Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Nat Commun ; 14(1): 686, 2023 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-36754960

RESUMEN

Deubiquitinating enzymes (DUBs) are an emerging drug target class of ~100 proteases that cleave ubiquitin from protein substrates to regulate many cellular processes. A lack of selective chemical probes impedes pharmacologic interrogation of this important gene family. DUBs engage their cognate ligands through a myriad of interactions. We embrace this structural complexity to tailor a chemical diversification strategy for a DUB-focused covalent library. Pairing our library with activity-based protein profiling as a high-density primary screen, we identify selective hits against 23 endogenous DUBs spanning four subfamilies. Optimization of an azetidine hit yields a probe for the understudied DUB VCPIP1 with nanomolar potency and in-family selectivity. Our success in identifying good chemical starting points as well as structure-activity relationships across the gene family from a modest but purpose-build library challenges current paradigms that emphasize ultrahigh throughput in vitro or virtual screens against an ever-increasing scope of chemical space.


Asunto(s)
Endopeptidasas , Ubiquitina , Ubiquitina/metabolismo , Endopeptidasas/metabolismo , Péptido Hidrolasas/metabolismo , Relación Estructura-Actividad , Enzimas Desubicuitinizantes/metabolismo , Ubiquitinación
2.
STAR Protoc ; 2(4): 100896, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34723213

RESUMEN

Identification of selective deubiquitinase (DUB) inhibitors is critical for probe development to further understand and explore DUB biological function. Here, we detail the optimization and deployment of an in vitro fluorogenic ubiquitin-rhodamine assay to conduct high-throughput screening of a small molecule library against a panel of DUBs. In screening the compound library against multiple DUBs in parallel, we describe an approach for identifying selective DUB inhibitors and provide a roadmap for enabling selective DUB inhibitor discovery. For complete details on the use and execution of this protocol, please refer to Varca et al. (2021).


Asunto(s)
Enzimas Desubicuitinizantes , Inhibidores Enzimáticos , Ensayos Analíticos de Alto Rendimiento/métodos , Rodaminas/metabolismo , Ubiquitina/metabolismo , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Enzimas Desubicuitinizantes/metabolismo , Pruebas de Enzimas , Escherichia coli/genética , Colorantes Fluorescentes/química , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Bibliotecas de Moléculas Pequeñas
3.
Cell Chem Biol ; 28(12): 1758-1771.e13, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34129829

RESUMEN

Deubiquitinating enzymes (DUBs) are a class of isopeptidases that regulate ubiquitin dynamics through catalytic cleavage of ubiquitin from protein substrates and ubiquitin precursors. Despite growing interest in DUB biological function and potential as therapeutic targets, few selective small-molecule inhibitors and no approved drugs currently exist. To identify chemical scaffolds targeting specific DUBs and establish a broader framework for future inhibitor development across the gene family, we performed high-throughput screening of a chemically diverse small-molecule library against eight different DUBs, spanning three well-characterized DUB families. Promising hit compounds were validated in a series of counter-screens and orthogonal assays, as well as further assessed for selectivity across expanded panels of DUBs. Through these efforts, we have identified multiple highly selective DUB inhibitors and developed a roadmap for rapidly identifying and validating selective inhibitors of related enzymes.


Asunto(s)
Enzimas Desubicuitinizantes/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Enzimas Desubicuitinizantes/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Femenino , Células HEK293 , Humanos , Estructura Molecular , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química
4.
Cell Chem Biol ; 28(1): 78-87.e3, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33007217

RESUMEN

Deubiquitinating enzymes (DUBs) catalyze the removal of ubiquitin, thereby reversing the activity of E3 ubiquitin ligases and are central to the control of protein abundance and function. Despite the growing interest in DUBs as therapeutic targets, cellular functions for DUBs remain largely unknown and technical challenges often preclude the identification of DUB substrates in a comprehensive manner. Here, we demonstrate that treatment with potent DUB inhibitors coupled to mass spectrometry-based proteomics can identify DUB substrates at a proteome-wide scale. We applied this approach to USP7, a DUB widely investigated as a therapeutic target and identified many known substrates and additional targets. We demonstrate that USP7 substrates are enriched for DNA repair enzymes and E3 ubiquitin ligases. This work provides not only a comprehensive annotation of USP7 substrates, but a general protocol widely applicable to other DUBs, which is critical for translational development of DUB targeted agents.


Asunto(s)
Proteómica , Peptidasa Específica de Ubiquitina 7/análisis , Biocatálisis , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Humanos , Especificidad por Sustrato , Peptidasa Específica de Ubiquitina 7/antagonistas & inhibidores , Peptidasa Específica de Ubiquitina 7/metabolismo , Ubiquitinación
5.
J Exp Med ; 215(1): 197-216, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29158376

RESUMEN

On-target drug delivery remains a challenge in cancer precision medicine; it is difficult to deliver a targeted therapy to cancer cells without incurring toxicity to normal tissues. The SERCA (sarco-endoplasmic reticulum Ca2+ ATPase) inhibitor thapsigargin inhibits mutant NOTCH1 receptors compared with wild type in T cell acute lymphoblastic leukemia (T-ALL), but its administration is predicted to be toxic in humans. Leveraging the addiction of ALL to folic acid, we conjugated folate to an alcohol derivative of thapsigargin via a cleavable ester linkage. JQ-FT is recognized by folate receptors on the plasma membrane and delivered into leukemia cells as a potent antileukemic agent. In mechanistic and translational models of T-ALL, we demonstrate NOTCH1 inhibition in vitro and in vivo. These proof-of-concept studies support the further optimization of this first-in-class NOTCH1 inhibitor with dual selectivity: leukemia over normal cells and NOTCH1 mutants over wild-type receptors. Furthermore, tumor-specific disruption of Notch signaling may overcome legitimate concerns associated with the tumor suppressor function of nontargeted Notch pathway inhibitors.


Asunto(s)
Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos , Leucemia/genética , Mutación , Receptor Notch1/antagonistas & inhibidores , Receptor Notch1/genética , Animales , Transporte Biológico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Endocitosis , Receptor 2 de Folato/genética , Receptor 2 de Folato/metabolismo , Ácido Fólico/química , Expresión Génica , Humanos , Leucemia/tratamiento farmacológico , Leucemia/metabolismo , Leucemia/patología , Ratones , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Unión Proteica , Receptor Notch1/metabolismo , Transducción de Señal/efectos de los fármacos , Tapsigargina/química , Ensayos Antitumor por Modelo de Xenoinjerto
6.
ACS Chem Biol ; 10(3): 667-74, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25397901

RESUMEN

The DOT1L lysine methyltransferase has emerged as a validated therapeutic target in MLL-rearranged (MLLr) acute leukemias. Although S-adenosylmethionine competitive inhibitors have demonstrated pharmacological proof-of-principle in MLLr-leukemia, these compounds require further optimization to improve cellular potency and pharmacokinetic stability. Limiting DOT1L inhibitor discovery and ligand optimization have been complex biochemical methods often using radionucleotides and cellular methods requiring prolonged culture. We therefore developed a new suite of assay technologies that allows comparative assessment of chemical tools for DOT1L in a miniaturized format. Coupling these assays with structural information, we developed new insights into DOT1L ligand binding and identified several functionalized probes with increased cellular potency (IC50 values ∼10 nM) and excellent selectivity for DOT1L. Together these assay technologies define a platform capability for discovery and optimization of small-molecule DOT1L inhibitors.


Asunto(s)
Antineoplásicos/síntesis química , Inhibidores Enzimáticos/síntesis química , Regulación Neoplásica de la Expresión Génica , Ensayos Analíticos de Alto Rendimiento , Histonas/antagonistas & inhibidores , Metiltransferasas/antagonistas & inhibidores , Adenosina/análogos & derivados , Adenosina/síntesis química , Adenosina/farmacología , Antineoplásicos/farmacología , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Células Epiteliales/patología , N-Metiltransferasa de Histona-Lisina , Histonas/genética , Histonas/metabolismo , Humanos , Ligandos , Metilación , Metiltransferasas/química , Metiltransferasas/genética , Metiltransferasas/metabolismo , Modelos Moleculares , Compuestos de Fenilurea/síntesis química , Compuestos de Fenilurea/farmacología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA