Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Cell ; 183(6): 1714-1731.e10, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-33275901

RESUMEN

Targeted protein degradation (TPD) refers to the use of small molecules to induce ubiquitin-dependent degradation of proteins. TPD is of interest in drug development, as it can address previously inaccessible targets. However, degrader discovery and optimization remains an inefficient process due to a lack of understanding of the relative importance of the key molecular events required to induce target degradation. Here, we use chemo-proteomics to annotate the degradable kinome. Our expansive dataset provides chemical leads for ∼200 kinases and demonstrates that the current practice of starting from the highest potency binder is an ineffective method for discovering active compounds. We develop multitargeted degraders to answer fundamental questions about the ubiquitin proteasome system, uncovering that kinase degradation is p97 dependent. This work will not only fuel kinase degrader discovery, but also provides a blueprint for evaluating targeted degradation across entire gene families to accelerate understanding of TPD beyond the kinome.


Asunto(s)
Proteínas Quinasas/metabolismo , Proteolisis , Proteoma/metabolismo , Adulto , Línea Celular , Bases de Datos de Proteínas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Quinasas/genética , Proteómica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Adulto Joven
2.
Nat Chem Biol ; 17(6): 711-717, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34035522

RESUMEN

The zinc-finger transcription factor Helios is critical for maintaining the identity, anergic phenotype and suppressive activity of regulatory T (Treg) cells. While it is an attractive target to enhance the efficacy of currently approved immunotherapies, no existing approaches can directly modulate Helios activity or abundance. Here, we report the structure-guided development of small molecules that recruit the E3 ubiquitin ligase substrate receptor cereblon to Helios, thereby promoting its degradation. Pharmacological Helios degradation destabilized the anergic phenotype and reduced the suppressive activity of Treg cells, establishing a route towards Helios-targeting therapeutics. More generally, this study provides a framework for the development of small-molecule degraders for previously unligandable targets by reprogramming E3 ligase substrate specificity.


Asunto(s)
Proteínas de Unión al ADN/efectos de los fármacos , Factor de Transcripción Ikaros/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Factores de Transcripción/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Humanos , Factor de Transcripción Ikaros/genética , Células Jurkat , Ratones , Modelos Moleculares , Estructura Molecular , Mutación/genética , Bibliotecas de Moléculas Pequeñas , Especificidad por Sustrato , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas/metabolismo
3.
Nat Chem Biol ; 16(1): 7-14, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31686031

RESUMEN

The investigational drugs E7820, indisulam and tasisulam (aryl-sulfonamides) promote the degradation of the splicing factor RBM39 in a proteasome-dependent mechanism. While the activity critically depends on the cullin RING ligase substrate receptor DCAF15, the molecular details remain elusive. Here we present the cryo-EM structure of the DDB1-DCAF15-DDA1 core ligase complex bound to RBM39 and E7820 at a resolution of 4.4 Å, together with crystal structures of engineered subcomplexes. We show that DCAF15 adopts a new fold stabilized by DDA1, and that extensive protein-protein contacts between the ligase and substrate mitigate low affinity interactions between aryl-sulfonamides and DCAF15. Our data demonstrate how aryl-sulfonamides neo-functionalize a shallow, non-conserved pocket on DCAF15 to selectively bind and degrade RBM39 and the closely related splicing factor RBM23 without the requirement for a high-affinity ligand, which has broad implications for the de novo discovery of molecular glue degraders.


Asunto(s)
Indoles/química , Péptidos y Proteínas de Señalización Intracelular/química , Proteolisis/efectos de los fármacos , Proteínas con Motivos de Reconocimiento de ARN/química , Sulfonamidas/química , Secuencias de Aminoácidos , Animales , Benzamidas/química , Benzamidas/farmacología , Microscopía por Crioelectrón , Transferencia Resonante de Energía de Fluorescencia , Humanos , Indoles/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Cinética , Unión Proteica , Dominios Proteicos , Mapeo de Interacción de Proteínas , Estructura Secundaria de Proteína , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Proteínas de Unión al ARN , Spodoptera , Sulfonamidas/farmacología , Ubiquitina-Proteína Ligasas/química , Xenopus
4.
Cell Chem Biol ; 29(10): 1470-1481.e31, 2022 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-36070758

RESUMEN

Targeted protein degradation (TPD) uses small molecules to recruit E3 ubiquitin ligases into the proximity of proteins of interest, inducing ubiquitination-dependent degradation. A major bottleneck in the TPD field is the lack of accessible E3 ligase ligands for developing degraders. To expand the E3 ligase toolbox, we sought to convert the Kelch-like ECH-associated protein 1 (KEAP1) inhibitor KI696 into a recruitment handle for several targets. While we were able to generate KEAP1-recruiting degraders of BET family and murine focal adhesion kinase (FAK), we discovered that the target scope of KEAP1 was narrow, as targets easily degraded using a cereblon (CRBN)-recruiting degrader were refractory to KEAP1-mediated degradation. Linking the KEAP1-binding ligand to a CRBN-binding ligand resulted in a molecule that induced degradation of KEAP1 but not CRBN. In sum, we characterize tool compounds to explore KEAP1-mediated ubiquitination and delineate the challenges of exploiting new E3 ligases for generating bivalent degraders.


Asunto(s)
Factor 2 Relacionado con NF-E2 , Ubiquitina-Proteína Ligasas , Ratones , Animales , Ubiquitina-Proteína Ligasas/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Ligandos , Factor 2 Relacionado con NF-E2/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Ubiquitinas/metabolismo
5.
Nat Cancer ; 3(5): 595-613, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35534777

RESUMEN

Acute myeloid leukemia (AML) remains difficult to treat and requires new therapeutic approaches. Potent inhibitors of the chromatin-associated protein MENIN have recently entered human clinical trials, opening new therapeutic opportunities for some genetic subtypes of this disease. Using genome-scale functional genetic screens, we identified IKAROS (encoded by IKZF1) as an essential transcription factor in KMT2A (MLL1)-rearranged (MLL-r) AML that maintains leukemogenic gene expression while also repressing pathways for tumor suppression, immune regulation and cellular differentiation. Furthermore, IKAROS displays an unexpected functional cooperativity and extensive chromatin co-occupancy with mixed lineage leukemia (MLL)1-MENIN and the regulator MEIS1 and an extensive hematopoietic transcriptional complex involving homeobox (HOX)A10, MEIS1 and IKAROS. This dependency could be therapeutically exploited by inducing IKAROS protein degradation with immunomodulatory imide drugs (IMiDs). Finally, we demonstrate that combined IKAROS degradation and MENIN inhibition effectively disrupts leukemogenic transcriptional networks, resulting in synergistic killing of leukemia cells and providing a paradigm for improved drug targeting of transcription and an opportunity for rapid clinical translation.


Asunto(s)
Leucemia Mieloide Aguda , Cromatina , Expresión Génica , Humanos , Factor de Transcripción Ikaros/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Factores de Transcripción/genética
6.
STAR Protoc ; 2(2): 100530, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-34041503

RESUMEN

Transcriptional changes happen within minutes; however, RNAi or genetic deletion requires days to weeks before transcription networks can be analyzed. This limitation has made it challenging to distinguish direct from indirect targets of sequence-specific transcription factors. This inability to define direct transcriptional targets hinders detailed studies of transcriptional mechanisms. This protocol combines rapid degradation of endogenous transcription factors with nascent transcript analysis to define the earliest, and likely direct, regulatory targets of transcription factors. For complete details on the use and execution of this protocol, please refer to Stengel et al., 2021).


Asunto(s)
Factores de Transcripción , Transcripción Genética , Animales , Células Cultivadas , Interferencia de ARN , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , Factores de Transcripción/análisis , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética/genética , Transcripción Genética/fisiología
7.
J Med Chem ; 64(15): 11637-11650, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34279939

RESUMEN

Chemical biology tools to modulate protein levels in cells are critical to decipher complex biology. Targeted protein degradation offers the potential for rapid and dose-dependent protein depletion through the use of protein fusion tags toward which protein degraders have been established. Here, we present a newly developed protein degradation tag BRD4BD1L94V along with the corresponding cereblon (CRBN)-based heterobifunctional degrader based on a "bump-and-hole" approach. The resulting compound XY-06-007 shows a half-degradation concentration (DC50, 6 h) of 10 nM against BRD4BD1L94V with no degradation of off-targets, as assessed by whole proteome mass spectrometry, and demonstrates suitable pharmacokinetics for in vivo studies. We demonstrate that BRD4BD1L94V can be combined with the dTAG approach to achieve simultaneous degrader-mediated depletion of their respective protein fusions. This orthogonal system complements currently available protein degradation tags and enables investigation into the consequences resulting from rapid degradation of previously undruggable disease codependencies.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Diseño de Fármacos , Factores de Transcripción/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Relación Estructura-Actividad , Factores de Transcripción/metabolismo
8.
Cell Chem Biol ; 28(10): 1514-1527.e4, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-34314730

RESUMEN

Targeted protein degradation refers to the use of small molecules that recruit a ubiquitin ligase to a target protein for ubiquitination and subsequent proteasome-dependent degradation. While degraders have been developed for many targets, key questions regarding degrader development and the consequences of acute pharmacological degradation remain, specifically for targets that exist in obligate multi-protein complexes. Here, we synthesize a pan-histone deacetylase (HDAC) degrader library for the chemo-proteomic exploration of acute degradation of a key class of chromatin-modifying enzymes. Using chemo-proteomics, we not only map the degradability of the zinc-dependent HDAC family identifying leads for targeting HDACs 1-8 and 10 but also explore important aspects of degrading epigenetic enzymes. We discover cell line-driven target specificity and that HDAC degradation often results in collateral loss of HDAC-containing repressive complexes. These findings potentially offer a new mechanism toward controlling chromatin structure, and our resource will facilitate accelerated degrader design and development for HDACs.


Asunto(s)
Histona Desacetilasas/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo , Línea Celular , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/metabolismo , Histona Desacetilasas/química , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Proteolisis , Proteómica/métodos , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
9.
Cell Chem Biol ; 27(1): 66-73.e7, 2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31859249

RESUMEN

The PI3K/AKT signaling cascade is one of the most commonly dysregulated pathways in cancer, with over half of tumors exhibiting aberrant AKT activation. Although potent small-molecule AKT inhibitors have entered clinical trials, robust and durable therapeutic responses have not been observed. As an alternative strategy to target AKT, we report the development of INY-03-041, a pan-AKT degrader consisting of the ATP-competitive AKT inhibitor GDC-0068 conjugated to lenalidomide, a recruiter of the E3 ubiquitin ligase substrate adaptor Cereblon (CRBN). INY-03-041 induced potent degradation of all three AKT isoforms and displayed enhanced anti-proliferative effects relative to GDC-0068. Notably, INY-03-041 promoted sustained AKT degradation and inhibition of downstream signaling effects for up to 96 h, even after compound washout. Our findings suggest that AKT degradation may confer prolonged pharmacological effects compared with inhibition, and highlight the potential advantages of AKT-targeted degradation.


Asunto(s)
Descubrimiento de Drogas , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteolisis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Humanos , Conformación Molecular , Piperazinas/química , Inhibidores de Proteínas Quinasas/química , Pirimidinas/química
10.
Cell Chem Biol ; 27(1): 57-65.e9, 2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31735695

RESUMEN

The G1/S cell cycle checkpoint is frequently dysregulated in cancer, leaving cancer cells reliant on a functional G2/M checkpoint to prevent excessive DNA damage. Wee1 regulates the G2/M checkpoint by phosphorylating CDK1 at Tyr15 to prevent mitotic entry. Previous drug development efforts targeting Wee1 resulted in the clinical-grade inhibitor, AZD1775. However, AZD1775 is burdened by dose-limiting adverse events, and has off-target PLK1 activity. In an attempt to overcome these limitations, we developed Wee1 degraders by conjugating AZD1775 to the cereblon (CRBN)-binding ligand, pomalidomide. The resulting lead compound, ZNL-02-096, degrades Wee1 while sparing PLK1, induces G2/M accumulation at 10-fold lower doses than AZD1775, and synergizes with Olaparib in ovarian cancer cells. We demonstrate that ZNL-02-096 has CRBN-dependent pharmacology that is distinct from AZD1775, which justifies further evaluation of selective Wee1 degraders.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Desarrollo de Medicamentos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/metabolismo , Proteolisis/efectos de los fármacos , Pirazoles/farmacología , Pirimidinonas/farmacología , Talidomida/análogos & derivados , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN , Femenino , Humanos , Estructura Molecular , Ftalazinas/química , Ftalazinas/farmacología , Piperazinas/química , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/química , Pirazoles/química , Pirimidinonas/química , Talidomida/química , Talidomida/farmacología
11.
Nat Commun ; 10(1): 3468, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31371704

RESUMEN

Targeted protein degradation is a promising drug development paradigm. Here we leverage this strategy to develop a new class of small molecule antivirals that induce proteasomal degradation of viral proteins. Telaprevir, a reversible-covalent inhibitor that binds to the hepatitis C virus (HCV) protease active site is conjugated to ligands that recruit the CRL4CRBN ligase complex, yielding compounds that can both inhibit and induce the degradation of the HCV NS3/4A protease. An optimized degrader, DGY-08-097, potently inhibits HCV in a cellular infection model, and we demonstrate that protein degradation contributes to its antiviral activity. Finally, we show that this new class of antiviral agents can overcome viral variants that confer resistance to traditional enzymatic inhibitors such as telaprevir. Overall, our work provides proof-of-concept that targeted protein degradation may provide a new paradigm for the development of antivirals with superior resistance profiles.


Asunto(s)
Antivirales/farmacología , Farmacorresistencia Viral/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Inhibidores de Proteasas/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antivirales/química , Línea Celular Tumoral , Diseño de Fármacos , Farmacorresistencia Viral/genética , Técnicas de Silenciamiento del Gen , Células HEK293 , Hepacivirus/efectos de los fármacos , Hepacivirus/metabolismo , Hepatitis C/tratamiento farmacológico , Hepatitis C/genética , Hepatitis C/virología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ligandos , Modelos Moleculares , Oligopéptidos/química , Oligopéptidos/farmacología , Prueba de Estudio Conceptual , Inhibidores de Proteasas/química , Proteolisis/efectos de los fármacos , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas no Estructurales Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA