 08 23.
Article de Anglais | MEDLINE | ID: mdl-27482083

RÉSUMÉ

The aggregation of α-synuclein (aSyn) leading to the formation of Lewy bodies is the defining pathological hallmark of Parkinson's disease (PD). Rare familial PD-associated mutations in aSyn render it aggregation-prone; however, PD patients carrying wild type (WT) aSyn also have aggregated aSyn in Lewy bodies. The mechanisms by which WT aSyn aggregates are unclear. Here, we report that inflammation can play a role in causing the aggregation of WT aSyn. We show that activation of the inflammasome with known stimuli results in the aggregation of aSyn in a neuronal cell model of PD. The insoluble aggregates are enriched with truncated aSyn as found in Lewy bodies of the PD brain. Inhibition of the inflammasome enzyme caspase-1 by chemical inhibition or genetic knockdown with shRNA abated aSyn truncation. In vitro characterization confirmed that caspase-1 directly cleaves aSyn, generating a highly aggregation-prone species. The truncation-induced aggregation of aSyn is toxic to neuronal culture, and inhibition of caspase-1 by shRNA or a specific chemical inhibitor improved the survival of a neuronal PD cell model. This study provides a molecular link for the role of inflammation in aSyn aggregation, and perhaps in the pathogenesis of sporadic PD as well.


Sujet(s)
Caspase-1/génétique , Inflammasomes/métabolisme , Corps de Lewy/métabolisme , Neurones/métabolisme , Agrégats de protéines/génétique , alpha-Synucléine/génétique , Alun/pharmacologie , Caspase-1/métabolisme , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Dipeptides/pharmacologie , Régulation de l'expression des gènes , Humains , Interleukine-1 bêta/génétique , Interleukine-1 bêta/métabolisme , Corps de Lewy/effets des médicaments et des substances chimiques , Corps de Lewy/anatomopathologie , Lipopolysaccharides/pharmacologie , Neurones/effets des médicaments et des substances chimiques , Neurones/anatomopathologie , Nigéricine/pharmacologie , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Transduction du signal , Ménadione/pharmacologie , alpha-Synucléine/composition chimique , alpha-Synucléine/métabolisme , para-Aminobenzoates/pharmacologie
17.
ACS Chem Biol ; 11(11): 3214-3225, 2016 11 18.
Article de Anglais | MEDLINE | ID: mdl-27552339

RÉSUMÉ

Loss of function mutations in Kelch-like ECH Associated Protein 1 (KEAP1), or gain-of-function mutations in nuclear factor erythroid 2-related factor 2 (NRF2), are common in non-small cell lung cancer (NSCLC) and associated with therapeutic resistance. To discover novel NRF2 inhibitors for targeted therapy, we conducted a quantitative high-throughput screen using a diverse set of ∼400 000 small molecules (Molecular Libraries Small Molecule Repository Library, MLSMR) at the National Center for Advancing Translational Sciences. We identified ML385 as a probe molecule that binds to NRF2 and inhibits its downstream target gene expression. Specifically, ML385 binds to Neh1, the Cap 'N' Collar Basic Leucine Zipper (CNC-bZIP) domain of NRF2, and interferes with the binding of the V-Maf Avian Musculoaponeurotic Fibrosarcoma Oncogene Homologue G (MAFG)-NRF2 protein complex to regulatory DNA binding sequences. In clonogenic assays, when used in combination with platinum-based drugs, doxorubicin or taxol, ML385 substantially enhances cytotoxicity in NSCLC cells, as compared to single agents. ML385 shows specificity and selectivity for NSCLC cells with KEAP1 mutation, leading to gain of NRF2 function. In preclinical models of NSCLC with gain of NRF2 function, ML385 in combination with carboplatin showed significant antitumor activity. We demonstrate the discovery and validation of ML385 as a novel and specific NRF2 inhibitor and conclude that targeting NRF2 may represent a promising strategy for the treatment of advanced NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Résistance aux médicaments antinéoplasiques , Protéine-1 de type kelch associée à ECH/génétique , Tumeurs du poumon/traitement médicamenteux , Facteur-2 apparenté à NF-E2/antagonistes et inhibiteurs , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire tumorale , Humains , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie
18.
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
19.
EBioMedicine ; 8: 49-59, 2016 Jun.
Article de Anglais | MEDLINE | ID: mdl-27428418

RÉSUMÉ

Excess lipid storage is an epidemic problem in human populations. Thus, the identification of small molecules to treat or prevent lipid storage-related metabolic complications is of great interest. Here we screened >320.000 compounds for their ability to prevent a cellular lipid accumulation phenotype. We used fly cells because the multifarious tools available for this organism should facilitate unraveling the mechanism-of-action of active small molecules. Of the several hundred lipid storage inhibitors identified in the primary screen we concentrated on three structurally diverse and potent compound classes active in cells of multiple species (including human) and negligible cytotoxicity. Together with Drosophila in vivo epistasis experiments, RNA-Seq expression profiles suggested that the target of one of the small molecules was diacylglycerol acyltransferase 1 (DGAT1), a key enzyme in the production of triacylglycerols and prominent human drug target. We confirmed this prediction by biochemical and enzymatic activity tests.


Sujet(s)
Diacylglycerol O-acyltransferase/métabolisme , Antienzymes/métabolisme , Génomique , Animaux , Cellules COS , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Chlorocebus aethiops , Diacylglycerol O-acyltransferase/antagonistes et inhibiteurs , Diacylglycerol O-acyltransferase/génétique , Drosophila/métabolisme , Antienzymes/composition chimique , Antienzymes/pharmacologie , Épistasie , Acides gras/métabolisme , Femelle , Humains , Peroxydation lipidique , Mâle , Souris , Phénotype , Pyrroles/composition chimique , Pyrroles/métabolisme , Pyrroles/pharmacologie , Analyse de séquence d'ARN , Bibliothèques de petites molécules/composition chimique , Bibliothèques de petites molécules/métabolisme , Bibliothèques de petites molécules/pharmacologie , Relation structure-activité
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...