Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Nat Chem Biol ; 18(12): 1388-1398, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36097295

RESUMEN

The Janus tyrosine kinase (JAK) family of non-receptor tyrosine kinases includes four isoforms (JAK1, JAK2, JAK3, and TYK2) and is responsible for signal transduction downstream of diverse cytokine receptors. JAK inhibitors have emerged as important therapies for immun(onc)ological disorders, but their use is limited by undesirable side effects presumed to arise from poor isoform selectivity, a common challenge for inhibitors targeting the ATP-binding pocket of kinases. Here we describe the chemical proteomic discovery of a druggable allosteric cysteine present in the non-catalytic pseudokinase domain of JAK1 (C817) and TYK2 (C838), but absent from JAK2 or JAK3. Electrophilic compounds selectively engaging this site block JAK1-dependent trans-phosphorylation and cytokine signaling, while appearing to act largely as 'silent' ligands for TYK2. Importantly, the allosteric JAK1 inhibitors do not impair JAK2-dependent cytokine signaling and are inactive in cells expressing a C817A JAK1 mutant. Our findings thus reveal an allosteric approach for inhibiting JAK1 with unprecedented isoform selectivity.


Asunto(s)
Cisteína , Proteómica , Transducción de Señal , Citocinas , Isoformas de Proteínas
3.
Bioorg Med Chem Lett ; 28(16): 2682-2687, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29731364

RESUMEN

Nicotinamide N-methyltransferase (NNMT) catalyzes the N-methylation of nicotinamide using S-adenosyl-L-methionine (SAM) as a methyl donor and, through doing so, can modulate cellular methylation potential to impact diverse epigenetic processes. NNMT has been implicated in a range of diseases, including cancer and metabolic disorders. Potent, selective, and cell-active inhibitors would constitute valuable probes to study the biological functions and therapeutic potential of NNMT. We previously reported the discovery of electrophilic small molecules that inhibit NNMT by reacting with an active-site cysteine residue in the SAM-binding pocket. Here, we have used activity-based protein profiling (ABPP)-guided medicinal chemistry to optimize the potency and selectivity of NNMT inhibitors, culminating in the discovery of multiple alpha-chloroacetamide (αCA) compounds with sub-µM IC50 values in vitro and excellent proteomic selectivity in cell lysates. However, these compounds showed much weaker inhibition of NNMT in cells, a feature that was not shared by off-targets of the αCAs. Our results show the potential for developing potent and selective covalent inhibitors of NNMT, but also highlight challenges that may be faced in targeting this enzyme in cellular systems.


Asunto(s)
Acetamidas/farmacología , Inhibidores Enzimáticos/farmacología , Nicotinamida N-Metiltransferasa/antagonistas & inhibidores , Acetamidas/síntesis química , Dominio Catalítico , Línea Celular Tumoral , Cisteína/química , Inhibidores Enzimáticos/síntesis química , Humanos , Nicotinamida N-Metiltransferasa/química
4.
Cell ; 171(3): 696-709.e23, 2017 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-28965760

RESUMEN

The transcription factor NRF2 is a master regulator of the cellular antioxidant response, and it is often genetically activated in non-small-cell lung cancers (NSCLCs) by, for instance, mutations in the negative regulator KEAP1. While direct pharmacological inhibition of NRF2 has proven challenging, its aberrant activation rewires biochemical networks in cancer cells that may create special vulnerabilities. Here, we use chemical proteomics to map druggable proteins that are selectively expressed in KEAP1-mutant NSCLC cells. Principal among these is NR0B1, an atypical orphan nuclear receptor that we show engages in a multimeric protein complex to regulate the transcriptional output of KEAP1-mutant NSCLC cells. We further identify small molecules that covalently target a conserved cysteine within the NR0B1 protein interaction domain, and we demonstrate that these compounds disrupt NR0B1 complexes and impair the anchorage-independent growth of KEAP1-mutant cancer cells. Our findings designate NR0B1 as a druggable transcriptional regulator that supports NRF2-dependent lung cancers.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/química , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/química , Neoplasias Pulmonares/genética , Proteoma/análisis , Transcriptoma , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Cisteína/metabolismo , Receptor Nuclear Huérfano DAX-1/metabolismo , Redes Reguladoras de Genes , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Ligandos , Neoplasias Pulmonares/metabolismo
5.
Nat Chem ; 9(3): 234-243, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28221344

RESUMEN

Activity-based protein profiling (ABPP) serves as a chemical proteomic platform to discover and characterize functional amino acids in proteins on the basis of their enhanced reactivity towards small-molecule probes. This approach, to date, has mainly targeted nucleophilic functional groups, such as the side chains of serine and cysteine, using electrophilic probes. Here we show that 'reverse-polarity' (RP)-ABPP using clickable, nucleophilic hydrazine probes can capture and identify protein-bound electrophiles in cells. Using this approach, we demonstrate that the pyruvoyl cofactor of S-adenosyl-L-methionine decarboxylase (AMD1) is dynamically controlled by intracellular methionine concentrations. We also identify a heretofore unknown modification-an N-terminally bound glyoxylyl group-in the poorly characterized protein secernin-3. RP-ABPP thus provides a versatile method to monitor the metabolic regulation of electrophilic cofactors and discover novel types of electrophilic modifications on proteins in human cells.


Asunto(s)
Adenosilmetionina Descarboxilasa/química , Proteínas del Tejido Nervioso/química , Proteómica , Adenosilmetionina Descarboxilasa/metabolismo , Aminoácidos/química , Aminoácidos/metabolismo , Supervivencia Celular , Colorantes Fluorescentes/química , Células HEK293 , Humanos , Hidrazinas/química , Metionina/química , Metionina/metabolismo , Estructura Molecular , Proteínas del Tejido Nervioso/metabolismo
6.
J Am Chem Soc ; 138(40): 13335-13343, 2016 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-27689866

RESUMEN

Methylation is a fundamental mechanism used in Nature to modify the structure and function of biomolecules, including proteins, DNA, RNA, and metabolites. Methyl groups are predominantly installed into biomolecules by a large and diverse class of S-adenosyl methionine (SAM)-dependent methyltransferases (MTs), of which there are ∼200 known or putative members in the human proteome. Deregulated MT activity contributes to numerous diseases, including cancer, and several MT inhibitors are in clinical development. Nonetheless, a large fraction of the human MT family remains poorly characterized, underscoring the need for new technologies to characterize MTs and their inhibitors in native biological systems. Here, we describe a suite of S-adenosyl homocysteine (SAH) photoreactive probes and their application in chemical proteomic experiments to profile and enrich a large number of MTs (>50) from human cancer cell lysates with remarkable specificity over other classes of proteins. We further demonstrate that the SAH probes can enrich MT-associated proteins and be used to screen for and assess the selectivity of MT inhibitors, leading to the discovery of a covalent inhibitor of nicotinamide N-methyltransferase (NNMT), an enzyme implicated in cancer and metabolic disorders. The chemical proteomics probes and methods for their utilization reported herein should prove of value for the functional characterization of MTs, MT complexes, and MT inhibitors in mammalian biology and disease.


Asunto(s)
Metiltransferasas/metabolismo , Proteómica , Línea Celular Tumoral , Activación Enzimática , Humanos , Sondas Moleculares/metabolismo , S-Adenosilhomocisteína/metabolismo , Rayos Ultravioleta
7.
Nature ; 534(7608): 570-4, 2016 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-27309814

RESUMEN

Small molecules are powerful tools for investigating protein function and can serve as leads for new therapeutics. Most human proteins, however, lack small-molecule ligands, and entire protein classes are considered 'undruggable'. Fragment-based ligand discovery can identify small-molecule probes for proteins that have proven difficult to target using high-throughput screening of complex compound libraries. Although reversibly binding ligands are commonly pursued, covalent fragments provide an alternative route to small-molecule probes, including those that can access regions of proteins that are difficult to target through binding affinity alone. Here we report a quantitative analysis of cysteine-reactive small-molecule fragments screened against thousands of proteins in human proteomes and cells. Covalent ligands were identified for >700 cysteines found in both druggable proteins and proteins deficient in chemical probes, including transcription factors, adaptor/scaffolding proteins, and uncharacterized proteins. Among the atypical ligand-protein interactions discovered were compounds that react preferentially with pro- (inactive) caspases. We used these ligands to distinguish extrinsic apoptosis pathways in human cell lines versus primary human T cells, showing that the former is largely mediated by caspase-8 while the latter depends on both caspase-8 and -10. Fragment-based covalent ligand discovery provides a greatly expanded portrait of the ligandable proteome and furnishes compounds that can illuminate protein functions in native biological systems.


Asunto(s)
Cisteína/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Proteoma/química , Proteoma/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Linfocitos T/metabolismo , Apoptosis , Caspasa 10/química , Caspasa 10/metabolismo , Caspasa 8/química , Caspasa 8/metabolismo , Células Cultivadas , Precursores Enzimáticos/química , Precursores Enzimáticos/metabolismo , Humanos , Ligandos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Linfocitos T/química , Factores de Transcripción/química , Factores de Transcripción/metabolismo
8.
J Am Chem Soc ; 135(17): 6442-5, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23586842

RESUMEN

A concise and highly enantioselective total synthesis of the akuammiline alkaloid (-)-vincorine has been accomplished. A key element of the synthesis is a stereoselective organocatalytic Diels-Alder, iminium cyclization cascade sequence, which serves to construct the tetracyclic alkaloid core architecture in one step from simple achiral precursors. The challenging seven-membered azepanyl ring system is installed by way of a single electron-mediated cyclization event initiated from an acyl telluride precursor. The total synthesis of (-)-vincorine is achieved in nine steps and 9% overall yield from commercially available starting materials.


Asunto(s)
Alcaloides de la Vinca/síntesis química , Ciclización , Indicadores y Reactivos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Estereoisomerismo , Alcaloides de la Vinca/química , Difracción de Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...