Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Chem Biol ; 31(6): 1036-1038, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38906107

RESUMEN

In this Voices piece, the Cell Chemical Biology editors ask researchers from a range of backgrounds: what are some exciting discoveries in the induced proximity field and the next frontier for therapeutic development?


Asunto(s)
Descubrimiento de Drogas , Humanos
2.
Nat Chem Biol ; 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38907113

RESUMEN

Metabolic alterations in cancer precipitate in associated dependencies that can be therapeutically exploited. To meet this goal, natural product-inspired small molecules can provide a resource of invaluable chemotypes. Here, we identify orpinolide, a synthetic withanolide analog with pronounced antileukemic properties, via orthogonal chemical screening. Through multiomics profiling and genome-scale CRISPR-Cas9 screens, we identify that orpinolide disrupts Golgi homeostasis via a mechanism that requires active phosphatidylinositol 4-phosphate signaling at the endoplasmic reticulum-Golgi membrane interface. Thermal proteome profiling and genetic validation studies reveal the oxysterol-binding protein OSBP as the direct and phenotypically relevant target of orpinolide. Collectively, these data reaffirm sterol transport as a therapeutically actionable dependency in leukemia and motivate ensuing translational investigation via the probe-like compound orpinolide.

4.
Nat Chem Biol ; 20(1): 93-102, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37679459

RESUMEN

Molecular glue degraders are an effective therapeutic modality, but their design principles are not well understood. Recently, several unexpectedly diverse compounds were reported to deplete cyclin K by linking CDK12-cyclin K to the DDB1-CUL4-RBX1 E3 ligase. Here, to investigate how chemically dissimilar small molecules trigger cyclin K degradation, we evaluated 91 candidate degraders in structural, biophysical and cellular studies and reveal all compounds acquire glue activity via simultaneous CDK12 binding and engagement of DDB1 interfacial residues, in particular Arg928. While we identify multiple published kinase inhibitors as cryptic degraders, we also show that these glues do not require pronounced inhibitory properties for activity and that the relative degree of CDK12 inhibition versus cyclin K degradation is tuneable. We further demonstrate cyclin K degraders have transcriptional signatures distinct from CDK12 inhibitors, thereby offering unique therapeutic opportunities. The systematic structure-activity relationship analysis presented herein provides a conceptual framework for rational molecular glue design.


Asunto(s)
Ciclinas , Ubiquitina-Proteína Ligasas , Ciclinas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteolisis , Relación Estructura-Actividad
5.
Nat Chem Biol ; 20(1): 13-14, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37978259
6.
Angew Chem Int Ed Engl ; 63(12): e202316730, 2024 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-38153885

RESUMEN

Degraders hold the promise to efficiently inactivate previously intractable disease-relevant targets. Unlike traditional inhibitors, degraders act substoichiometrically and rely on the hijacked proteolysis machinery, which can also act as an entry point for resistance. To fully harness the potential of targeted protein degradation, it is crucial to comprehend resistance mechanisms and formulate effective strategies to overcome them. We conducted a chemical screening to identify synthetic lethal vulnerabilities of cancer cells that exhibit widespread resistance to degraders. Comparative profiling followed by tailored optimization delivered the small molecule RBS-10, which shows preferential cytotoxicity against cells pan-resistant to degraders. Multiomics deconvolution of the mechanism of action revealed that RBS-10 acts as a prodrug bioactivated by the oxidoreductase enzyme NQO1, which is highly overexpressed in our resistance models. Collectively, our work informs on NQO1 as an actionable vulnerability to overcome resistance to degraders and as a biomarker to selectively exploit bioactivatable prodrugs in cancer.


Asunto(s)
Neoplasias , Profármacos , Humanos , Profármacos/farmacología , Proteolisis , NAD(P)H Deshidrogenasa (Quinona)/metabolismo
7.
Nat Commun ; 14(1): 3318, 2023 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-37308482

RESUMEN

p38α is a versatile protein kinase that can control numerous processes and plays important roles in the cellular responses to stress. Dysregulation of p38α signaling has been linked to several diseases including inflammation, immune disorders and cancer, suggesting that targeting p38α could be therapeutically beneficial. Over the last two decades, numerous p38α inhibitors have been developed, which showed promising effects in pre-clinical studies but results from clinical trials have been disappointing, fueling the interest in the generation of alternative mechanisms of p38α modulation. Here, we report the in silico identification of compounds that we refer to as non-canonical p38α inhibitors (NC-p38i). By combining biochemical and structural analyses, we show that NC-p38i efficiently inhibit p38α autophosphorylation but weakly affect the activity of the canonical pathway. Our results demonstrate how the structural plasticity of p38α can be leveraged to develop therapeutic opportunities targeting a subset of the functions regulated by this pathway.


Asunto(s)
Inflamación , Transducción de Señal , Humanos , Fosforilación
8.
Chembiochem ; 24(10): e202300163, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-37018140

RESUMEN

Degraders have illustrated that compound-induced proximity to E3 ubiquitin ligases can prompt the ubiquitination and degradation of disease-relevant proteins. Hence, this pharmacology is becoming a promising alternative and complement to available therapeutic interventions (e. g., inhibitors). Degraders rely on protein binding instead of inhibition and, hence, they hold the promise to broaden the druggable proteome. Biophysical and structural biology approaches have been the cornerstone of understanding and rationalizing degrader-induced ternary complex formation. Computational models have now started to harness the experimental data from these approaches with the aim to identify and rationally help design new degraders. This review outlines the current experimental and computational strategies used to study ternary complex formation and degradation and highlights the importance of effective crosstalk between these approaches in the advancement of the targeted protein degradation (TPD) field. As our understanding of the molecular features that govern drug-induced interactions grows, faster optimizations and superior therapeutic innovations for TPD and other proximity-inducing modalities are sure to follow.


Asunto(s)
Proteoma , Ubiquitina-Proteína Ligasas , Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Unión Proteica , Proteoma/metabolismo
9.
Chem Soc Rev ; 51(13): 5498-5517, 2022 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-35723413

RESUMEN

Protein-protein interactions (PPIs) govern all biological processes. Some small molecules modulate PPIs through induced protein proximity. In particular, molecular glue degraders are monovalent compounds that orchestrate interactions between a target protein and an E3 ubiquitin ligase, prompting the proteasomal degradation of the former. This and other pharmacological strategies of targeted protein degradation (e.g. proteolysis-targeting chimeras - PROTACs) overcome some limitations of traditional occupancy-based therapeutics. Here, we provide an overview of the "molecular glue" concept, with a special focus on natural and synthetic inducers of proximity to E3s. We then briefly highlight the serendipitous discoveries of some clinical and preclinical molecular glue degraders, and discuss the first examples of intentional discoveries. Specifically, we outline the different screening strategies reported in this rapidly evolving arena and our thoughts on future perspectives. By mastering the ability to influence PPIs, molecular glue degraders can induce the degradation of unligandable proteins, thus providing an exciting path forward to broaden the targetable proteome.


Asunto(s)
Proteínas , Ubiquitina-Proteína Ligasas , Proteínas/metabolismo , Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo
10.
Cell Chem Biol ; 28(7): 1048-1060, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-33811812

RESUMEN

The therapeutic modality of targeted protein degradation promises to overcome limitations of traditional pharmacology. Small-molecule degraders recruit disease-causing proteins to E3 ubiquitin ligases, prompting their ubiquitination and degradation by the proteasome. The discovery, mechanistic elucidation, and selectivity profiling of novel degraders are often conducted in cellular systems. This highlights the need for unbiased multi-omics strategies that inform on the functionally involved components. Here, we review how proteomics and functional genomics can be integrated to identify and mechanistically understand degraders, their target selectivity as well as putative resistance mechanisms.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Humanos , Proteolisis/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/efectos de los fármacos
12.
Nat Chem Biol ; 16(11): 1199-1207, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32747809

RESUMEN

Targeted protein degradation is a new therapeutic modality based on drugs that destabilize proteins by inducing their proximity to E3 ubiquitin ligases. Of particular interest are molecular glues that can degrade otherwise unligandable proteins by orchestrating direct interactions between target and ligase. However, their discovery has so far been serendipitous, thus hampering broad translational efforts. Here, we describe a scalable strategy toward glue degrader discovery that is based on chemical screening in hyponeddylated cells coupled to a multi-omics target deconvolution campaign. This approach led us to identify compounds that induce ubiquitination and degradation of cyclin K by prompting an interaction of CDK12-cyclin K with a CRL4B ligase complex. Notably, this interaction is independent of a dedicated substrate receptor, thus functionally segregating this mechanism from all described degraders. Collectively, our data outline a versatile and broadly applicable strategy to identify degraders with nonobvious mechanisms and thus empower future drug discovery efforts.


Asunto(s)
Acetamidas/química , Antibacterianos/farmacología , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Doxiciclina/farmacología , Hidrazinas/química , Indoles/química , Proteolisis/efectos de los fármacos , Proteína 7 de Unión a Retinoblastoma/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica , Humanos , Estructura Molecular , Unión Proteica , Conformación Proteica , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/efectos de los fármacos
13.
Mol Cell ; 75(4): 849-858.e8, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31442425

RESUMEN

Inducing protein degradation via small molecules is a transformative therapeutic paradigm. Although structural requirements of target degradation are emerging, mechanisms determining the cellular response to small-molecule degraders remain poorly understood. To systematically delineate effectors required for targeted protein degradation, we applied genome-scale CRISPR/Cas9 screens for five drugs that hijack different substrate receptors (SRs) of cullin RING ligases (CRLs) to induce target proteolysis. We found that sensitivity to small-molecule degraders is dictated by shared and drug-specific modulator networks, including the COP9 signalosome and the SR exchange factor CAND1. Genetic or pharmacologic perturbation of these effectors impairs CRL plasticity and arrests a wide array of ligases in a constitutively active state. Resulting defects in CRL decommissioning prompt widespread CRL auto-degradation that confers resistance to multiple degraders. Collectively, our study informs on regulation and architecture of CRLs amenable for targeted protein degradation and outlines biomarkers and putative resistance mechanisms for upcoming clinical investigation.


Asunto(s)
Complejo del Señalosoma COP9/metabolismo , Proteínas Cullin/metabolismo , Proteolisis , Factores de Transcripción/metabolismo , Complejo del Señalosoma COP9/genética , Proteínas Cullin/genética , Humanos , Factores de Transcripción/genética
14.
Drug Discov Today Technol ; 31: 81-90, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31200863

RESUMEN

Target(ed) protein degradation (TPD) is a novel paradigm in drug discovery and a promising therapeutic strategy. TPD is based on small-molecules that catalyze the degradation of proteins by re-directing the ubiquitination activity of ubiquitin E3 ligases. Its unique molecular pharmacology enables robust, selective and fast elimination of proteins in cellular assays and in vivo. In addition to possible clinical applications, TPD is also emerging as an attractive alternative to traditional pharmacologic or genetic perturbation strategies. Directly acting degraders, as well as chemical-genetics derivatives offer unique opportunities in the pre-clinical identification, characterization and mechanistic validation of therapeutic targets.


Asunto(s)
Neoplasias/metabolismo , Proteolisis , Animales , Descubrimiento de Drogas , Humanos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Proteínas/metabolismo
15.
Genes Dev ; 32(7-8): 568-576, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29650524

RESUMEN

MEK inhibition in combination with a glycogen synthase kinase-3ß (GSK3ß) inhibitor, referred as the 2i condition, favors pluripotency in embryonic stem cells (ESCs). However, the mechanisms by which the 2i condition limits ESC differentiation and whether RAS proteins are involved in this phenomenon remain poorly understood. Here we show that RAS nullyzygosity reduces the growth of mouse ESCs (mESCs) and prohibits their differentiation. Upon RAS deficiency or MEK inhibition, ERF (E twenty-six 2 [Ets2]-repressive factor), a transcriptional repressor from the ETS domain family, translocates to the nucleus, where it binds to the enhancers of pluripotency factors and key RAS targets. Remarkably, deletion of Erf rescues the proliferative defects of RAS-devoid mESCs and restores their capacity to differentiate. Furthermore, we show that Erf loss enables the development of RAS nullyzygous teratomas. In summary, this work reveals an essential role for RAS proteins in pluripotency and identifies ERF as a key mediator of the response to RAS/MEK/ERK inhibition in mESCs.


Asunto(s)
Células Madre Embrionarias/citología , Genes ras , Proteínas Represoras/fisiología , Animales , Diferenciación Celular , Línea Celular , Células Madre Embrionarias/metabolismo , Elementos de Facilitación Genéticos , Eliminación de Gen , Ratones , Ratones Desnudos , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Teratoma/genética
16.
J Mol Biol ; 429(18): 2780-2789, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28782559

RESUMEN

The development of haploid mammalian cell lines, coupled to next-generation sequencing, has recently facilitated forward genetic screenings in mammals. For mutagenesis, retrovirus- or transposon-based gene traps are frequently used. Current methods to map gene-trap insertions are based on inverse or splinkerette PCR, which despite their efficacy are prone to artifacts and do not provide information on expression of the targeted gene. Here, we describe a new RNA sequencing-based method (TrapSeq) to map gene-trap insertions. By recognizing chimeric mRNAs containing gene-trap sequences spliced to an exon, our method identifies insertions that lead to productive trapping. When applied to individual mutant clones, our method provides a fast and cost-effective way that not only identifies the insertion site but also reveals its impact on the expression of the trapped gene. As proof of principle, we conducted two independent screenings for resistance against 6-thioguanine and an ATR inhibitor, which identified mutations known to provide resistance to these reagents and revealed ECT2 as a novel determinant for the sensitivity to ATR inhibition.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Mutagénesis Insercional/métodos , Análisis de Secuencia de ARN/métodos , Animales , Análisis Costo-Beneficio , Humanos , Factores de Tiempo
17.
Proc Natl Acad Sci U S A ; 114(35): 9367-9372, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28808015

RESUMEN

The recent development of haploid cell lines has facilitated forward genetic screenings in mammalian cells. These lines include near-haploid human cell lines isolated from a patient with chronic myelogenous leukemia (KBM7 and HAP1), as well as haploid embryonic stem cells derived from several organisms. In all cases, haploidy was shown to be an unstable state, so that cultures of mammalian haploid cells rapidly become enriched in diploids. Here we show that the observed diploidization is due to a proliferative disadvantage of haploid cells compared with diploid cells. Accordingly, single-cell-sorted haploid mammalian cells maintain the haploid state for prolonged periods, owing to the absence of competing diploids. Although the duration of interphase is similar in haploid and diploid cells, haploid cells spend longer in mitosis, indicative of problems in chromosome segregation. In agreement with this, a substantial proportion of the haploids die at or shortly after the last mitosis through activation of a p53-dependent cytotoxic response. Finally, we show that p53 deletion stabilizes haploidy in human HAP1 cells and haploid mouse embryonic stem cells. We propose that, similar to aneuploidy or tetraploidy, haploidy triggers a p53-dependent response that limits the fitness of mammalian cells.


Asunto(s)
Supervivencia Celular/fisiología , Haploidia , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular , Proliferación Celular/fisiología , Segregación Cromosómica , Humanos , Ratones , Células Madre Embrionarias de Ratones/fisiología
18.
Mol Cell ; 62(2): 307-313, 2016 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-27067599

RESUMEN

One recurring theme in drug development is to exploit synthetic lethal properties as means to preferentially damage the DNA of cancer cells. We and others have previously developed inhibitors of the ATR kinase, shown to be particularly genotoxic for cells expressing certain oncogenes. In contrast, the mechanisms of resistance to ATR inhibitors remain unexplored. We report here on a genome-wide CRISPR-Cas9 screen that identified CDC25A as a major determinant of sensitivity to ATR inhibition. CDC25A-deficient cells resist high doses of ATR inhibitors, which we show is due to their failure to prematurely enter mitosis in response to the drugs. Forcing mitotic entry with WEE1 inhibitors restores the toxicity of ATR inhibitors in CDC25A-deficient cells. With ATR inhibitors now entering the clinic, our work provides a better understanding of the mechanisms by which these compounds kill cells and reveals genetic interactions that could be used for their rational use.


Asunto(s)
Antineoplásicos/farmacología , Sistemas CRISPR-Cas , Resistencia a Antineoplásicos/genética , Células Madre Embrionarias/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Fosfatasas cdc25/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Células Madre Embrionarias/enzimología , Células Madre Embrionarias/patología , Estudio de Asociación del Genoma Completo , Humanos , Mitosis/efectos de los fármacos , Terapia Molecular Dirigida , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Transfección , Fosfatasas cdc25/genética
19.
Nat Chem Biol ; 10(9): 768-773, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25064833

RESUMEN

Genotoxic chemotherapy is the most common cancer treatment strategy. However, its untargeted generic DNA-damaging nature and associated systemic cytotoxicity greatly limit its therapeutic applications. Here, we used a haploid genetic screen in human cells to discover an absolute dependency of the clinically evaluated anticancer compound YM155 on solute carrier family member 35 F2 (SLC35F2), an uncharacterized member of the solute carrier protein family that is highly expressed in a variety of human cancers. YM155 generated DNA damage through intercalation, which was contingent on the expression of SLC35F2 and its drug-importing activity. SLC35F2 expression and YM155 sensitivity correlated across a panel of cancer cell lines, and targeted genome editing verified SLC35F2 as the main determinant of YM155-mediated DNA damage toxicity in vitro and in vivo. These findings suggest a new route to targeted DNA damage by exploiting tumor and patient-specific import of YM155.


Asunto(s)
Daño del ADN/efectos de los fármacos , Imidazoles/farmacología , Sustancias Intercalantes/farmacología , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Naftoquinonas/farmacología , Animales , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , División Celular/fisiología , Línea Celular Tumoral , Supervivencia Celular , Clonación Molecular , Ensayo Cometa , Genoma Humano/efectos de los fármacos , Genoma Humano/genética , Haploidia , Humanos , Imidazoles/metabolismo , Inmunohistoquímica , Ratones , Ratones SCID , Naftoquinonas/metabolismo , ARN Neoplásico/química , ARN Neoplásico/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...